151
|
Mamun A, Yokoyama U, Saito J, Ito S, Hiromi T, Umemura M, Fujita T, Yasuda S, Minami T, Goda M, Uchida K, Suzuki S, Masuda M, Ishikawa Y. A selective antagonist of prostaglandin E receptor subtype 4 attenuates abdominal aortic aneurysm. Physiol Rep 2018; 6:e13878. [PMID: 30230255 PMCID: PMC6144453 DOI: 10.14814/phy2.13878] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/30/2018] [Accepted: 09/02/2018] [Indexed: 12/15/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a progressive disease that has an increasing prevalence with aging, but no effective pharmacological therapy to attenuate AAA progression is currently available. We reported that the prostaglandin E receptor EP4 plays roles in AAA progression. Here, we show the effect of CJ-42794, a selective EP4 antagonist, on AAA using two mouse models (angiotensin II- and CaCl2 -induced AAAs) and human aortic smooth muscle cells isolated from AAA tissue. Oral administration of CJ-42794 (0.2 mg/kg per day) for 4 weeks significantly decreased AAA formation in ApoE-/- mice infused with angiotensin II (1 μg/kg per min), in which elastic fiber degradation and activations of matrix metalloproteinase (MMP)-2 and MMP-9 were attenuated. Interleukin-6 (IL-6) proteins were highly expressed in the medial layer of angiotensin II-induced mouse AAA tissues, whereas this expression was significantly decreased in mice treated with CJ-42794. AAA formation induced by periaortic CaCl2 application in wild-type mice was also reduced by oral administration of CJ-42794 for 4 weeks. After oral administration of CJ-42794 beginning 2 weeks after periaortic CaCl2 application and continuing for an additional 4 weeks, the aortic diameter and elastic fiber degradation grade were significantly smaller in CJ-42794-treated mice than in untreated mice. Additionally, in smooth muscle cells isolated from human AAA tissues, stimulation of CJ-42794 inhibited PGE2 -induced IL-6 secretion in a dose-dependent manner and decreased PGE2 -induced MMP-2 activity. These data suggest that inhibition of EP4 has the potential to be a pharmacological strategy for attenuation of AAA progression.
Collapse
MESH Headings
- Animals
- Aorta, Abdominal/drug effects
- Aorta, Abdominal/metabolism
- Aorta, Abdominal/pathology
- Aortic Aneurysm, Abdominal/drug therapy
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Apolipoproteins E/deficiency
- Cells, Cultured
- Humans
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors
- Receptors, Prostaglandin E, EP4 Subtype/metabolism
- Sulfonylurea Compounds/pharmacology
- Sulfonylurea Compounds/therapeutic use
Collapse
Affiliation(s)
- Al Mamun
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Utako Yokoyama
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Junichi Saito
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Satoko Ito
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Taro Hiromi
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
- Department of Emergency medicineGraduate School of MedicineYokohama City UniversityYokohamaJapan
| | - Masanari Umemura
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Takayuki Fujita
- Cardiovascular Research InstituteYokohama City UniversityYokohamaJapan
| | - Shota Yasuda
- Department of SurgeryYokohama City UniversityYokohamaJapan
| | - Tomoyuki Minami
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | - Motohiko Goda
- Department of SurgeryYokohama City UniversityYokohamaJapan
| | - Keiji Uchida
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | | | - Munetaka Masuda
- Department of SurgeryYokohama City UniversityYokohamaJapan
- Cardiovascular CenterYokohama City University Medical CenterYokohamaJapan
| | | |
Collapse
|
152
|
Li H, Luo Y, Xu Y, Yang L, Hu C, Chen Q, Yang Y, Ma J, Zhang J, Xia H, Li Y, Yang J. Meloxicam Improves Cognitive Impairment of Diabetic Rats through COX2-PGE2-EPs-cAMP/pPKA Pathway. Mol Pharm 2018; 15:4121-4131. [PMID: 30109938 DOI: 10.1021/acs.molpharmaceut.8b00532] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Diabetics often face greater risk of cognitive impairment than nondiabetics. However, how to prevent this disease is still unconfirmed. In this study, we investigated the potential protection and mechanism of meloxicam on cognitive impairment in diabetic rats. The diabetic rat model was established with a high-fat diet and a small dose of streptozotocin (40 mg/kg). The changes of spatial learning and memory, histopathology, and the protein expressions of amyloid protein precursor (APP) and β-amyloid (Aβ) indicated that diabetic rats had neuronal injury and cognitive impairment. Tumor necrosis factor α (TNFα), interleukin 6 (IL-6), C reactive protein (CRP) and prostaglandin E2 (PGE2) levels, and microglial cell number were significantly increased in the diabetic rat brain. Meanwhile, the protein expressions of APP, Aβ, cyclooxygenases2 (COX2), E-type prostanoid recptors 1 (EP1) and EP2, and the level of cyclic adenosine monophosphate (cAMP) were significantly increased, while the protein expressions of EP3 and phosphorylated protein kinase A (pPKA) were significantly decreased in the diabetic rat hippocampus and cortex. However, the EP4 protein expression had no significant changes. Meloxicam significantly improved neuronal injury and cognitive impairment, and significantly decreased inflammatory cytokines levels. Meloxicam also significantly decreased the protein expressions of APP, Aβ, COX2, EP1 and EP2, and the level of cAMP and significantly increased the EP3 and pPKA protein expressions in rat hippocampus and cortex. However, meloxicam did not significantly influence the levels of blood glucose, lipids, and insulin of rats. Our results suggest that meloxicam could significantly protect diabetic rats from cognitive impairment via a mechanism that may be associated with rebalancing the COX2-PGE2-EPs-cAMP/PKA pathway.
Collapse
Affiliation(s)
- Huan Li
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Ying Luo
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Ying Xu
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences , State University of New York at Buffalo , Buffalo , New York 14214 , United States
| | - Lu Yang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Congli Hu
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Qi Chen
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Yang Yang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Jie Ma
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Jiahua Zhang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Hui Xia
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Yuke Li
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| | - Junqing Yang
- The Key Laboratory of Biochemistry and Molecular Pharmacology, Department of Pharmacology , Chongqing Medical University , Chongqing 400016 , China
| |
Collapse
|
153
|
|
154
|
Sáez JC, Contreras-Duarte S, Gómez GI, Labra VC, Santibañez CA, Gajardo-Gómez R, Avendaño BC, Díaz EF, Montero TD, Velarde V, Orellana JA. Connexin 43 Hemichannel Activity Promoted by Pro-Inflammatory Cytokines and High Glucose Alters Endothelial Cell Function. Front Immunol 2018; 9:1899. [PMID: 30158937 PMCID: PMC6104120 DOI: 10.3389/fimmu.2018.01899] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
The present work was done to elucidate whether hemichannels of a cell line derived from endothelial cells are affected by pro-inflammatory conditions (high glucose and IL-1β/TNF-α) known to lead to vascular dysfunction. We used EAhy 926 cells treated with high glucose and IL-1β/TNF-α. The hemichannel activity was evaluated with the dye uptake method and was abrogated with selective inhibitors or knocking down of hemichannel protein subunits with siRNA. Western blot analysis, cell surface biotinylation, and confocal microscopy were used to evaluate total and plasma membrane amounts of specific proteins and their cellular distribution, respectively. Changes in intracellular Ca2+ and nitric oxide (NO) signals were estimated by measuring FURA-2 and DAF-FM probes, respectively. High glucose concentration was found to elevate dye uptake, a response that was enhanced by IL-1β/TNF-α. High glucose plus IL-1β/TNF-α-induced dye uptake was abrogated by connexin 43 (Cx43) but not pannexin1 knockdown. Furthermore, Cx43 hemichannel activity was associated with enhanced ATP release and activation of p38 MAPK, inducible NO synthase, COX2, PGE2 receptor EP1, and P2X7/P2Y1 receptors. Inhibition of the above pathways prevented completely the increase in Cx43 hemichannel activity of cells treated high glucose and IL-1β/TNF-α. Both synthetic and endogenous cannabinoids (CBs) also prevented the increment in Cx43 hemichannel opening, as well as the subsequent generation and release of ATP and NO induced by pro-inflammatory conditions. The counteracting action of CBs also was extended to other endothelial alterations evoked by IL-1β/TNF-α and high glucose, including increased ATP-dependent Ca2+ dynamics and insulin-induced NO production. Finally, inhibition of Cx43 hemichannels also prevented the ATP release from endothelial cells treated with IL-1β/TNF-α and high glucose. Therefore, we propose that reduction of hemichannel activity could represent a strategy against the activation of deleterious pathways that lead to endothelial dysfunction and possibly cell damage evoked by high glucose and pro-inflammatory conditions during cardiovascular diseases.
Collapse
Affiliation(s)
- Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Susana Contreras-Duarte
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile.,Departamento de Ginecología y Obstetricia, Escuela de Medicina, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo I Gómez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valeria C Labra
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Cristian A Santibañez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rosario Gajardo-Gómez
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Beatriz C Avendaño
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Esteban F Díaz
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Trinidad D Montero
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Victoria Velarde
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago de Chile, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
155
|
Expression of E-prostanoid receptors in nasal polyp tissues of smoking and nonsmoking patients with chronic rhinosinusitis. PLoS One 2018; 13:e0200989. [PMID: 30040868 PMCID: PMC6057645 DOI: 10.1371/journal.pone.0200989] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 07/08/2018] [Indexed: 11/19/2022] Open
Abstract
Background Different inflammatory reactions have been observed in the polyp tissues of nonsmokers and smokers with chronic rhinosinusitis (CRS). E-prostanoid (EP) receptors play a role in the inflammatory processes. Cigarette smoke (CS) exposure regulates EP-receptor expression levels promoting inflammatory mediator release from various inflammatory cells. In this study, we characterize the EP-receptor expression profiles in the polyps of nonsmoking and smoking CRS patients to explore the possible role of CS in the pathogenesis of chronic rhinosinusitis with nasal polyps (CRSwNP). Methods Polyp biopsies were obtained from 28 non-smoking and 21 smoking CRSwNP patients. Histopathological characteristics were observed under a light microscope. The prostaglandin E2 (PGE2), TNF-α, and IL-8 contents in polyp tissues were detected using enzyme-linked immunosorbent assay. Immunostaining was used to locate EP receptors in polyps. Messenger RNA and protein expression of EP receptors were examined using quantitative real-time polymerase chain reaction and Western blot, respectively. Results More severe inflammatory reactions occurred in polyp tissues of smoking CRSwNP patients. The PGE2, TNF-α, and IL-8 in tissue homogenate levels were significantly higher in smoking CRSwNP patients than those in nonsmoking CRSwNP patients. Moreover, the distribution of each EP receptor subtype was similar in both groups. Compared with the EP-receptor expression in nonsmokers, messenger RNA and protein of EP2 and EP4 receptor were significantly down-expressed in smoking patients, but EP1 and EP3 receptors did not show significant differences. Conclusion CS exposure downregulates the expression levels of EP2 and EP4 receptors and stimulates the production of PGE2 and the proinflammatory cytokine IL-8 and TNF-α in polyp tissues of CRS patients. The down-expressed EP2 and EP4 receptors might be associated with severe inflammatory reactions in smoking CRSwNP patients.
Collapse
|
156
|
General Pathways of Pain Sensation and the Major Neurotransmitters Involved in Pain Regulation. Int J Mol Sci 2018; 19:ijms19082164. [PMID: 30042373 PMCID: PMC6121522 DOI: 10.3390/ijms19082164] [Citation(s) in RCA: 316] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/19/2018] [Accepted: 07/20/2018] [Indexed: 12/30/2022] Open
Abstract
Pain has been considered as a concept of sensation that we feel as a reaction to the stimulus of our surrounding, putting us in harm's way and acting as a form of defense mechanism that our body has permanently installed into its system. However, pain leads to a huge chunk of finances within the healthcare system with continuous rehabilitation of patients with adverse pain sensations, which might reduce not only their quality of life but also their productivity at work setting back the pace of our economy. It may not look like a huge deal but factor in pain as an issue for majority of us, it becomes an economical burden. Although pain has been researched into and understood by numerous researches, from its definition, mechanism of action to its inhibition in hopes of finding an absolute solution for victims of pain, the pathways of pain sensation, neurotransmitters involved in producing such a sensation are not comprehensively reviewed. Therefore, this review article aims to put in place a thorough understanding of major pain conditions that we experience-nociceptive, inflammatory and physiologically dysfunction, such as neuropathic pain and its modulation and feedback systems. Moreover, the complete mechanism of conduction is compiled within this article, elucidating understandings from various researches and breakthroughs.
Collapse
|
157
|
Ohba M, Saeki K, Koga T, Okuno T, Kobayashi Y, Yokomizo T. Profiling of bioactive lipids in different dendritic cell subsets using an improved multiplex quantitative LC-MS/MS method. Biochem Biophys Res Commun 2018; 504:562-568. [PMID: 29890138 DOI: 10.1016/j.bbrc.2018.06.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 06/01/2018] [Accepted: 06/07/2018] [Indexed: 12/31/2022]
Abstract
Lipids are an energy source and key components of the cell membrane; however, they are also bioactive mediators of physiological and pathophysiological phenomena. Quantification of bioactive lipids is not easy because they have diverse chemical properties and are present in trace amounts. Here, we improved a multiplex method of quantifying bioactive lipids, thereby enabling measurement of 90 compounds simultaneously. We then used this system to quantify bioactive lipids produced by two subsets of dendritic cells (DCs): all-trans retinoic acid-treated DCs (RA-DCs) (a type of tolerogenic DC (tDC)) and conventional DCs (cDCs). We found that cDCs produced inflammatory lipid mediators such as leukotrienes, whereas RA-DCs produced anti-inflammatory lipid mediators such as prostaglandin I2. Consistent with this, cDCs expressed larger amounts of mRNA encoding 5-lipoxygenase and LTA4 hydrolase (both responsible for leukotriene biosynthesis) and RA-DCs produced larger amounts of mRNA encoding prostaglandin I2 synthase. Taken together, the results suggest that the method is useful for clarifying the roles of bioactive lipids during immune responses.
Collapse
Affiliation(s)
- Mai Ohba
- Department of Biochemistry, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan
| | - Kazuko Saeki
- Department of Biochemistry, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan.
| | - Tomoaki Koga
- Department of Biochemistry, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan; Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Honjo 2-2-1, Chuo-ku, Kumamoto, Japan
| | - Toshiaki Okuno
- Department of Biochemistry, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan
| | - Yuichi Kobayashi
- Department of Biotechnology, Tokyo Institute of Technology, Nagatsuta-cho 4259, Midori-ku, Yokohama, Japan
| | - Takehiko Yokomizo
- Department of Biochemistry, Juntendo University School of Medicine, Hongo 2-1-1, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
158
|
Sharif NA, Klimko PG. Prostaglandin FP receptor antagonists: discovery, pharmacological characterization and therapeutic utility. Br J Pharmacol 2018; 176:1059-1078. [PMID: 29679483 DOI: 10.1111/bph.14335] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 03/19/2018] [Accepted: 03/28/2018] [Indexed: 12/19/2022] Open
Abstract
In contrast to the availability of potent and selective antagonists of several prostaglandin receptor types (including DP1 , DP2 , EP and TP receptors), there has been a paucity of well-characterized, selective FP receptor antagonists. The earliest ones included dimethyl amide and dimethyl amine derivatives of PGF2α , but these have failed to gain prominence. The fluorinated PGF2α analogues, AL-8810 and AL-3138, were subsequently discovered as competitive and non-competitive FP receptor antagonists respectively. Non-prostanoid structures, such as the thiazolidinone AS604872, the D-amino acid-based oligopeptide PDC31 and its peptidomimic analogue PDC113.824 came next, but the latter two are allosteric inhibitors of FP receptor signalling. AL-8810 has a sub-micromolar in vitro potency and ≥2 log unit selectivity against most other PG receptors when tested in several cell- and tissue-based functional assays. Additionally, AL-8810 has demonstrated therapeutic efficacy as an FP receptor antagonist in animal models of stroke, traumatic brain injury, multiple sclerosis, allodynia and endometriosis. Consequently, it appears that AL-8810 has become the FP receptor antagonist of choice. LINKED ARTICLES: This article is part of a themed section on Eicosanoids 35 years from the 1982 Nobel: where are we now? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.8/issuetoc.
Collapse
Affiliation(s)
| | - Peter G Klimko
- Novartis Pharmaceuticals Corporation, Fort Worth, TX, 76134, USA
| |
Collapse
|
159
|
da Cruz Fernandes IM, Pinto RZ, Ferreira P, Lira FS. Low back pain, obesity, and inflammatory markers: exercise as potential treatment. J Exerc Rehabil 2018; 14:168-174. [PMID: 29740548 PMCID: PMC5931150 DOI: 10.12965/jer.1836070.035] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 03/26/2018] [Indexed: 12/21/2022] Open
Abstract
Low back pain is a health issue with significant impact to patients and society. This narrative review aims to synthesize the relationship between obesity, low-grade inflammation and low back pain. It is known that a sedentary lifestyle is a risk factor for obesity and related disorders. The adipose tissue of obese people secretes a range of cytokines of character pro- and anti-inflammatory, with many molecular effects. In addition, pro-inflammatory cytokines are sensitizers of C-reactive protein (CRP), a marker of acute inflammation that can be linked to the musculoskeletal pain sensation individuals with back pain. Another inflammatory marker deserves mention, prostaglandin E2. Prostaglandin E2 is important in the process of triggering actions such as pyrexia, sensation of pain and inflammation, which are exhibited in low back pain condition. The potential for exercises and physical activity to control these mediators and act as a preventative measure for back pain are important because they work as a nonpharmacological strategy to this target audience. There are two types of exercise discussed in this review, the moderate-intensity continuous training and high-intensity interval training.
Collapse
Affiliation(s)
- Isabela Maia da Cruz Fernandes
- Exercise and Immunometabolism Research Group, Department of Physical Education, Faculty of Sciences and Technology, São Paulo State University, Presidente Prudente, São Paulo,
Brazil
| | - Rafael Zambelli Pinto
- Department of Physical Therapy, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte,
Brazil
| | - Paulo Ferreira
- Discipline of Physiotherapy, The University of Sydney – Faculty of Health Sciences, Sydney,
Australia
| | - Fábio Santos Lira
- Exercise and Immunometabolism Research Group, Department of Physical Education, Faculty of Sciences and Technology, São Paulo State University, Presidente Prudente, São Paulo,
Brazil
| |
Collapse
|
160
|
Selectively targeting prostanoid E (EP) receptor-mediated cell signalling pathways: Implications for lung health and disease. Pulm Pharmacol Ther 2018; 49:75-87. [DOI: 10.1016/j.pupt.2018.01.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 01/05/2018] [Accepted: 01/25/2018] [Indexed: 12/18/2022]
|
161
|
Hao H, Hu S, Wan Q, Xu C, Chen H, Zhu L, Xu Z, Meng J, Breyer RM, Li N, Liu DP, FitzGerald GA, Wang M. Protective Role of mPGES-1 (Microsomal Prostaglandin E Synthase-1)-Derived PGE 2 (Prostaglandin E 2) and the Endothelial EP4 (Prostaglandin E Receptor) in Vascular Responses to Injury. Arterioscler Thromb Vasc Biol 2018; 38:1115-1124. [PMID: 29599139 DOI: 10.1161/atvbaha.118.310713] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/12/2018] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Deletion of mPGES-1 (microsomal prostaglandin E synthase-1)-an anti-inflammatory target alternative to COX (cyclooxygenase)-2-attenuates injury-induced neointima formation in mice. This is attributable to the augmented levels of PGI2 (prostacyclin)-a known restraint of the vascular response to injury, acting via IP (I prostanoid receptor). To examine the role of mPGES-1-derived PGE2 (prostaglandin E2) in vascular remodeling without the IP. APPROACH AND RESULTS Mice deficient in both IP and mPGES-1 (DKO [double knockout] and littermate controls [IP KO (knockout)]) were subjected to angioplasty wire injury. Compared with the deletion of IP alone, coincident deletion of IP and mPGES-1 increased neointima formation, without affecting media area. Early pathological changes include impaired reendothelialization and increased leukocyte invasion in neointima. Endothelial cells (ECs), but not vascular smooth muscle cells, isolated from DKOs exhibited impaired cell proliferation. Activation of EP (E prostanoid receptor) 4 (and EP2, to a lesser extent), but not of EP1 or EP3, promoted EC proliferation. EP4 antagonism inhibited proliferation of mPGES-1-competent ECs, but not of mPGES-1-deficient ECs, which showed suppressed PGE2 production. EP4 activation inhibited leukocyte adhesion to ECs in vitro, promoted reendothelialization, and limited neointima formation post-injury in the mouse. Endothelium-restricted deletion of EP4 in mice suppressed reendothelialization, increased neointimal leukocytes, and exacerbated neointimal formation. CONCLUSIONS Removal of the IP receptors unmasks a protective role of mPGES-1-derived PGE2 in limiting injury-induced vascular hyperplasia. EP4, in the endothelial compartment, is essential to promote reendothelialization and restrain neointimal formation after injury. Activating EP4 bears therapeutic potential to prevent restenosis after percutaneous coronary intervention.
Collapse
Affiliation(s)
- Huifeng Hao
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Sheng Hu
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Qing Wan
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Chuansheng Xu
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Hong Chen
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Liyuan Zhu
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Zhenyu Xu
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | - Jian Meng
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.)
| | | | - Nailin Li
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden (N.L.).,Clinical Pharmacology, Karolinska University Hospital, Stockholm, Sweden (N.L.)
| | - De-Pei Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing (D.-P.L.)
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia (G.A.F.)
| | - Miao Wang
- From the State Key Laboratory of Cardiovascular Disease (H.H., S.H., Q.W., C.X., H.C., L.Z., Z.X., J.M., M.W.) .,Clinical Pharmacology Center (M.W.), Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing
| |
Collapse
|
162
|
Gallic Acid-L-Leucine Conjugate Protects Mice against LPS-Induced Inflammation and Sepsis via Correcting Proinflammatory Lipid Mediator Profiles and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1081287. [PMID: 29765489 PMCID: PMC5889890 DOI: 10.1155/2018/1081287] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 12/19/2017] [Accepted: 01/08/2018] [Indexed: 12/20/2022]
Abstract
The pathology of endotoxin LPS-induced sepsis is hallmarked by aberrant production of proinflammatory lipid mediators and nitric oxide (NO). The aim of the present study was to determine whether the new product gallic acid-L-leucine (GAL) conjugate could ameliorate the LPS-induced dysregulation of arachidonic acid metabolism and NO production. We first investigated the effects of GAL conjugate on the expression of proinflammatory enzymes and the production of proinflammatory NO and lipid mediators in mouse macrophage cell line RAW264.7, primary peritoneal macrophages, and mouse model. Western blot analyses revealed that GAL attenuated LPS-induced expression of iNOS, COX-2, and 5-LOX in a concentration-dependent manner. Consistently, probing NO-mediated fluorescence revealed that GAL antagonized the stimulatory effect of LPS on iNOS activity. By profiling of lipid mediators with ESI-MS-based lipidomics, we found that GAL suppressed LPS-induced overproduction of prostaglandin E2, prostaglandin F2, leukotriene B4, and thromboxane B2. We further discovered that GAL might exhibit anti-inflammatory activities by the following mechanisms: (1) suppressing LPS-induced activation of MAP kinases (i.e., ERK1/2, JNK, and p38); (2) reducing the production of reactive oxygen species (ROS); and (3) preventing LPS-induced nuclear translocation of transcription factors NF-κB and AP-1. Consequently, GAL significantly decreased the levels of COX-2 and iNOS expression and the plasma levels of proinflammatory lipid mediators in LPS-treated mice. GAL pretreatment enhanced the survival of mice against LPS-induced endotoxic shock. Taken together, our results suggest that GAL may be a potential anti-inflammatory drug for the treatment of endotoxemia and sepsis.
Collapse
|
163
|
Shankar V, Goddard WA, Kim SK, Abrol R, Liu F. The 3D Structure of Human DP Prostaglandin G-Protein-Coupled Receptor Bound to Cyclopentanoindole Antagonist, Predicted Using the DuplexBiHelix Modification of the GEnSeMBLE Method. J Chem Theory Comput 2018; 14:1624-1642. [PMID: 29268008 DOI: 10.1021/acs.jctc.7b00842] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Prostaglandins play a critical physiological role in both cardiovascular and immune systems, acting through their interactions with 9 prostanoid G protein-coupled receptors (GPCRs). These receptors are important therapeutic targets for a variety of diseases including arthritis, allergies, type 2 diabetes, and cancer. The DP prostaglandin receptor is of interest because it has unique structural and physiological properties. Most notably, DP does not have the 3-6 ionic lock common to Class A GPCRs. However, the lack of X-ray structures for any of the 9 prostaglandin GPCRs hampers the application of structure-based drug design methods to develop more selective and active medications to specific receptors. We predict here 3D structures for the DP prostaglandin GPCR, based on the GEnSeMBLE complete sampling with hierarchical scoring (CS-HS) methodology. This involves evaluating the energy of 13 trillion packings to finally select the best 20 that are stable enough to be relevant for binding to antagonists, agonists, and modulators. To validate the predicted structures, we predict the binding site for the Merck cyclopentanoindole (CPI) selective antagonist docked to DP. We find that the CPI binds vertically in the 1-2-7 binding pocket, interacting favorably with residues R3107.40 and K762.54 with additional interactions with S3137.43, S3167.46, S191.35, etc. This binding site differs significantly from that of antagonists to known Class A GPCRs where the ligand binds in the 3-4-5-6 region. We find that the predicted binding site leads to reasonable agreement with experimental Structure-Activity Relationship (SAR). We suggest additional mutation experiments including K762.54, E1293.49, L1233.43, M2706.40, F2746.44 to further validate the structure, function, and activation mechanism of receptors in the prostaglandin family. Our structures and binding sites are largely consistent and improve upon the predictions by Li et al. ( J. Am. Chem. Soc. 2007 , 129 ( 35 ), 10720 ) that used our earlier MembStruk prediction methodology.
Collapse
Affiliation(s)
- Vishnu Shankar
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - William A Goddard
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Soo-Kyung Kim
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Ravinder Abrol
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| | - Fan Liu
- Materials and Process Simulation Center (139-74) , California Institute of Technology , 1200 E. California Blvd. , Pasadena , California 91125 , United States
| |
Collapse
|
164
|
Abstract
Platelet P2Y1 receptor signalling via RhoGTPases is necessary for platelet-dependent leukocyte recruitment, where no platelet aggregation is observed. We investigated signalling cascades involved in distinct P2Y1-dependent platelet activities in vitro, using specific inhibitors for phospholipase C (PLC) (U73122, to inhibit the canonical pathway), and RhoGTPases: Rac1 (NSC23766) and RhoA (ROCK inhibitor GSK429286). Human platelet rich plasma (for platelet aggregation) or isolated washed platelets (for chemotaxis assays) was treated with U73122, GSK429286 or NSC23766 prior to stimulation with adenosine diphosphate (ADP) or the P2Y1 specific agonist MRS2365. Aggregation, chemotaxis (towards f-MLP), or platelet-induced human neutrophil chemotaxis (PINC) towards macrophage derived chemokine (MDC) was assessed. Molecular docking of ADP and MRS2365 to P2Y1 was analysed using AutoDock Smina followed by GOLD molecular docking in the Accelrys Discovery Studio software. Inhibition of PLC, but not Rac1 or RhoA, suppressed platelet aggregation induced by ADP and MRS2365. In contrast, platelet chemotaxis and PINC, were significantly attenuated by inhibition of platelet Rac1 or RhoA, but not PLC. MRS2365, compared to ADP had a less pronounced effect on P2Y1-induced aggregation, but a similar efficacy to stimulate platelet chemotaxis and PINC, which might be explained by differences in molecular interaction of ADP compared to MRS2365 with the P2Y1 receptor. Platelet P2Y1 receptor activation during inflammation signals through alternate pathways involving Rho GTPases in contrast to canonical P2Y1 receptor induced PLC signalling. This might be explained by selective molecular interactions of ligands within the orthosteric site of the P2Y1 receptor.
Collapse
|
165
|
Caselli G, Bonazzi A, Lanza M, Ferrari F, Maggioni D, Ferioli C, Giambelli R, Comi E, Zerbi S, Perrella M, Letari O, Di Luccio E, Colovic M, Persiani S, Zanelli T, Mennuni L, Piepoli T, Rovati LC. Pharmacological characterisation of CR6086, a potent prostaglandin E 2 receptor 4 antagonist, as a new potential disease-modifying anti-rheumatic drug. Arthritis Res Ther 2018; 20:39. [PMID: 29490676 PMCID: PMC5831858 DOI: 10.1186/s13075-018-1537-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/04/2018] [Indexed: 12/22/2022] Open
Abstract
Background Prostaglandin E2 (PGE2) acts via its EP4 receptor as a cytokine amplifier (e.g., interleukin [IL]-6) and induces the differentiation and expansion of inflammatory T-helper (Th) lymphocytes. These mechanisms play a key role in the onset and progression of rheumatoid arthritis (RA). We present the pharmacological characterisation of CR6086, a novel EP4 receptor antagonist, and provide evidence for its potential as a disease-modifying anti-rheumatic drug (DMARD). Methods CR6086 affinity and pharmacodynamics were studied in EP4-expressing HEK293 cells by radioligand binding and cyclic adenosine monophosphate (cAMP) production, respectively. In immune cells, IL-6 and vascular endothelial growth factor (VEGF) expression were analysed by RT-PCR, and IL-23 and IL-17 release were measured by enzyme-linked immunosorbent assay (ELISA). In collagen-induced arthritis (CIA) models, rats or mice were immunised with bovine collagen type II. Drugs were administered orally (etanercept and methotrexate intraperitoneally) starting at disease onset. Arthritis progression was evaluated by oedema, clinical score and histopathology. Anti-collagen II immunoglobulin G antibodies were measured by ELISA. Results CR6086 showed selectivity and high affinity for the human EP4 receptor (Ki = 16.6 nM) and functioned as a pure antagonist (half-maximal inhibitory concentration, 22 nM) on PGE2-stimulated cAMP production. In models of human immune cells in culture, CR6086 reduced key cytokine players of RA (IL-6 and VEGF expression in macrophages, IL-23 release from dendritic cells, IL-17 release from Th17 cells). In the CIA model of RA in rats and mice, CR6086 significantly improved all features of arthritis: severity, histology, inflammation and pain. In rats, CR6086 was better than the selective cyclooxygenase-2 inhibitor rofecoxib and at least as effective as the Janus kinase inhibitor tofacitinib. In mice, CR6086 and the biologic DMARD etanercept were highly effective, whereas the non-steroidal anti-inflammatory drug naproxen was ineffective. Importantly, in a study of CR6086/methotrexate, combined treatment greatly improved the effect of a fully immunosuppressive dose of methotrexate. Conclusions CR6086 is a novel, potent EP4 antagonist showing favourable immunomodulatory properties, striking DMARD effects in rodents, and anti-inflammatory activity targeted to immune-mediated inflammatory diseases and distinct from the general effects of cyclooxygenase inhibitors. These results support the clinical development of CR6086, both as a stand-alone DMARD and as a combination therapy with methotrexate. The proof-of-concept trial in patients with RA is ongoing. Electronic supplementary material The online version of this article (10.1186/s13075-018-1537-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Albino Bonazzi
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Marco Lanza
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Flora Ferrari
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Daniele Maggioni
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Cristian Ferioli
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Roberto Giambelli
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Eleonora Comi
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Silvia Zerbi
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Marco Perrella
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Ornella Letari
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Elena Di Luccio
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Milena Colovic
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Stefano Persiani
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Tiziano Zanelli
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Laura Mennuni
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | - Tiziana Piepoli
- Rottapharm Biotech, Via Valosa di Sopra 9, I-20900, Monza, MB, Italy
| | | |
Collapse
|
166
|
Koeberle A, Werz O. Natural products as inhibitors of prostaglandin E 2 and pro-inflammatory 5-lipoxygenase-derived lipid mediator biosynthesis. Biotechnol Adv 2018; 36:1709-1723. [PMID: 29454981 DOI: 10.1016/j.biotechadv.2018.02.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Revised: 01/19/2018] [Accepted: 02/14/2018] [Indexed: 12/31/2022]
Abstract
Non-steroidal anti-inflammatory drugs (NSAIDs) inhibit prostanoid formation and represent prevalent therapeutics for treatment of inflammatory disorders. However, NSAIDs are afflicted with severe side effects, which might be circumvented by more selective suppression of pro-inflammatory eicosanoid biosynthesis. This concept led to dual inhibitors of microsomal prostaglandin E2 synthase (mPGES)-1 and 5-lipoxygenase that are crucial enzymes in the biosynthesis of pro-inflammatory prostaglandin E2 and leukotrienes. The potential of their dual inhibition in light of superior efficacy and safety is discussed. Focus is placed on natural products, for which direct inhibition of mPGES-1 and leukotriene biosynthesis has been confirmed.
Collapse
Affiliation(s)
- Andreas Koeberle
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, Jena 07743, Germany.
| | - Oliver Werz
- Chair of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich-Schiller-University Jena, Philosophenweg 14, Jena 07743, Germany.
| |
Collapse
|
167
|
Wan M, Tang X, Rekha RS, Muvva SSVJR, Brighenti S, Agerberth B, Haeggström JZ. Prostaglandin E 2 suppresses hCAP18/LL-37 expression in human macrophages via EP2/EP4: implications for treatment of Mycobacterium tuberculosis infection. FASEB J 2018; 32:2827-2840. [PMID: 29401596 DOI: 10.1096/fj.201701308] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Prostaglandin (PG)E2 is an arachidonic acid-derived lipid mediator that plays an important role in inflammation and immunity. In this study, we demonstrate that PGE2 suppresses basal and 1,25-dihydroxy vitamin D3 (VD3)-induced expression of hCAP18/LL-37 via E prostanoid (EP)2 and EP4 receptors. In humans, VD3 up-regulates vitamin D receptor (VDR) expression and promotes transcription of the cathelicidin hCAP18/LL-37 gene, whereas PGE2 counteracts this effect. We find that PGE2 induces the cAMP/PKA-signaling pathway and enhances the expression of the inhibitory transcription factor cAMP-responsive modulator/inducible cAMP early repressor, which prevents VDR expression and induction of hCAP18/LL-37 in human macrophages. The negative regulation by PGE2 was evident in M1- and M2-polarized human macrophages, although PGE2 displayed more profound inhibitory effects in M2 cells. PGE2 impaired VD3-induced expression of cathelicidin and concomitant activation of autophagy during Mycobacterium tuberculosis (Mtb) infection and facilitated intracellular Mtb growth in human macrophages. An EP4 agonist also significantly promoted Mtb survival in human macrophages. Our results indicate that PGE2 inhibits hCAP18/LL-37 expression, especially VD3-induced cathelicidin and autophagy, which may reduce host defense against Mtb. Accordingly, antagonists of EP4 may constitute a novel adjunctive therapy in Mtb infection.-Wan, M., Tang, X., Rekha, R. S., Muvva, S. S. V. J. R., Brighenti, S., Agerberth, B., Haeggström, J. Z. Prostaglandin E2 suppresses hCAP18/LL-37 expression in human macrophages via EP2/EP4: implications for treatment of Mycobacterium tuberculosis infection.
Collapse
Affiliation(s)
- Min Wan
- Division of Physiology Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Xiao Tang
- Division of Physiology Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Rokeya Sultana Rekha
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska University Hospital, Huddinge, Stockholm, Sweden; and
| | | | - Susanna Brighenti
- Center for Infectious Medicine, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | - Birgitta Agerberth
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska University Hospital, Huddinge, Stockholm, Sweden; and
| | - Jesper Z Haeggström
- Division of Physiology Chemistry II, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
168
|
Baothman BK, Smith J, Kay LJ, Suvarna SK, Peachell PT. Prostaglandin D2 generation from human lung mast cells is catalysed exclusively by cyclooxygenase-1. Eur J Pharmacol 2018; 819:225-232. [DOI: 10.1016/j.ejphar.2017.12.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 11/08/2017] [Accepted: 12/04/2017] [Indexed: 11/28/2022]
|
169
|
Non-asthmatic eosinophilic bronchitis and its relationship with asthma. Pulm Pharmacol Ther 2017; 47:66-71. [DOI: 10.1016/j.pupt.2017.07.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 11/21/2022]
|
170
|
Nisar SP, Lordkipanidzé M, Jones ML, Dawood BB, Murden S, Cunningham MR, Mumford AD, Wilde JT, Watson SP, Lowe GC, Mundell SJ. A novel thromboxane A2 receptor N42S variant results in reduced surface expression and platelet dysfunction. Thromb Haemost 2017; 111:923-32. [DOI: 10.1160/th13-08-0672] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 12/19/2013] [Indexed: 11/05/2022]
Abstract
SummaryA small number of thromboxane receptor variants have been described in patients with a bleeding history that result in platelet dysfunction. We have identified a patient with a history of significant bleeding, who expresses a novel heterozygous thromboxane receptor variant that predicts an asparagine to serine substitution (N42S). This asparagine is conserved across all class A GPCRs, suggesting a vital role for receptor structure and function. We investigated the functional consequences of the TP receptor heterozygous N42S substitution by performing platelet function studies on platelet-rich plasma taken from the patient and healthy controls. We investigated the N42S mutation by expressing the wild-type (WT) and mutant receptor in human embryonic kidney (HEK) cells. Aggregation studies showed an ablation of arachidonic acid responses in the patient, whilst there was right-ward shift of the U46619 concentration response curve (CRC). Thromboxane generation was unaffected. Calcium mobilisation studies in cells lines showed a rightward shift of the U46619 CRC in N42S–expressing cells compared to WT. Radioligand binding studies revealed a reduction in BMax in platelets taken from the patient and in N42S–expressing cells, whilst cell studies confirmed poor surface expression. We have identified a novel thromboxane receptor variant, N42S, which results in platelet dysfunction due to reduced surface expression. It is associated with a significant bleeding history in the patient in whom it was identified. This is the first description of a naturally occurring variant that results in the substitution of this highly conserved residue and confirms the importance of this residue for correct GPCR function.
Collapse
|
171
|
Li HH, Hsu HH, Chang GJ, Chen IC, Ho WJ, Hsu PC, Chen WJ, Pang JHS, Huang CC, Lai YJ. Prostanoid EP 4 agonist L-902,688 activates PPARγ and attenuates pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2017; 314:L349-L359. [PMID: 29146573 DOI: 10.1152/ajplung.00245.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Prostacyclin agonists that bind the prostacyclin receptor (IP) to stimulate cAMP synthesis are effective vasodilators for the treatment of idiopathic pulmonary arterial hypertension (IPAH), but this signaling may occur through nuclear peroxisome proliferator-activated receptor-γ (PPARγ). There is evidence of scant IP and PPARγ expression but stable prostanoid EP4 receptor (EP4) expression in IPAH patients. Both IP and EP4 functionally couple with stimulatory G protein (Gs), which activates signal transduction. We investigated the effect of an EP4-specific agonist on pulmonary arterial remodeling and its regulatory mechanisms in pulmonary arterial smooth muscle cells (PASMCs). Immunoblotting evealed IP, EP4, and PPARγ expression in human pulmonary arterial hypertension (PAH) and monocrotaline (MCT)-induced PAH rat lung tissue. Isolated PASMCs from MCT-induced PAH rats (MCT-PASMCs) were treated with L-902,688, a selective EP4 agonist, to investigate the anti-vascular remodeling effect. Scant expression of IP and PPARγ but stable expression of EP4 was observed in IPAH patient lung tissues and MCT-PASMCs. L-902,688 inhibited IP-insufficient MCT-PASMC proliferation and migration by activating PPARγ in a time- and dose-dependent manner, but these effects were reversed by AH-23848 (an EP4 antagonist) and H-89 [a protein kinase A (PKA) inhibitor], highlighting the crucial role of PPARγ in the activity of this EP4 agonist. L-902,688 attenuated pulmonary arterial remodeling in hypoxic PAH mice and MCT-induced PAH rats; therefore, we conclude that the selective EP4 agonist L-902,688 reverses vascular remodeling by activating PPARγ. This study identified a novel EP4-PKA-PPARγ pathway, and we propose EP4 as a potential therapeutic target for PAH.
Collapse
Affiliation(s)
- Hsin-Hsien Li
- Department of Respiratory Therapy, Chang-Gung University College of Medicine , Tao-Yuan , Taiwan
| | - Hsao-Hsun Hsu
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine , Taipei , Taiwan
| | - Gwo-Jyh Chang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University , Tao-Yuan , Taiwan
| | - I-Chen Chen
- Graduate Institute of Clinical Medical Sciences, Chang Gung University , Tao-Yuan , Taiwan
| | - Wan-Jing Ho
- Cardiovascular Division, Chang Gung Memorial Hospital , Tao-Yuan , Taiwan
| | - Pei-Chen Hsu
- Division of Thoracic Surgery, Department of Surgery, National Taiwan University Hospital and National Taiwan University College of Medicine , Taipei , Taiwan
| | - Wei-Jan Chen
- Cardiovascular Division, Chang Gung Memorial Hospital , Tao-Yuan , Taiwan
| | - Jong-Hwei S Pang
- Graduate Institute of Clinical Medical Sciences, Chang Gung University , Tao-Yuan , Taiwan
| | - Chung-Chi Huang
- Department of Respiratory Therapy, Chang-Gung University College of Medicine , Tao-Yuan , Taiwan.,Division of Thoracic Medicine, Chang Gung Memorial Hospital , Tao-Yuan , Taiwan
| | - Ying-Ju Lai
- Department of Respiratory Therapy, Chang-Gung University College of Medicine , Tao-Yuan , Taiwan.,Cardiovascular Division, Chang Gung Memorial Hospital , Tao-Yuan , Taiwan.,Department of Respiratory Care, Chang-Gung University of Science and Technology, Chia-Yi, Taiwan
| |
Collapse
|
172
|
Araki Y, Suganami A, Endo S, Masuda Y, Fukushima K, Regan JW, Murayama T, Tamura Y, Fujino H. PGE1and E3show lower efficacies than E2to β-catenin-mediated activity as biased ligands of EP4 prostanoid receptors. FEBS Lett 2017; 591:3771-3780. [DOI: 10.1002/1873-3468.12878] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 09/10/2017] [Accepted: 09/19/2017] [Indexed: 12/24/2022]
Affiliation(s)
- Yumi Araki
- Department of Molecular Pharmacology; Graduate School of Pharmaceutical Sciences and Graduate School of Biomedical Sciences; Tokushima University; Japan
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Japan
| | - Akiko Suganami
- Department of Bioinformatics; Graduate School of Medicine; Chiba University; Japan
| | - Suzu Endo
- Department of Molecular Pharmacology; Graduate School of Pharmaceutical Sciences and Graduate School of Biomedical Sciences; Tokushima University; Japan
| | - Yuta Masuda
- Department of Molecular Pharmacology; Graduate School of Pharmaceutical Sciences and Graduate School of Biomedical Sciences; Tokushima University; Japan
| | - Keijo Fukushima
- Department of Molecular Pharmacology; Graduate School of Pharmaceutical Sciences and Graduate School of Biomedical Sciences; Tokushima University; Japan
| | - John W. Regan
- Department of Pharmacology and Toxicology; College of Pharmacy; The University of Arizona; Tucson AZ USA
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology; Graduate School of Pharmaceutical Sciences; Chiba University; Japan
| | - Yutaka Tamura
- Department of Bioinformatics; Graduate School of Medicine; Chiba University; Japan
| | - Hiromichi Fujino
- Department of Molecular Pharmacology; Graduate School of Pharmaceutical Sciences and Graduate School of Biomedical Sciences; Tokushima University; Japan
| |
Collapse
|
173
|
Sharma A, Nakade UP, Jaitley P, Sharma V, Choudhury S, Garg SK. WITHDRAWN: Differential involvement of L- and T-type Ca 2+ channels, store-operated calcium channel (TRPC) and Rho-kinase signaling pathway(s) in PGF 2α-induced contractions in myometrium of non-pregnant and pregnant buffaloes (Bubalus bubalis). Prostaglandins Other Lipid Mediat 2017:S1098-8823(17)30021-7. [PMID: 28916261 DOI: 10.1016/j.prostaglandins.2017.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 09/05/2017] [Accepted: 09/06/2017] [Indexed: 11/30/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at https://www.elsevier.com/about/our-business/policies/article-withdrawal.
Collapse
Affiliation(s)
- Abhishek Sharma
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Udayraj P Nakade
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Pooja Jaitley
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Vipin Sharma
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Soumen Choudhury
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India
| | - Satish Kumar Garg
- Experimental Pharmacology Laboratory, Department of Pharmacology & Toxicology, College of Veterinary Science and Animal Husbandry, U.P. Pandit Deen Dayal Upadhyaya Pashu Chikitsa Vigyan Vishwavidyalaya, Evam Go-Anusandhan Sansthan (DUVASU), Mathura 281001, India.
| |
Collapse
|
174
|
Deriving vascular smooth muscle cells from mesenchymal stromal cells: Evolving differentiation strategies and current understanding of their mechanisms. Biomaterials 2017; 145:9-22. [PMID: 28843066 DOI: 10.1016/j.biomaterials.2017.08.028] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/07/2017] [Accepted: 08/14/2017] [Indexed: 12/11/2022]
Abstract
Vascular smooth muscle cells (VSMCs) play essential roles in regulating blood vessel form and function. Regeneration of functional vascular smooth muscle tissue to repair vascular diseases is an area of intense research in tissue engineering and regenerative medicine. For functional vascular smooth muscle tissue regeneration to become a practical therapy over the next decade, the field will need to have access to VSMC sources that are effective, robust and safe. While pluripotent stem cells hold good future promise to this end, more immediate translation is expected to come from approaches that generate functional VSMCs from adult sources of multipotent adipose-derived and bone marrow-derived mesenchymal stromal cells (ASCs and BMSCs). The research to this end is extensive and is dominated by studies relating to classical biochemical signalling molecules used to induce differentiation of ASCs and BMSCs. However, prolonged use of the biochemical induction factors is costly and can cause potential endotoxin contamination in the culture. Over recent years several non-traditional differentiation approaches have been devised to mimic defined aspects of the native micro-environment in which VSMCs reside to contribute to the differentiation of VSMC-like cells from ASCs and BMSCs. In this review, the promises and limitations of several non-traditional culture approaches (e.g., co-culture, biomechanical, and biomaterial stimuli) targeting VSMC differentiation are discussed. The extensive crosstalk between the underlying signalling cascades are delineated and put into a translational context. It is expected that this review will not only provide significant insight into VSMC differentiation strategies for vascular smooth muscle tissue engineering applications, but will also highlight the fundamental importance of engineering the cellular microenvironment on multiple scales (with consideration of different combinatorial pathways) in order to direct cell differentiation fate and obtain cells of a desired and stable phenotype. These strategies may ultimately be applied to different sources of stem cells in the future for a range of biomaterial and tissue engineering disciplines.
Collapse
|
175
|
Cooper A, Singh S, Hook S, Tyndall JDA, Vernall AJ. Chemical Tools for Studying Lipid-Binding Class A G Protein-Coupled Receptors. Pharmacol Rev 2017; 69:316-353. [PMID: 28655732 DOI: 10.1124/pr.116.013243] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 05/15/2017] [Indexed: 12/16/2022] Open
Abstract
Cannabinoid, free fatty acid, lysophosphatidic acid, sphingosine 1-phosphate, prostanoid, leukotriene, bile acid, and platelet-activating factor receptor families are class A G protein-coupled receptors with endogenous lipid ligands. Pharmacological tools are crucial for studying these receptors and addressing the many unanswered questions surrounding expression of these receptors in normal and diseased tissues. An inherent challenge for developing tools for these lipid receptors is balancing the often lipophilic requirements of the receptor-binding pharmacophore with favorable physicochemical properties to optimize highly specific binding. In this study, we review the radioligands, fluorescent ligands, covalent ligands, and antibodies that have been used to study these lipid-binding receptors. For each tool type, the characteristics and design rationale along with in vitro and in vivo applications are detailed.
Collapse
Affiliation(s)
- Anna Cooper
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sameek Singh
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | - Sarah Hook
- School of Pharmacy, University of Otago, Dunedin, New Zealand
| | | | | |
Collapse
|
176
|
Sommakia S, Baker OJ. Regulation of inflammation by lipid mediators in oral diseases. Oral Dis 2017; 23:576-597. [PMID: 27426637 PMCID: PMC5243936 DOI: 10.1111/odi.12544] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 07/08/2016] [Accepted: 07/13/2016] [Indexed: 02/06/2023]
Abstract
Lipid mediators (LM) of inflammation are a class of compounds derived from ω-3 and ω-6 fatty acids that play a wide role in modulating inflammatory responses. Some LM possess pro-inflammatory properties, while others possess proresolving characteristics, and the class switch from pro-inflammatory to proresolving is crucial for tissue homeostasis. In this article, we review the major classes of LM, focusing on their biosynthesis and signaling pathways, and their role in systemic and, especially, oral health and disease. We discuss the detection of these LM in various body fluids, focusing on diagnostic and therapeutic applications. We also present data showing gender-related differences in salivary LM levels in healthy controls, leading to a hypothesis on the etiology of inflammatory diseases, particularly Sjögren's syndrome. We conclude by enumerating open areas of research where further investigation of LM is likely to result in therapeutic and diagnostic advances.
Collapse
Affiliation(s)
- Salah Sommakia
- School of Dentistry, The University of Utah, Salt Lake City, UT, USA
| | - Olga J. Baker
- School of Dentistry, The University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
177
|
The role of prostaglandins E1 and E2, dinoprostone, and misoprostol in cervical ripening and the induction of labor: a mechanistic approach. Arch Gynecol Obstet 2017; 296:167-179. [DOI: 10.1007/s00404-017-4418-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/30/2017] [Indexed: 12/23/2022]
|
178
|
Pluchart H, Khouri C, Blaise S, Roustit M, Cracowski JL. Targeting the Prostacyclin Pathway: Beyond Pulmonary Arterial Hypertension. Trends Pharmacol Sci 2017; 38:512-523. [DOI: 10.1016/j.tips.2017.03.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/03/2017] [Accepted: 03/06/2017] [Indexed: 01/08/2023]
|
179
|
Honorato Pérez J. Selexipag, a selective prostacyclin receptor agonist in pulmonary arterial hypertension: a pharmacology review. Expert Rev Clin Pharmacol 2017; 10:753-762. [DOI: 10.1080/17512433.2017.1322900] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Jesús Honorato Pérez
- Medicina y Cirugía, Universidad Complutense de Madrid, Madrid, Spain
- Medicina Interna y Farmacología Clínica, Universidad de Navarra, Pamplona, Spain
- Servicio de Farmacología Clínica, Clínica Universidad de Navarra, Pamplona, Spain
| |
Collapse
|
180
|
Lebkowska-Wieruszewska B, De Vito V, Owen H, Poapholatep A, Giorgi M. Pharmacokinetics of grapiprant, a selective EP4
prostaglandin PGE2
receptor antagonist, after 2 mg/kg oral and i.v. administrations in cats. J Vet Pharmacol Ther 2017; 40:e11-e15. [DOI: 10.1111/jvp.12414] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Accepted: 03/31/2017] [Indexed: 01/12/2023]
Affiliation(s)
| | - V. De Vito
- Department of Veterinary Medicine; University of Sassari; Sassari Italy
| | - H. Owen
- School of Veterinary Science; The University of Queensland; Gatton Queensland Australia
| | - A. Poapholatep
- Department of Pharmacology; Faculty of Veterinary Medicine; Kasetsart University; Bangkok Thailand
| | - M. Giorgi
- Department of Veterinary Sciences; University of Pisa; Pisa San Piero a Grado Italy
| |
Collapse
|
181
|
Proschak E, Heitel P, Kalinowsky L, Merk D. Opportunities and Challenges for Fatty Acid Mimetics in Drug Discovery. J Med Chem 2017; 60:5235-5266. [PMID: 28252961 DOI: 10.1021/acs.jmedchem.6b01287] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Fatty acids beyond their role as an endogenous energy source and storage are increasingly considered as signaling molecules regulating various physiological effects in metabolism and inflammation. Accordingly, the molecular targets involved in formation and physiological activities of fatty acids hold significant therapeutic potential. A number of these fatty acid targets are addressed by some of the oldest and most widely used drugs such as cyclooxygenase inhibiting NSAIDs, whereas others remain unexploited. Compounds orthosterically binding to proteins that endogenously bind fatty acids are considered as fatty acid mimetics. On the basis of their structural resemblance, fatty acid mimetics constitute a family of bioactive compounds showing specific binding thermodynamics and following similar pharmacokinetic mechanisms. This perspective systematically evaluates targets for fatty acid mimetics, investigates their common structural characteristics, and highlights demands in their discovery and design. In summary, fatty acid mimetics share particularly favorable characteristics justifying the conclusion that their therapeutic potential vastly outweighs the challenges in their design.
Collapse
Affiliation(s)
- Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Pascal Heitel
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Lena Kalinowsky
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe-University Frankfurt , Max-von-Laue-Straße 9, 60438 Frankfurt, Germany
| |
Collapse
|
182
|
Nagano T, Nishiyama R, Sanada A, Mutaguchi Y, Ioku A, Umeki H, Kishimoto S, Yamanaka D, Kimura SH, Takemura M. Prostaglandin E 2 potentiates interferon-γ-induced nitric oxide production in cultured rat microglia. J Neurochem 2017; 140:605-612. [PMID: 27973680 DOI: 10.1111/jnc.13926] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/26/2022]
Abstract
Prostaglandin E2 (PGE2 ) plays crucial roles in managing microglial activation through the prostanoid EP2 receptor, a PGE2 receptor subtype. In this study, we report that PGE2 enhances interferon-γ (IFN-γ)-induced nitric oxide production in microglia. IFN-γ increased the release of nitrite, a metabolite of nitric oxide, which was augmented by PGE2 , although PGE2 by itself slightly affects nitrite release. The potentiating effect of PGE2 was positively associated with increased expression of inducible nitric oxide synthase. In contrast to nitrite release induced by IFN-γ, lipopolysaccharide-induced nitrite release was not affected by PGE2 . An EP2 agonist, ONO-AE1-259-01 also augmented IFN-γ-induced nitrite release, while an EP1 agonist, ONO-DI-004, an EP3 agonist, ONO-AE-248, or an EP4 agonist, ONO-AE1-329, did not. In addition, the potentiating effect of PGE2 was inhibited by an EP2 antagonist, PF-04418948, but not by an EP1 antagonist, ONO-8713, an EP3 antagonist, ONO-AE3-240, or an EP4 antagonist, ONO-AE3-208, at 10-6 M. Among the EP agonists, ONO-AE1-259-01 alone was able to accumulate cyclic adenosine monophosphate (AMP), and among the EP antagonists, PF-04418948 was the only one able to inhibit PGE2 -increased intracellular cyclic AMP accumulation. On the other hand, IFN-γ promoted phosphorylation of signal transducer and activator of transcription 1, which was not affected by PGE2 . Furthermore, other prostanoid receptor agonists, PGD2 , PGF2α , iloprost, and U-46119, slightly affected IFN-γ-induced nitrite release. These results indicate that PGE2 potentiates IFN-γ-induced nitric oxide production in microglia through the EP2 receptor, which may shed light on one of the pro-inflammatory aspects of PGE2 .
Collapse
Affiliation(s)
- Takayuki Nagano
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ryo Nishiyama
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Ayaka Sanada
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Yukiko Mutaguchi
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Anna Ioku
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Hirohisa Umeki
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Satoshi Kishimoto
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Daisuke Yamanaka
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Shinya H Kimura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| | - Motohiko Takemura
- Department of Pharmacology, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
183
|
O'Sullivan AG, Mulvaney EP, Kinsella BT. Regulation of protein kinase C-related kinase (PRK) signalling by the TPα and TPβ isoforms of the human thromboxane A 2 receptor: Implications for thromboxane- and androgen- dependent neoplastic and epigenetic responses in prostate cancer. Biochim Biophys Acta Mol Basis Dis 2017; 1863:838-856. [PMID: 28108419 DOI: 10.1016/j.bbadis.2017.01.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 01/11/2017] [Accepted: 01/16/2017] [Indexed: 12/11/2022]
Abstract
The prostanoid thromboxane (TX) A2 and its T Prostanoid receptor (the TP) are increasingly implicated in prostate cancer (PCa). Mechanistically, we recently discovered that both TPα and TPβ form functional signalling complexes with members of the protein kinase C-related kinase (PRK) family, AGC- kinases essential for the epigenetic regulation of androgen receptor (AR)-dependent transcription and promising therapeutic targets for treatment of castrate-resistant prostate cancer (CRPC). Critically, similar to androgens, activation of the PRKs through the TXA2/TP signalling axis induces phosphorylation of histone H3 at Thr11 (H3Thr11), a marker of androgen-induced chromatin remodelling and transcriptional activation, raising the possibility that TXA2-TP signalling can mimic and/or enhance AR-induced cellular changes even in the absence of circulating androgens such as in CRPC. Hence the aim of the current study was to investigate whether TXA2/TP-induced PRK activation can mimic and/or enhance AR-mediated cellular responses in the model androgen-responsive prostate adenocarcinoma LNCaP cell line. We reveal that TXA2/TP signalling can act as a neoplastic- and epigenetic-regulator, promoting and enhancing both AR-associated chromatin remodelling (H3Thr11 phosphorylation, WDR5 recruitment and acetylation of histone H4 at lysine 16) and AR-mediated transcriptional activation (e.g of the KLK3/prostate-specific antigen and TMPRSS2 genes) through mechanisms involving TPα/TPβ mediated-PRK1 and PRK2, but not PRK3, signalling complexes. Overall, these data demonstrate that TPα/TPβ can act as neoplastic and epigenetic regulators by mimicking and/or enhancing the actions of androgens within the prostate and provides further mechanistic insights into the role of the TXA2/TP signalling axis in PCa, including potentially in CRPC.
Collapse
Affiliation(s)
- Aine G O'Sullivan
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Eamon P Mulvaney
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - B Therese Kinsella
- UCD School of Biomolecular and Biomedical Sciences, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
184
|
Wang JW, Vu C, Poloso NJ. A Prostacyclin Analog, Cicaprost, Exhibits Potent Anti-Inflammatory Activity in Human Primary Immune Cells and a Uveitis Model. J Ocul Pharmacol Ther 2017; 33:186-192. [PMID: 28072560 DOI: 10.1089/jop.2016.0167] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To investigate the therapeutic potential of a prostacyclin (IP) receptor agonist for ocular inflammation and the effect on immune cells. METHODS The anti-inflammatory activities of cicaprost were determined in primary human monocyte-derived macrophages and human monocyte-derived dendritic cells (MoDC), as well as a lipopolysaccharides (LPS)-induced rat uveitis model. Multiple cytokine release was measured by utilizing Luminex Technology. Prostacyclin (IP) Receptor expression was detected by reverse transcription-polymerase chain receptor. Leukocyte infiltration and protein exudation in the rat uveitis model were measured using a hemocytometer and protein concentration by a NanoDrop instrument. RESULTS Cicapost, an IP receptor agonist, potently inhibits proinflammatory chemokines/cytokine production not only from LPS- or TNFα (tumor necrosis factor-alpha)-induced primary human monocyte-derived macrophages, but also from LPS-stimulated MoDC. While constitutively expressed in macrophages, the IP receptor was inducible by LPS stimulation in MoDCs. In a LPS-induced rat uveitis model, cicaprost efficaciously prevents ocular inflammatory cell and protein leakage, as well as inflammatory cytokine release. CONCLUSION The IP receptor agonist cicaprost is a potent anti-inflammatory agent, implicating that the tightly controlled PGI2/IP signaling pathway is important in regulating inflammation. This response could be harnessed in ocular inflammatory disease where steroids are currently the standard of care.
Collapse
Affiliation(s)
- Jenny W Wang
- Department of Biological Sciences, Allergan , Irvine, California
| | - Chau Vu
- Department of Biological Sciences, Allergan , Irvine, California
| | - Neil J Poloso
- Department of Biological Sciences, Allergan , Irvine, California
| |
Collapse
|
185
|
Wang M, Shibamoto T, Kuda Y, Tanida M, Zhang T, Song J, Mukai K, Kurata Y. The Responses of Pulmonary and Systemic Circulation and Airway to Allergic Mediators in Anesthetized Rats. Biol Pharm Bull 2017; 39:556-63. [PMID: 27040628 DOI: 10.1248/bpb.b15-00864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Lung allergic diseases sometimes accompany pulmonary vaso- and broncho-constriction. Rats are currently used for the experimental study of lung allergies. However, their hemodynamic mechanisms are not fully understood. Therefore the effects of allergic mediators were determined systematically in vivo in rats in terms of pulmonary vascular resistance (PVR), airway pressure (AWP) and total peripheral resistance (TPR). We directly measured pulmonary arterial pressure, left atrial pressure, systemic arterial pressure, central venous pressure and aortic blood flow to determine PVR and TPR, as well as AWP, following injections of platelet-activating factor (PAF), histamine, serotonin, leukotriene (LT) C4, and prostaglandin (PG) D2 in anesthetized open-chest artificially ventilated Sprague-Dawley (SD) rats. PVR was dose-dependently increased by consecutive administration of PAF, LTC4, and PGD2, with the maximal responsiveness being PAF>LTC4>PGD2. However, neither histamine nor serotonin changed PVR. TPR was decreased by all agents except LTC4 which actually increased it. PAF and serotonin, but not the other agents, increased AWP. In conclusion, allergic mediators exert non-uniform actions on pulmonary and systemic circulation and airways in anesthetized SD rats: PAF, LTC4 and PGD2, but not histamine or serotonin, caused substantial pulmonary vasoconstriction; LTC4 yielded systemic vasoconstriction, while the others caused systemic vasodilatation; only two mediators, PAF and serotonin, induce airway constriction.
Collapse
Affiliation(s)
- Mofei Wang
- Department of Physiology II, Kanazawa Medical University
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Exogenous fatty acids and niacin on acute prostaglandin D 2 production in human myeloid cells. J Nutr Biochem 2017; 39:22-31. [DOI: 10.1016/j.jnutbio.2016.09.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 09/22/2016] [Accepted: 09/22/2016] [Indexed: 12/31/2022]
|
187
|
Moreno JJ. Eicosanoid receptors: Targets for the treatment of disrupted intestinal epithelial homeostasis. Eur J Pharmacol 2016; 796:7-19. [PMID: 27940058 DOI: 10.1016/j.ejphar.2016.12.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/30/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022]
Abstract
The importance of cyclooxygenase and lipoxygenase pathways and the consequent eicosanoid synthesis in the physiology and pathophysiology of the intestinal epithelium is currently being established. Each eicosanoid (prostanoid, leukotriene, hydroxyeicosatetraenoic acid) preferentially recognizes one or more receptors coupled to one or more signal-transduction processes. This overview focuses on the role of eicosanoid receptors in the maintenance of intestinal epithelium physiology through the control of proliferation/differentiation/apoptosis processes. Furthermore, it is reported that the role of these receptors on the regulation of the barrier function of the intestinal epithelium have arisen through the regulation of absorption/secretion processes, tight-junction state and the control of the intestinal immune response. Also, this review considers the implication of AA cascade in the disruption of epithelial homeostasis during inflammatory bowel diseases and colorectal cancer as well as the therapeutic values and potential of the eicosanoid receptors as novel targets for the treatments of the pathologies above mentioned.
Collapse
Affiliation(s)
- Juan J Moreno
- Department of Nutrition, Food Sciences and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Nutrition and Food Safety (INSA-UB), University of Barcelona, Avda. Prat de la Riba 171, E-08921 Santa Coloma de Gramenet, Spain.
| |
Collapse
|
188
|
Capra V, Mauri M, Guzzi F, Busnelli M, Accomazzo MR, Gaussem P, Nisar SP, Mundell SJ, Parenti M, Rovati GE. Impaired thromboxane receptor dimerization reduces signaling efficiency: A potential mechanism for reduced platelet function in vivo. Biochem Pharmacol 2016; 124:43-56. [PMID: 27845050 DOI: 10.1016/j.bcp.2016.11.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Accepted: 11/10/2016] [Indexed: 12/16/2022]
Abstract
Thromboxane A2 is a potent mediator of inflammation and platelet aggregation exerting its effects through the activation of a G protein-coupled receptor (GPCR), termed TP. Although the existence of dimers/oligomers in Class A GPCRs is widely accepted, their functional significance still remains controversial. Recently, we have shown that TPα and TPβ homo-/hetero-dimers interact through an interface of residues in transmembrane domain 1 (TM1) whose disruption impairs dimer formation. Here, biochemical and pharmacological characterization of this dimer deficient mutant (DDM) in living cells indicates a significant impairment in its response to agonists. Interestingly, two single loss-of-function TPα variants, namely W29C and N42S recently identified in two heterozygous patients affected by bleeding disorders, match some of the residues mutated in our DDM. These two naturally occurring variants display a reduced potency to TP agonists and are characterized by impaired dimer formation in transfected HEK-293T cells. These findings provide proofs that lack of homo-dimer formation is a crucial process for reduced TPα function in vivo, and might represent one molecular mechanism through which platelet TPα receptor dysfunction affects the patient(s) carrying these mutations.
Collapse
Key Words
- (Z)-7-[(1R,2R,3R,4S)-3-[[2-(phenylcarbamoyl)hydrazinyl]methyl]-7-oxabicyclo[2.2.1]heptan-2-yl]hept-5-enoic acid
- (Z)-7-[(1S,2S,3R,4R)-3-[(E,3S)-3-hydroxyoct-1-enyl]-5-oxabicyclo[2.2.1]heptan-2-yl]hept-5-enoic acid
- (Z)-7-[(1S,2S,3S,4R)-3-[(E,3R)-3-hydroxy-4-(4-iodophenoxy)but-1-enyl]-7-oxabicyclo[2.2.1]heptan-2-yl]hept-5-enoic acid
- (Z)-7-[(1S,3R,4R,5S)-3-[(E,3R)-3-hydroxyoct-1-enyl]-6,6-dimethyl-4-bicyclo[3.1.1]heptanyl]hept-5-enoic acid
- 3-[(3R)-3-[(4-fluorophenyl)sulfonylamino]-1,2,3,4-tetrahydrocarbazol-9-yl]propanoic acid
- Eicosanoids
- G protein coupled receptors
- I-BOP (PubChem CID: 51015454)
- Pinane Thromboxane A2 (PTA2) (PubChem CID: 25834471)
- Platelets
- Ramatroban (PubChem CID: 123879)
- Receptor dimer
- SQ29,548 (PubChem CID: 6437074)
- Signal transduction
- Thromboxane A(2)
- U46619 (PubChem CID: 5311493)
Collapse
Affiliation(s)
- Valérie Capra
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy; Department of Health Science, University of Milan, Milano, Italy.
| | - Mario Mauri
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Francesca Guzzi
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - Marta Busnelli
- CNR, Institute of Neuroscience, University of Milan, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| | - Maria Rosa Accomazzo
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy.
| | - Pascale Gaussem
- Inserm UMR-S1140, Faculte' de Pharmacie, Université Paris Descartes, Sorbonne Paris Cité, Paris and AP-HP, Hopital Européen Georges Pompidou, Service d'Hématologie Biologique, Paris, France.
| | - Shaista P Nisar
- School of Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK.
| | - Stuart J Mundell
- School of Physiology and Pharmacology, University of Bristol, Bristol BS8 1TD, UK.
| | - Marco Parenti
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy.
| | - G Enrico Rovati
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milano, Italy.
| |
Collapse
|
189
|
Woodward DF, Wenthur SL, Rudebush TL, Fan S, Toris CB. Prostanoid Receptor Antagonist Effects on Intraocular Pressure, Supported by Ocular Biodisposition Experiments. J Ocul Pharmacol Ther 2016; 32:606-622. [PMID: 27763812 DOI: 10.1089/jop.2016.0069] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
PURPOSE Since all prostanoid receptors affect intraocular pressure (IOP) and endogenous prostanoids are found in ocular tissues, the pressor effects of prostanoid antagonists were comprehensively evaluated. The absence of effects of most of these antagonists was not entirely anticipated. To ensure no false-negative results, ocular biodisposition studies were conducted. METHODS Monkeys with laser-induced ocular hypertension were used to study antagonist effects on IOP. Ocular biodisposition of each antagonist was assessed in rabbits, with LC/MS/MS analyses of tissue extracts and blood. RESULTS EP1, EP2, EP3, EP4, FP, IP, and TP prostanoid receptor antagonists did not affect IOP, even at a high 1% dose. These studies were followed by ocular biodisposition studies. Striking differences in ocular tissue bioavailability were observed, which were independent of solubility. Only the EP1 antagonist SC-51322 failed to penetrate sufficiently to be bioavailable in the aqueous humor and ciliary body/iris. This obliged testing an alternative EP1 antagonist, namely ONO-8713, to reliably conclude that an EP1 antagonist does not alter IOP. CONCLUSIONS These antagonist studies provided no evidence for individual endogenous prostanoids exerting a meaningful role in regulating IOP. They do reaffirm the critical importance of studying ocular bioavailability for confirming negative data. Large differences among the antagonists in anterior segment and even ocular surface tissue biodisposition were observed in rabbits. It appears from these monkey studies, supported by rabbit ocular bioavailability data, that an absence of drug effect in the eye cannot be adequately substantiated without determination of ocular pharmacokinetics.
Collapse
Affiliation(s)
- David F Woodward
- 1 Department of Biological Sciences, Allergan, Inc. , Irvine, California
| | - Stacey L Wenthur
- 2 Department of Ophthalmology and Visual Science, School of Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Tara L Rudebush
- 2 Department of Ophthalmology and Visual Science, School of Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Shan Fan
- 2 Department of Ophthalmology and Visual Science, School of Medicine, University of Nebraska Medical Center , Omaha, Nebraska
| | - Carol B Toris
- 2 Department of Ophthalmology and Visual Science, School of Medicine, University of Nebraska Medical Center , Omaha, Nebraska.,3 Department of Ophthalmology and Visual Science, School of Medicine, Case Western Reserve University , Cleveland, Ohio
| |
Collapse
|
190
|
Yun B, Leslie CC. Cellular Assays for Evaluating Calcium-Dependent Translocation of cPLA 2α to Membrane. Methods Enzymol 2016; 583:71-99. [PMID: 28063500 DOI: 10.1016/bs.mie.2016.09.006] [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] [Indexed: 12/18/2022]
Abstract
The group IVA phospholipase A2, commonly called cytosolic phospholipase A2α (cPLA2α), is a widely expressed enzyme that hydrolyzes membrane phospholipid to produce arachidonic acid and lysophospholipids, which are precursors for a number of bioactive lipid mediators. Arachidonic acid is metabolized through the cyclooxygenase and lipoxygenase pathways for production of prostaglandins and leukotrienes that regulate normal physiological processes and contribute to disease pathogenesis. cPLA2α is composed of an N-terminal C2 domain and a C-terminal catalytic domain that contains the Ser-Asp catalytic dyad. The catalytic domain contains phosphorylation sites and basic residues that regulate the catalytic activity of cPLA2α. In response to cell stimulation, cPLA2α is rapidly activated by posttranslational mechanisms including increases in intracellular calcium and phosphorylation by mitogen-activated protein kinases. In resting cells, cPLA2α is localized in the cytosol but translocates to membrane including the Golgi, endoplasmic reticulum, and the peri-nuclear membrane in response to increases in intracellular calcium. Calcium binds to the C2 domain, which promotes the interaction of cPLA2α with membrane through hydrophobic interactions. In this chapter, we describe assays used to study the calcium-dependent translocation of cPLA2α to membrane, a regulatory step necessary for access to phospholipid and release of arachidonic acid.
Collapse
Affiliation(s)
- B Yun
- National Jewish Health, Denver, CO, United States
| | - C C Leslie
- National Jewish Health, Denver, CO, United States; University of Colorado Denver, Aurora, CO, United States.
| |
Collapse
|
191
|
Łebkowska-Wieruszewska B, Barsotti G, Lisowski A, Gazzano A, Owen H, Giorgi M. Pharmacokinetics and estimated bioavailability of grapiprant, a novel selective prostaglandin E2 receptor antagonist, after oral administration in fasted and fed dogs. N Z Vet J 2016; 65:19-23. [DOI: 10.1080/00480169.2016.1241727] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
| | - G Barsotti
- Department of Veterinary Sciences, University of Pisa, Via Livornese (lato monte), San Piero a Grado, Pisa, Italy
| | - A Lisowski
- Department of Cattle Breeding and Protection of Genetic Resources, University of Life Sciences, Lublin, Poland
| | - A Gazzano
- Department of Veterinary Sciences, University of Pisa, Via Livornese (lato monte), San Piero a Grado, Pisa, Italy
| | - H Owen
- School of Veterinary Science, The University of Queensland, Gatton Campus, Gatton, Queensland 4343, Australia
| | - M Giorgi
- Department of Veterinary Sciences, University of Pisa, Via Livornese (lato monte), San Piero a Grado, Pisa, Italy
| |
Collapse
|
192
|
Suganami A, Fujino H, Okura I, Yanagisawa N, Sugiyama H, Regan JW, Tamura Y, Murayama T. Human DP and EP2 prostanoid receptors take on distinct forms depending on the diverse binding of different ligands. FEBS J 2016; 283:3931-3940. [PMID: 27636113 DOI: 10.1111/febs.13899] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/12/2016] [Accepted: 09/14/2016] [Indexed: 11/30/2022]
Abstract
Human D-type prostanoid (DP) and E-type prostanoid 2 (EP2) receptors are G protein-coupled receptors and are regarded as the most closely related receptors among prostanoid receptors because they are generated by tandem duplication. The DP receptor-cognate ligand, prostaglandin D2 (PGD2 ) has the ability to activate not only DP receptors but also EP2 receptors. Likewise, the EP2 receptor-cognate ligand, prostaglandin E2 (PGE2 ) has the ability to activate DP receptors in addition to EP receptors in order to stimulate cAMP formation. However, since PGD2 and/or PGE2 activate DP and EP2 receptors to similar maximal levels, that is, their similar efficacies, differences between the ligands in each receptor have not yet been determined in detail except for their different affinities. Herein we demonstrated, using an in silico simulation to predict binding patterns among DP or EP2 receptors and PGD2 , PGE2 , or prostaglandin F2α as the reference prostanoid, that DP and EP2 receptors plausibly take on distinct forms depending on the diverse binding of different ligands. Since these ligands have the potential to make these receptors form distinct conformations with discrete signaling pathways, they are consequently regarded as endogenous biased ligands. Moreover, by using functional assays, the susceptibilities of the DP receptors to the noncognate ligands were approximately 10 times lower than those of EP2 receptors. Thus, EP2 receptors seem to be able to distinguish endogenous ligands better than DP receptors, thereby both receptors are plausibly gaining role-sharing functions with respect to one another as the copies of duplicated gene.
Collapse
Affiliation(s)
- Akiko Suganami
- Department of Bioinformatics, Graduate School of Medicine, Chiba University, Japan
| | - Hiromichi Fujino
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Japan.,Department of Molecular Pharmacology, Faculty of Pharmaceutical Sciences & Institute of Biomedical Sciences, Graduate School, Tokushima University, Japan
| | - Iori Okura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | - Naoki Yanagisawa
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| | | | - John W Regan
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, AZ, USA
| | - Yutaka Tamura
- Department of Bioinformatics, Graduate School of Medicine, Chiba University, Japan
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, Japan
| |
Collapse
|
193
|
Guan NN, Svennersten K, de Verdier PJ, Wiklund NP, Gustafsson LE. Prostaglandin D 2 effects and DP 1 /DP 2 receptor distribution in guinea pig urinary bladder out-flow region. J Cell Mol Med 2016; 21:234-243. [PMID: 27664012 PMCID: PMC5264142 DOI: 10.1111/jcmm.12959] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 07/21/2016] [Indexed: 12/23/2022] Open
Abstract
The proximal urethra and urinary bladder trigone play important roles in continence. We have previously shown that PGD2 is released from guinea pig bladder urothelium/suburothelium and can inhibit detrusor contractile responses. We presently wished to investigate PGD2 actions in guinea pig out‐flow region and the distribution of DP1/DP2 receptors. The effects of PGD2 on urothelium‐intact trigone and proximal urethra contractility were studied in organ bath experiments. Expression of DP1/DP2 receptor proteins was analysed by western blot. Immunohistochemistry was used to identify distribution of DP1/DP2 receptors. PGD2 in a dose‐dependent manner inhibited trigone contractions induced by electrical field stimulation (EFS) and inhibited spontaneous contractions of the proximal urethra. PGD2 was equally (trigone) or slightly less potent (urethra) compared with PGE2. Expression of DP1 and DP2 receptors was found in male guinea pig bladder trigone, neck and proximal urethra. In the trigone and proximal urethra, DP1 receptors were found on the membrane of smooth muscle cells and weak immunoreactivty was observed in the urothelium. DP2 receptors were distributed more widespread, weakly and evenly in the urothelium and smooth muscles. Inhibitory effects by PGD2 on motor activity of guinea pig trigone and proximal urethra are consistent with finding DP1 and DP2 receptors located in the urothelium and smooth muscle cells of the trigone and proximal urethra, and PGD2 may therefore be a modulator of the bladder out‐flow region, possibly having a function in regulation of micturition and a role in overactive bladder syndrome.
Collapse
Affiliation(s)
- Na N Guan
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Svennersten
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Petra J de Verdier
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - N Peter Wiklund
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Lars E Gustafsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
194
|
Tunaru S, Chennupati R, Nüsing RM, Offermanns S. Arachidonic Acid Metabolite 19(S)-HETE Induces Vasorelaxation and Platelet Inhibition by Activating Prostacyclin (IP) Receptor. PLoS One 2016; 11:e0163633. [PMID: 27662627 PMCID: PMC5035018 DOI: 10.1371/journal.pone.0163633] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 09/12/2016] [Indexed: 01/08/2023] Open
Abstract
19(S)-hydroxy-eicosatetraenoic acid (19(S)-HETE) belongs to a family of arachidonic acid metabolites produced by cytochrome P450 enzymes, which play critical roles in the regulation of cardiovascular, renal and pulmonary functions. Although it has been known for a long time that 19(S)-HETE has vascular effects, its mechanism of action has remained unclear. In this study we show that 19(S)-HETE induces cAMP accumulation in the human megakaryoblastic leukemia cell line MEG-01. This effect was concentration-dependent with an EC50 of 520 nM, insensitive to pharmacological inhibition of COX-1/2 and required the expression of the G-protein Gs. Systematic siRNA-mediated knock-down of each G-protein coupled receptor (GPCR) expressed in MEG-01 followed by functional analysis identified the prostacyclin receptor (IP) as the mediator of the effects of 19(S)-HETE, and the heterologously expressed IP receptor was also activated by 19(S)-HETE in a concentration-dependent manner with an EC50 of 567 nM. Pretreatment of isolated murine platelets with 19(S)-HETE blocked thrombin-induced platelets aggregation, an effect not seen in platelets from mice lacking the IP receptor. Furthermore, 19(S)-HETE was able to relax mouse mesenteric artery- and thoracic aorta-derived vessel segments. While pharmacological inhibition of COX-1/2 enzymes had no effect on the vasodilatory activity of 19(S)-HETE these effects were not observed in vessels from mice lacking the IP receptor. These results identify a novel mechanism of action for the CYP450-dependent arachidonic acid metabolite 19(S)-HETE and point to the existence of a broader spectrum of naturally occurring prostanoid receptor agonists.
Collapse
Affiliation(s)
- Sorin Tunaru
- Max-Planck-Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
- * E-mail: (ST); (SO)
| | - Ramesh Chennupati
- Max-Planck-Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
| | - Rolf M. Nüsing
- Institute for Clinical Pharmacology, J.W. Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
| | - Stefan Offermanns
- Max-Planck-Institute for Heart and Lung Research, Department of Pharmacology, Ludwigstr. 43, 61231 Bad Nauheim, Germany
- Medical Faculty, J.W. Goethe University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany
- * E-mail: (ST); (SO)
| |
Collapse
|
195
|
Theiler A, Konya V, Pasterk L, Maric J, Bärnthaler T, Lanz I, Platzer W, Schuligoi R, Heinemann A. The EP1/EP3 receptor agonist 17-pt-PGE 2 acts as an EP4 receptor agonist on endothelial barrier function and in a model of LPS-induced pulmonary inflammation. Vascul Pharmacol 2016; 87:180-189. [PMID: 27664754 DOI: 10.1016/j.vph.2016.09.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 09/16/2016] [Accepted: 09/20/2016] [Indexed: 12/18/2022]
Abstract
Endothelial dysfunction is a hallmark of inflammatory conditions. We recently demonstrated that prostaglandin (PG)E2 enhances the resistance of pulmonary endothelium in vitro and counteracts lipopolysaccharide (LPS)-induced pulmonary inflammation in vivo via EP4 receptors. The aim of this study was to investigate the role of the EP1/EP3 receptor agonist 17-phenyl-trinor-(pt)-PGE2 on acute lung inflammation in a mouse model. In LPS-induced pulmonary inflammation in mice, 17-pt-PGE2 reduced neutrophil infiltration and inhibited vascular leakage. These effects were unaltered by an EP1 antagonist, but reversed by EP4 receptor antagonists. 17-pt-PGE2 increased the resistance of pulmonary microvascular endothelial cells and prevented thrombin-induced disruption of endothelial junctions. Again, these effects were not mediated via EP1 or EP3 but through activation of the EP4 receptor, as demonstrated by the lack of effect of more selective EP1 and EP3 receptor agonists, prevention of these effects by EP4 antagonists and EP4 receptor knock-down by siRNA. In contrast, the aggregation enhancing effect of 17-pt-PGE2 in human platelets was mediated via EP3 receptors. Our results demonstrate that 17-pt-PGE2 enhances the endothelial barrier in vitro on pulmonary microvascular endothelial cells, and accordingly ameliorates the recruitment of neutrophils, via EP4 receptors in vivo. This suggests a beneficial effect of 17-pt-PGE2 on pulmonary inflammatory diseases.
Collapse
Affiliation(s)
- Anna Theiler
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Viktoria Konya
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Lisa Pasterk
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Jovana Maric
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Thomas Bärnthaler
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Ilse Lanz
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Wolfgang Platzer
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Rufina Schuligoi
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| | - Akos Heinemann
- Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitaetsplatz 4, 8010 Graz, Austria.
| |
Collapse
|
196
|
Mastalerz L, Celejewska-Wójcik N, Wójcik K, Gielicz A, Ćmiel A, Ignacak M, Oleś K, Szczeklik A, Sanak M. Induced sputum supernatant bioactive lipid mediators can identify subtypes of asthma. Clin Exp Allergy 2016; 45:1779-89. [PMID: 26449970 DOI: 10.1111/cea.12654] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Revised: 09/20/2015] [Accepted: 09/22/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Induced sputum (IS) allows to measure mediators of asthmatic inflammation in bronchial secretions. The specific role of induced sputum supernatant (ISS) endogenous bioactive lipid mediators in subtypes of asthma is not well understood. OBJECTIVE To investigate the interactions between airway inflammation and clinical phenotypes of asthma, we integrated induced sputum supernatant (ISS) eicosanoids and quantitative assessment of infiltrating cells into new subtypes with the means of latent class analysis (LCA). METHODS One hundred and thirty-nine asthmatics with and without aspirin hypersensitivity underwent sputum induction. High-performance liquid chromatography or gas chromatography coupled with mass spectrometry was used to profile eicosanoids. Nineteen variables covering clinical characteristics, IS inflammatory cells and eicosanoids were considered in the LCA. RESULTS Four phenotypic asthma classes were distinguished. Class 1 with mild-to-moderate asthma, chronic rhinosinusitis (CRS), high PGA2 in ISS and almost equal distribution of inflammation cell patterns. Class 3 subjects also had mild-to-moderate asthma but without upper airway symptoms. Induced sputum was often paucigranulocytic with low levels of lipid mediators. Classes 2 and 4 represented severe asthma with CRS and impaired lung function despite high doses of steroids. High blood and sputum eosinophilia was in line with high cysteinyl leukotrienes and PGD2 in ISS only in class 2. Class 4 subjects tended to have increased sputum neutrophilia and PGE2 in ISS. Aspirin hypersensitivity was most frequent among class 2 subjects. CONCLUSIONS & CLINICAL RELEVANCE The LCA revealed four distinct asthma classes differing in eicosanoid pathways.
Collapse
Affiliation(s)
- L Mastalerz
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - N Celejewska-Wójcik
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - K Wójcik
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - A Gielicz
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - A Ćmiel
- Department of Applied Mathematics, AGH University of Science and Technology, Cracow, Poland
| | - M Ignacak
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - K Oleś
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| | - A Szczeklik
- Department of Applied Mathematics, AGH University of Science and Technology, Cracow, Poland
| | - M Sanak
- Department of Medicine, Jagiellonian University School of Medicine, Cracow, Poland
| |
Collapse
|
197
|
Gill SK, Yao Y, Kay LJ, Bewley MA, Marriott HM, Peachell PT. The anti-inflammatory effects of PGE 2 on human lung macrophages are mediated by the EP 4 receptor. Br J Pharmacol 2016; 173:3099-3109. [PMID: 27460634 DOI: 10.1111/bph.13565] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 07/19/2016] [Accepted: 07/19/2016] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE PGE2 inhibits cytokine generation from human lung macrophages. However, the EP receptor that mediates this beneficial anti-inflammatory effect of PGE2 has not been defined. The aim of this study was to identify the EP receptor by which PGE2 inhibits cytokine generation from human lung macrophages. This was determined by using recently developed EP receptor ligands. EXPERIMENTAL APPROACH The effects of PGE2 and EP-selective agonists on LPS-induced generation of TNF-α and IL-6 from macrophages were evaluated. The effects of EP2 -selective (PF-04852946, PF-04418948) and EP4 -selective (L-161,982, CJ-042794) receptor antagonists on PGE2 responses were studied. The expression of EP receptor subtypes by human lung macrophages was determined by RT-PCR. KEY RESULTS PGE2 inhibited LPS-induced and Streptococcus pneumoniae-induced cytokine generation from human lung macrophages. Analysis of mRNA levels indicated that macrophages expressed EP2 and EP4 receptors. L-902,688 (EP4 receptor-selective agonist) was considerably more potent than butaprost (EP2 receptor-selective agonist) as an inhibitor of TNF-α generation from macrophages. EP2 receptor-selective antagonists had marginal effects on the PGE2 inhibition of TNF-α generation, whereas EP4 receptor-selective antagonists caused rightward shifts in the PGE2 concentration-response curves. CONCLUSIONS AND IMPLICATIONS These studies demonstrate that the EP4 receptor is the principal receptor that mediates the anti-inflammatory effects of PGE2 on human lung macrophages. This suggests that EP4 receptor agonists could be effective anti-inflammatory agents in human lung disease.
Collapse
Affiliation(s)
- Sharonjit K Gill
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Yiwen Yao
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Linda J Kay
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Martin A Bewley
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Helen M Marriott
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK
| | - Peter T Peachell
- Academic Unit of Respiratory Medicine, Department of Infection, Immunity and Cardiovascular Disease, The Medical School (Floor L), University of Sheffield, Sheffield, UK.
| |
Collapse
|
198
|
Jayaraja S, Dakhama A, Yun B, Ghosh M, Lee H, Redente EF, Uhlson CL, Murphy RC, Leslie CC. Cytosolic phospholipase A2 contributes to innate immune defense against Candida albicans lung infection. BMC Immunol 2016; 17:27. [PMID: 27501951 PMCID: PMC4977843 DOI: 10.1186/s12865-016-0165-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 07/25/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The lung is exposed to airborne fungal spores, and fungi that colonize the oral cavity such as Candida albicans, but does not develop disease to opportunistic fungal pathogens unless the immune system is compromised. The Group IVA cytosolic phospholipase A2 (cPLA2α) is activated in response to Candida albicans infection resulting in the release of arachidonic acid for eicosanoid production. Although eicosanoids such as prostaglandins and leukotrienes modulate inflammation and immune responses, the role of cPLA2α and eicosanoids in regulating C. albicans lung infection is not understood. METHODS The responses of cPLA2α(+/+) and cPLA2α(-/-) Balb/c mice to intratracheal instillation of C. albicans were compared. After challenge, we evaluated weight loss, organ fungal burden, and the recruitment of cells and the levels of cytokines and eicosanoids in bronchoalveolar lavage fluid. The ability of macrophages and neutrophils from cPLA2α(+/+) and cPLA2α(-/-) mice to recognize and kill C. albicans was also compared. RESULTS After C. albicans instillation, cPLA2α(+/+) mice recovered a modest weight loss by 48 h and completely cleared fungi from the lung by 12 h with no dissemination to the kidneys. In cPLA2α(-/-) mice, weight loss continued for 72 h, C. albicans was not completely cleared from the lung and disseminated to the kidneys. cPLA2α(-/-) mice exhibited greater signs of inflammation including higher neutrophil influx, and elevated levels of albumin and pro-inflammatory cytokines/chemokines (IL1α, IL1β, TNFα, IL6, CSF2, CXCL1, CCL20) in bronchoalveolar lavage fluid. The amounts of cysteinyl leukotrienes, thromboxane B2 and prostaglandin E2 were significantly lower in bronchoalveolar lavage fluid from C. albicans-infected cPLA2α(-/-) mice compared to cPLA2α(+/+) mice. Alveolar macrophages and neutrophils from uninfected cPLA2α(-/-) mice exhibited less killing of C. albicans in vitro than cells from cPLA2α(+/+) mice. In addition alveolar macrophages from cPLA2α(-/-) mice isolated 6 h after instillation of GFP-C. albicans contained fewer internalized fungi than cPLA2α(+/+) macrophages. CONCLUSIONS The results demonstrate that cPLA2α contributes to immune surveillance and host defense in the lung to prevent infection by the commensal fungus C. albicans and to dampen inflammation.
Collapse
Affiliation(s)
- Sabarirajan Jayaraja
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - Azzeddine Dakhama
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - Bogeon Yun
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - Moumita Ghosh
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - HeeJung Lee
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - Elizabeth F Redente
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA
| | - Charis L Uhlson
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | - Robert C Murphy
- Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA
| | - Christina C Leslie
- Department of Pediatrics, National Jewish Health, 1400 Jackson St., Denver, Colorado, 80206, USA. .,Department of Pharmacology, University of Colorado Denver, Aurora, Colorado, USA. .,Department of Pathology, University of Colorado Denver, Aurora, Colorado, USA.
| |
Collapse
|
199
|
Structural features of subtype-selective EP receptor modulators. Drug Discov Today 2016; 22:57-71. [PMID: 27506873 DOI: 10.1016/j.drudis.2016.08.003] [Citation(s) in RCA: 123] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 06/20/2016] [Accepted: 08/01/2016] [Indexed: 12/11/2022]
Abstract
Prostaglandin E2 is a potent endogenous molecule that binds to four different G-protein-coupled receptors: EP1-4. Each of these receptors is a valuable drug target, with distinct tissue localisation and signalling pathways. We review the structural features of EP modulators required for subtype-selective activity, as well as the structural requirements for improved pharmacokinetic parameters. Novel EP receptor subtype selective agonists and antagonists appear to be valuable drug candidates in the therapy of many pathophysiological states, including ulcerative colitis, glaucoma, bone healing, B cell lymphoma, neurological diseases, among others, which have been studied in vitro, in vivo and in early phase clinical trials.
Collapse
|
200
|
Richter MJ, Gall H, Grimminger J, Grimminger F, Ghofrani HA. Selexipag for the treatment of pulmonary arterial hypertension. Expert Opin Pharmacother 2016; 17:1825-34. [DOI: 10.1080/14656566.2016.1215429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
- Manuel Jonas Richter
- Department of Internal Medicine, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany, member of the German Center of Lung Research (DZL)
- Department of Pneumology, Kerckhoff Heart and Thoracic Center, Bad Nauheim, Germany
| | - Henning Gall
- Department of Internal Medicine, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany, member of the German Center of Lung Research (DZL)
| | - Jan Grimminger
- Department of Internal Medicine, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany, member of the German Center of Lung Research (DZL)
- Center for Pulmonary Hypertension, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Grimminger
- Department of Internal Medicine, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany, member of the German Center of Lung Research (DZL)
| | - Hossein-Ardeschir Ghofrani
- Department of Internal Medicine, University of Giessen and Marburg Lung Center (UGMLC), Giessen, Germany, member of the German Center of Lung Research (DZL)
- Department of Pneumology, Kerckhoff Heart and Thoracic Center, Bad Nauheim, Germany
- Department of Medicine, Imperial College London, London, UK
| |
Collapse
|