1
|
Sternak M, Stojak M, Banasik T, Kij A, Bar A, Pacia MZ, Wojnar-Lason K, Chorazy N, Mohaissen T, Marczyk B, Czyzynska-Cichon I, Berkimbayeva Z, Mika A, Chlopicki S. Vascular ATGL-dependent lipolysis and the activation of cPLA 2-PGI 2 pathway protect against postprandial endothelial dysfunction. Cell Mol Life Sci 2024; 81:125. [PMID: 38467757 PMCID: PMC10927860 DOI: 10.1007/s00018-024-05167-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 01/05/2024] [Accepted: 02/02/2024] [Indexed: 03/13/2024]
Abstract
Adipose triglyceride lipase (ATGL) is involved in lipolysis and displays a detrimental pathophysiological role in cardio-metabolic diseases. However, the organo-protective effects of ATGL-induced lipolysis were also suggested. The aim of this work was to characterize the function of lipid droplets (LDs) and ATGL-induced lipolysis in the regulation of endothelial function. ATGL-dependent LDs hydrolysis and cytosolic phospholipase A2 (cPLA2)-derived eicosanoids production were studied in the aorta, endothelial and smooth muscle cells exposed to exogenous oleic acid (OA) or arachidonic acid (AA). Functional effects of ATGL-dependent lipolysis and subsequent activation of cPLA2/PGI2 pathway were also studied in vivo in relation to postprandial endothelial dysfunction.The formation of LDs was invariably associated with elevated production of endogenous AA-derived prostacyclin (PGI2). In the presence of the inhibitor of ATGL or the inhibitor of cytosolic phospholipase A2, the production of eicosanoids was reduced, with a concomitant increase in the number of LDs. OA administration impaired endothelial barrier integrity in vitro that was further impaired if OA was given together with ATGL inhibitor. Importantly, in vivo, olive oil induced postprandial endothelial dysfunction that was significantly deteriorated by ATGL inhibition, cPLA2 inhibition or by prostacyclin (IP) receptor blockade.In summary, vascular LDs formation induced by exogenous AA or OA was associated with ATGL- and cPLA2-dependent PGI2 production from endogenous AA. The inhibition of ATGL resulted in an impairment of endothelial barrier function in vitro. The inhibition of ATGL-cPLA2-PGI2 dependent pathway resulted in the deterioration of endothelial function upon exposure to olive oil in vivo. In conclusion, vascular ATGL-cPLA2-PGI2 dependent pathway activated by lipid overload and linked to LDs formation in endothelium and smooth muscle cells has a vasoprotective role by counterbalancing detrimental effects of lipid overload on endothelial function.
Collapse
Affiliation(s)
- M Sternak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland.
| | - M Stojak
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - T Banasik
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - A Kij
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - A Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - M Z Pacia
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - K Wojnar-Lason
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
- Medical College, Chair of Pharmacology, Jagiellonian University, Grzegorzecka 16, Krakow, Poland
| | - N Chorazy
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
- Doctoral School of Exact and Natural Sciences, Jagiellonian University, Lojasiewicza 11, Krakow, Poland
| | - T Mohaissen
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - B Marczyk
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - I Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - Z Berkimbayeva
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland
| | - A Mika
- Department of Environmental Analysis, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, Poland
- Department of Pharmaceutical Biochemistry, Medical University of Gdansk, Debinki 1, Gdansk, Poland
| | - S Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, Krakow, Poland.
- Medical College, Chair of Pharmacology, Jagiellonian University, Grzegorzecka 16, Krakow, Poland.
| |
Collapse
|
2
|
Watabe T, Yamahira S, Matsuda M, Terai K. Visual quantification of prostaglandin E 2 discharge from a single cell. Cell Struct Funct 2023; 48:241-249. [PMID: 37813623 PMCID: PMC11496778 DOI: 10.1247/csf.23047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023] Open
Abstract
Calcium transients drive cells to discharge prostaglandin E2 (PGE2). We visualized PGE2-induced protein kinase A (PKA) activation and quantitated PGE2 secreted from a single cell by combining fluorescence microscopy and a simulation model. For this purpose, we first prepared PGE2-producer cells that express either an optogenetic or a chemogenetic calcium channel stimulator: OptoSTIM1 or Gq-DREADD, respectively. Second, we prepared reporter cells expressing the Gs-coupled PGE2 reporter EP2 and the PKA biosensor Booster-PKA, which is based on the principle of Förster resonance energy transfer (FRET). Upon the stimulation-induced triggering of calcium transients, a single producer cell discharges PGE2 to stimulate PKA in the surrounding reporter cells. Due to the flow of the medium, the PKA-activated area exhibited a comet-like smear when HeLa cells were used. In contrast, radial PKA activation was observed when confluent MDCK cells were used, indicating that PGE2 diffusion was restricted to the basolateral space. By fitting the radius of the PKA-activated area to a simulation model based on simple diffusion, we estimated that a single HeLa cell secretes 0.25 fmol PGE2 upon a single calcium transient to activate PKA in more than 1000 neighboring cells. This model also predicts that the PGE2 discharge rate is comparable to the diffusion rate. Thus, our method quantitatively envisions that a single calcium transient affects more than 1000 neighboring cells via PGE2.Key words: prostaglandin E2, imaging, intercellular communication, biosensor, quantification.
Collapse
Affiliation(s)
- Tetsuya Watabe
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Shinya Yamahira
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
| | - Michiyuki Matsuda
- Laboratory of Bioimaging and Cell Signaling, Graduate School of Biostudies, Kyoto University, Kyoto 606-8315, Japan
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8315, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto 606-8315, Japan
| | - Kenta Terai
- Department of Pathology and Biology of Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8315, Japan
| |
Collapse
|
3
|
Jeong M, Ju Y, Kwon H, Kim Y, Hyun KY, Choi GE. Protocatechuic Acid and Syringin from Saussurea neoserrata Nakai Attenuate Prostaglandin Production in Human Keratinocytes Exposed to Airborne Particulate Matter. Curr Issues Mol Biol 2023; 45:5950-5966. [PMID: 37504292 PMCID: PMC10378452 DOI: 10.3390/cimb45070376] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/14/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023] Open
Abstract
Saussurea neoserrata Nakai offers a reliable and efficient source of antioxidants that can help alleviate adverse skin reactions triggered by air pollutants. Air pollutants, such as particulate matter (PM), have the ability to infiltrate the skin and contribute to the higher occurrence of cardiovascular, cerebrovascular, and respiratory ailments. Individuals with compromised skin barriers are particularly susceptible to the impact of PM since it can be absorbed more readily through the skin. This study investigated the impact of protocatechuic acid and syringin, obtained from the n-BuOH extract of S. neoserrata Nakai, on the release of PGE2 and PGD2 induced by PM10. Additionally, it examined the gene expression of the synthesis of PGE2 and PGD2 in human keratinocytes. The findings of this research highlight the potential of utilizing safe and efficient plant-derived antioxidants in dermatological and cosmetic applications to mitigate the negative skin reactions caused by exposure to air pollution.
Collapse
Affiliation(s)
- Myeongguk Jeong
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeongdon Ju
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
- Medical Science Research Center, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hyeokjin Kwon
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Yeeun Kim
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| | - Kyung-Yae Hyun
- Department of Clinical Laboratory Science, Dong-Eui University, Busan 47340, Republic of Korea
| | - Go-Eun Choi
- Department of Biomedical Laboratory Science, College of Health Sciences, Catholic University of Pusan, Busan 46252, Republic of Korea
| |
Collapse
|
4
|
Zhang Y, Sun L, Wang X, Zhou Q. Integrative analysis of HASMCs gene expression profile revealed the role of thrombin in the pathogenesis of atherosclerosis. BMC Cardiovasc Disord 2023; 23:191. [PMID: 37046189 PMCID: PMC10091598 DOI: 10.1186/s12872-023-03211-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 03/28/2023] [Indexed: 04/14/2023] Open
Abstract
We explored the effect of thrombin on human aortic smooth muscle cells (HASMCs) and further analyzed its role in the pathogenesis of atherosclerosis (AS). Thrombin-induced differentially expressed genes (DEGs) in HASMCs were identified by analyzing expression profiles from the GEO. Subsequently, enrichment analysis, GSEA, PPI network, and gene-microRNAs networks were interrogated to identify hub genes and associated pathways. Enrichment analysis results indicated that thrombin causes HASMCs to secrete various pro-inflammatory cytokines and chemokines, exacerbating local inflammatory response in AS. Moreover, we identified 9 HUB genes in the PPI network, which are closely related to the inflammatory response and the promotion of the cell cycle. Additionally, we found that thrombin inhibits lipid metabolism and autophagy of HASMCs, potentially contributing to smooth muscle-derived foam cell formation. Our study deepens a mechanistic understanding of the effect of thrombin on HASMCs and provides new insight into treating AS.
Collapse
Affiliation(s)
- Yichen Zhang
- The Second Hospital of Shandong University, Jinan, Shandong Province, China
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Lin Sun
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Xingsheng Wang
- Jinan Central Hospital, Shandong University, Jinan, Shandong Province, China
| | - Qingbo Zhou
- The Second Hospital of Shandong University, Jinan, Shandong Province, China.
| |
Collapse
|
5
|
Weaver LM, Loftin CD, Zhan CG. Development of pharmacotherapies for abdominal aortic aneurysms. Biomed Pharmacother 2022; 153:113340. [PMID: 35780618 PMCID: PMC9514980 DOI: 10.1016/j.biopha.2022.113340] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 11/23/2022] Open
Abstract
The cardiovascular field is still searching for a treatment for abdominal aortic aneurysms (AAA). This inflammatory disease often goes undiagnosed until a late stage and associated rupture has a high mortality rate. No pharmacological treatment options are available. Three hallmark factors of AAA pathology include inflammation, extracellular matrix remodeling, and vascular smooth muscle dysfunction. Here we discuss drugs for AAA treatment that have been studied in clinical trials by examining the drug targets and data present for each drug's ability to regulate the aforementioned three hallmark pathways in AAA progression. Historically, drugs that were examined in interventional clinical trials for treatment of AAA were repurposed therapeutics. Novel treatments (biologics, small-molecule compounds etc.) have not been able to reach the clinic, stalling out in pre-clinical studies. Here we discuss the backgrounds of previous investigational drugs in hopes of better informing future development of potential therapeutics. Overall, the highlighted themes discussed here stress the importance of both centralized anti-inflammatory drug targets and rigor of translatability. Exceedingly few murine studies have examined an intervention-based drug treatment in halting further growth of an established AAA despite interventional treatment being the therapeutic approach taken to treat AAA in a clinical setting. Additionally, data suggest that a potentially successful drug target may be a central inflammatory biomarker. Specifically, one that can effectively modulate all three hallmark factors of AAA formation, not just inflammation. It is suggested that inhibiting PGE2 formation with an mPGES-1 inhibitor is a leading drug target for AAA treatment to this end.
Collapse
Affiliation(s)
- Lauren M Weaver
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| | - Charles D Loftin
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA; Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, 789 South Limestone Street, Lexington, KY 40536, USA.
| |
Collapse
|
6
|
Araujo P, Espe M, Lucena E, Yang Y, Holen E. Differential production of prostaglandins and prostacyclins by liver and head kidney cells from Atlantic salmon challenged with arachidonic and eicosapentaenoic acids. FISH AND SHELLFISH IMMUNOLOGY REPORTS 2021; 2:100015. [DOI: 10.1016/j.fsirep.2021.100015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/26/2021] [Accepted: 06/28/2021] [Indexed: 10/20/2022] Open
|
7
|
Zhu X, Yao Y, Yang J, Zhengxie J, Li X, Hu S, Zhang A, Dong J, Zhang C, Gan G. COX-2-PGE 2 signaling pathway contributes to hippocampal neuronal injury and cognitive impairment in PTZ-kindled epilepsy mice. Int Immunopharmacol 2020; 87:106801. [PMID: 32702600 DOI: 10.1016/j.intimp.2020.106801] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/01/2020] [Accepted: 07/09/2020] [Indexed: 10/23/2022]
Abstract
Epilepsy is one of the most common neurological diseases. It adversely affects cognitive function. Neuroinflammation has been widely recognized as an important factor involved in the pathophysiology of epilepsy. Cyclooxygenase (COX) is a type of oxidoreductase enzyme that acts in the metabolic pathway converting arachidonic acid to prostaglandins, which mediate inflammatory reactions. The activation of inducible cyclooxygenase-2 (COX-2) is considered to be a precipitating factor of neuroinflammation in the brain. Neuroinflammatory processes in the brain are known to contribute to the cascade of events leading to neuronal injury, which may consequently cause cognitive decline. Here in this study, we showed that pentylenetetrazole (PTZ)-kindled mice exhibited an increased level of COX-2 and its main product prostaglandin E2 (PGE2) along with neuroinflammation and neuronal injury in the hippocampus. Pharmacological inhibition of COX-2 by celecoxib, however, significantly reduced hippocampal neuroinflammation and neuronal injury. Furthermore, inhibition of COX-2 by celecoxib attenuated cognitive impairment in the PTZ-kindled mice, suggesting that COX-2-PGE2 signaling pathway mediated neuroinflammation and neuronal injury contributes to cognitive dysfunction in the PTZ-kindled epilepsy mice. Targeting COX-2-PGE2 signaling pathway in the epileptic brain appears to be a viable strategy for attenuating neuronal injury and preventing cognitive deficits in epilepsy patients.
Collapse
Affiliation(s)
- Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| | - Yuanyuan Yao
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Jiurong Yang
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Junhao Zhengxie
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Xinyan Li
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Sijin Hu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Aifeng Zhang
- Department of Pathology, Medical School of Southeast University, Nanjing, China
| | - Jingde Dong
- Department of Geriatric Neurology, Nanjing Brain Hospital Affiliated with Nanjing Medical University, Nanjing, China
| | - Chenchen Zhang
- Transmission Electron Microscopy Center, Medical School of Southeast University, Nanjing, China
| | - Guangming Gan
- Transmission Electron Microscopy Center, Medical School of Southeast University, Nanjing, China; Department of Genetics and Developmental Biology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
8
|
Spartalis E, Spartalis M, Athanasiou A, Paschou SA, Patelis N, Voudris V, Iliopoulos DC. Endothelium in Aortic Aneurysm Disease: New Insights. Curr Med Chem 2020; 27:1081-1088. [PMID: 31549591 DOI: 10.2174/0929867326666190923151959] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 04/10/2019] [Accepted: 04/28/2019] [Indexed: 01/12/2023]
Abstract
Inflammation is recognized as a fundamental element in the development and growth of aortic aneurysms. Aortic aneurysm is correlated with aortic wall deformities and injury, as a result of inflammation, matrix metalloproteinases activation, oxidative stress, and apoptosis of vascular smooth muscle cells. The endothelial wall has a critical part in the inflammation of the aorta and endothelial heterogeneity has proven to be significant for modeling aneurysm formation. Endothelial shear stress and blood flow affect the aortic wall through hindrance of cytokines and adhesion molecules excreted by endothelial cells, causing reduction of the inflammation process in the media and adventitia. This pathophysiological process results in the disruption of elastic fibers, degradation of collagen fibers, and destruction of vascular smooth muscle cells. Consequently, the aortic wall is impaired due to reduced thickness, decreased mechanical function, and cannot tolerate the impact of blood flow leading to aortic expansion. Surgery is still considered the mainstay therapy for large aortic aneurysms. The prevention of aortic dilation, though, is based on the hinderance of endothelial dysregulation with drugs, the reduction of reactive oxygen and nitrogen species, and also the reduction of pro-inflammatory molecules and metalloproteinases. Further investigations are required to enlighten the emerging role of endothelial cells in aortic disease.
Collapse
Affiliation(s)
- Eleftherios Spartalis
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Michael Spartalis
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Antonios Athanasiou
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Stavroula A Paschou
- Division of Endocrinology and Diabetes, "Aghia Sophia" Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Patelis
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| | - Vassilis Voudris
- Division of Cardiology, Onassis Cardiac Surgery Center, Athens, Greece
| | - Dimitrios C Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research, University of Athens, Medical School, Athens, Greece
| |
Collapse
|
9
|
Sun J, Zheng S, Yang N, Chen B, He G, Zhu T. Dexmedetomidine inhibits apoptosis and expression of COX-2 induced by lipopolysaccharide in primary human alveolar epithelial type 2 cells. Biochem Biophys Res Commun 2019; 517:89-95. [DOI: 10.1016/j.bbrc.2019.07.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 07/06/2019] [Indexed: 01/23/2023]
|
10
|
Schwarzbach SV, Melo CF, Xavier PLP, Roballo KC, Cordeiro YG, Ambrósio CE, Fukumasu H, Carregaro AB. Morphine, but not methadone, inhibits microsomal prostaglandin E synthase-1 and prostaglandin-endoperoxide synthase 2 in lipopolysaccharide-stimulated horse synoviocytes. Biochimie 2019; 160:28-33. [PMID: 30763639 DOI: 10.1016/j.biochi.2019.02.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 02/08/2019] [Indexed: 11/15/2022]
Abstract
Osteoarthritis (OA) is one of the main locomotor disorders in horses. Although nonsteroidal anti-inflammatory drugs are the first-line treatment for OA, opioids could also be used. In previous studies, opioids showed promising anti-inflammatory and analgesic effects. In this study, we aimed to investigate the effects of two opioids (morphine and methadone) against inflammation in lipopolysaccharide (LPS)-stimulated synoviocytes by analyzing microsomal prostaglandin E synthase-1 (mPGES-1) and prostaglandin-endoperoxide synthase 2 (PTGS2) expression. Synoviocytes were obtained from the joints at the distal limbs of dead animals. The cytotoxic effects of LPS, morphine, and methadone were investigated by using a cell viability assay with crystal violet dye. Synoviocytes were treated with LPS, LPS plus morphine, or LPS plus methadone for 3, 6, and 12 h, and mPGES-1 and PTGS2 expression was measured using real-time polymerase chain reaction. LPS, and morphine did not affect the viability of synoviocytes, even at high concentrations. LPS treatment increased mPGES-1 and PTGS2 expression, whereas morphine inhibited the increase in mPGES-1 and PTGS2 expression in LPS-stimulated synoviocytes. Methadone did not inhibit mPGES-1 or PTGS2 expression. These results suggest that morphine may exhibit anti-inflammatory effect; therefore, it might be beneficial for the treatment of OA.
Collapse
Affiliation(s)
- S V Schwarzbach
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - C F Melo
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - P L P Xavier
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - K C Roballo
- Innovative Therapies Group (GDTI), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - Y G Cordeiro
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - C E Ambrósio
- Innovative Therapies Group (GDTI), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - H Fukumasu
- Laboratory of Comparative and Translational Oncology (LOCT), Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil
| | - A B Carregaro
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga, SP, Brazil.
| |
Collapse
|
11
|
Qasem H, Al-Ayadhi L, Bjørklund G, Chirumbolo S, El-Ansary A. Impaired lipid metabolism markers to assess the risk of neuroinflammation in autism spectrum disorder. Metab Brain Dis 2018; 33:1141-1153. [PMID: 29569150 DOI: 10.1007/s11011-018-0206-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/19/2018] [Indexed: 01/19/2023]
Abstract
Autism spectrum disorder (ASD) is a multifactorial disorder caused by an interaction between environmental risk factors and a genetic background. It is characterized by impairment in communication, social interaction, repetitive behavior, and sensory processing. The etiology of ASD is still not fully understood, and the role of neuroinflammation in autism behaviors needs to be further investigated. The aim of the present study was to test the possible association between prostaglandin E2 (PGE2), cyclooxygenase-2 (COX-2), microsomal prostaglandin E synthase-1 (mPGES-1), prostaglandin PGE2 EP2 receptors and nuclear kappa B (NF-κB) and the severity of cognitive disorders, social impairment, and sensory dysfunction. PGE2, COX-2, mPGES-1, PGE2-EP2 receptors and NF-κB as biochemical parameters related to neuroinflammation were determined in the plasma of 47 Saudi male patients with ASD, categorized as mild to moderate and severe as indicated by the Childhood Autism Rating Scale (CARS) or the Social Responsiveness Scale (SRS) or the Short Sensory Profile (SSP) and compared to 46 neurotypical controls. The data indicated that ASD patients have remarkably higher levels of the measured parameters compared to neurotypical controls, except for EP2 receptors that showed an opposite trend. While the measured parameter did not correlate with the severity of social and cognitive dysfunction, PGE2, COX-2, and mPGES-1 were remarkably associated with the dysfunction in sensory processing. NF-κB was significantly increased in relation to age. Based on the discussed data, the positive correlation between PGE2, COX-2, and mPGES-1 confirm the role of PGE2 pathway and neuroinflammation in the etiology of ASD, and the possibility of using PGE2, COX-2 and mPGES-1 as biomarkers of autism severity. NF-κB as inflammatory inducer showed an elevated level in plasma of ASD individuals. Receiver operating characteristic analysis together with predictiveness diagrams proved that the measured parameters could be used as predictive biomarkers of biochemical correlates to ASD.
Collapse
Affiliation(s)
- Hanan Qasem
- Biochemistry Department, Science College, King Saud University, Riyadh, Saudi Arabia
| | - Laila Al-Ayadhi
- Autism Research and Treatment Center, Riyadh, Saudi Arabia
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia
- Physiology Department, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Mo i Rana, Norway
| | - Salvatore Chirumbolo
- Department of Neurological and Movement Sciences, University of Verona, Verona, Italy
| | - Afaf El-Ansary
- Autism Research and Treatment Center, Riyadh, Saudi Arabia.
- Shaik AL-Amodi Autism Research Chair, King Saud University, Riyadh, Saudi Arabia.
- Central laboratory, Female Centre for Scientific and Medical Studies, King Saud University, Riyadh, Saudi Arabia.
- Therapeutic Chemistry Department, National Research Center, Dokki, Guiza, Egypt.
| |
Collapse
|
12
|
Microsomal Prostaglandin E Synthase-1 Expression by Aortic Smooth Muscle Cells Attenuates the Differentiated Phenotype. J Cardiovasc Pharmacol 2017; 68:127-42. [PMID: 27159620 DOI: 10.1097/fjc.0000000000000395] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The development of numerous types of cardiovascular disease is associated with alteration of the vascular smooth muscle cell (SMC) phenotype. We have previously shown that abdominal aortic aneurysm progression in a mouse model of the disease is associated with reduced differentiation of SMCs within the lesion and that cyclooxygenase-2 (COX-2) is critical to initiation and progression of the aneurysms. The current studies used human aortic SMC (hASMC) cultures to better characterize mechanisms responsible for COX-2-dependent modulation of the SMC phenotype. Depending on the culture conditions, hASMCs expressed multiple characteristics of a differentiated and contractile phenotype, or a dedifferentiated and secretory phenotype. The pharmacological inhibition of COX-2 promoted the differentiated phenotype, whereas treatment with the COX-2-derived metabolite prostaglandin E2 (PGE2) increased characteristics of the dedifferentiated phenotype. Furthermore, pharmacological inhibition or siRNA-mediated knockdown of microsomal prostaglandin E synthase-1 (mPGES-1), the enzyme that functions downstream of COX-2 during the synthesis of PGE2, significantly increased expression of characteristics of the differentiated SMC phenotype. Therefore, our findings suggest that COX-2 and mPGES-1-dependent synthesis of PGE2 contributes to a dedifferentiated hASMC phenotype and that mPGES-1 may provide a novel pharmacological target for treatment of cardiovascular diseases where altered SMC differentiation has a causative role.
Collapse
|
13
|
Ozen G, Gomez I, Daci A, Deschildre C, Boubaya L, Teskin O, Uydeş-Doğan BS, Jakobsson PJ, Longrois D, Topal G, Norel X. Inhibition of microsomal PGE synthase-1 reduces human vascular tone by increasing PGI 2 : a safer alternative to COX-2 inhibition. Br J Pharmacol 2017; 174:4087-4098. [PMID: 28675448 DOI: 10.1111/bph.13939] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Revised: 05/29/2017] [Accepted: 06/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND AND PURPOSE The side effects of cyclooxygenase-2 (COX-2) inhibitors on the cardiovascular system could be associated with reduced prostaglandin (PG)I2 synthesis. Microsomal PGE synthase-1 (mPGES-1) catalyses the formation of PGE2 from COX-derived PGH2 . This enzyme is induced under inflammatory conditions and constitutes an attractive target for novel anti-inflammatory drugs. However, it is not known whether mPGES-1 inhibitors could be devoid of cardiovascular side effects. The aim of this study was to compare, in vitro, the effects of mPGES-1 and COX-2 inhibitors on vascular tone in human blood vessels. EXPERIMENTAL APPROACH The vascular tone and prostanoid release from internal mammary artery (IMA) and saphenous vein (SV) incubated for 30 min with inhibitors of mPGES-1 or COX-2 were investigated under normal and inflammatory conditions. KEY RESULTS In inflammatory conditions, mPGES-1 and COX-2 proteins were more expressed, and increased levels of PGE2 and PGI2 were released. COX-2 and NOS inhibitors increased noradrenaline induced vascular contractions in IMA under inflammatory conditions while no effect was observed in SV. Interestingly, the mPGES-1 inhibitor significantly reduced (30-40%) noradrenaline-induced contractions in both vessels. This effect was reversed by an IP (PGI2 receptor) antagonist but not modified by NOS inhibition. Moreover, PGI2 release was increased with the mPGES-1 inhibitor and decreased with the COX-2 inhibitor, while both inhibitors reduced PGE2 release. CONCLUSIONS AND IMPLICATIONS In contrast to COX-2 inhibition, inhibition of mPGES-1 reduced vasoconstriction by increasing PGI2 synthesis. Targeting mPGES-1 could provide a lower risk of cardiovascular side effects, compared with those of the COX-2 inhibitors. LINKED ARTICLES This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.
Collapse
Affiliation(s)
- Gulsev Ozen
- INSERM U1148, Paris, France.,Faculty of Pharmacy, Department of Pharmacology, Istanbul University, Istanbul, Turkey
| | - Ingrid Gomez
- INSERM U1148, Paris, France.,Department of Infection, Immunity and Cardiovascular Disease, School of Medicine and Biomedical Sciences, University of Sheffield, Sheffield, UK
| | - Armond Daci
- Faculty of Pharmacy, Department of Pharmacology, Istanbul University, Istanbul, Turkey
| | | | | | - Onder Teskin
- Department of Cardiovascular Surgery, Aile Hospital, Istanbul, Turkey
| | - B Sonmez Uydeş-Doğan
- Faculty of Pharmacy, Department of Pharmacology, Istanbul University, Istanbul, Turkey
| | - Per-Johan Jakobsson
- Unit of Rheumatology, Department of Medicine Solna, Karolinska Institute and Unit of Rheumatology, Karolinska University Hospital, Stockholm, Sweden
| | - Dan Longrois
- INSERM U1148, Paris, France.,AP-HP CHU X. Bichat, Department of Anesthesia and Intensive Care, University Paris Diderot, Sorbonne Paris-Cité, UMR-S1148, Paris, France
| | - Gokce Topal
- Faculty of Pharmacy, Department of Pharmacology, Istanbul University, Istanbul, Turkey
| | - Xavier Norel
- INSERM U1148, Paris, France.,University Paris Diderot, Sorbonne Paris-Cité, UMR-S1148, Paris, France
| |
Collapse
|
14
|
Decreased vasorelaxation induced by iloprost during acute inflammation in human internal mammary artery. Eur J Pharmacol 2017; 804:31-37. [PMID: 28373136 DOI: 10.1016/j.ejphar.2017.03.060] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 03/27/2017] [Accepted: 03/30/2017] [Indexed: 12/25/2022]
Abstract
Cyclooxygenase-2 (COX-2) induction in human internal mammary arteries (IMA) under inflammatory conditions has been associated with attenuated norepinephrine (NE)-induced vasoconstriction. This effect was associated with increased prostaglandin (PG) E2 and prostacyclin (PGI2) releases. The present study was designed to assess the role of these PG and their receptors (EP and IP, respectively) on the vascular reactivity during acute inflammation. Isolated IMA were cultured in the absence (Control conditions) or presence (Inflammatory conditions) of both interleukin-1 beta (IL-1β) and lipopolysaccharide (LPS). The vasorelaxation and the increased content of cyclic adenosine monophosphate (cAMP) induced by iloprost, a PGI2 analogue, were significantly reduced under inflammatory conditions and restored in preparations cultured with the IP antagonist (CAY10441). Decreased cAMP levels under inflammatory conditions are due to at least increased phosphodiesterase (PDE) 4B expression. On the other hand, PGE2, thromboxane analogues and EP agonists-induced vasoconstrictions were not affected under inflammatory conditions. No vasorelaxation was observed with PGD2, PGE2 or the EP2/4 agonists in pre-contracted IMA. Finally, using RT-qPCR and immunohistochemistry, the COX-2, IP receptor and PGI2 synthase (PGIS) were detected. A significant increase of COX-2 and moderate increase of IP mRNA expression was observed under inflammatory conditions, whereas PGIS mRNA level was not affected. This study demonstrates that PGI2/IP receptor signalling and PGI2-induced relaxation are impaired in human IMA during acute inflammation, whereas the responses induced by other prostanoids are not affected. These results could explain some of the mechanisms of vascular dysfunction reported in inflammatory conditions.
Collapse
|
15
|
Identification of the two-phase mechanism of arachidonic acid regulating inflammatory prostaglandin E2 biosynthesis by targeting COX-2 and mPGES-1. Arch Biochem Biophys 2016; 603:29-37. [DOI: 10.1016/j.abb.2016.04.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 01/15/2023]
|
16
|
Pruzanski W, Kopilov J, Kuksis A. Hydrolysis of lipoproteins by sPLA2's enhances mitogenesis and eicosanoid release from vascular smooth muscle cells: Diverse activity of sPLA2's IIA, V and X. Prostaglandins Other Lipid Mediat 2015; 122:64-8. [PMID: 26711221 DOI: 10.1016/j.prostaglandins.2015.12.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Revised: 12/01/2015] [Accepted: 12/08/2015] [Indexed: 11/30/2022]
Abstract
Mitogenesis of Vascular Smooth Muscle Cells (VSMC) plays an important role in atherogenesis. Until recently, the effect of lipid subfractions has not been clarified. Secretory phospholipases A2 (sPLA2's) hydrolyse glycerophospholipids and release pro-inflammatory lyso-lipids, oxidized and non-oxidized fatty acids and isoprostanes. They localize in the vascular wall. We hypothesized that structurally similar sPLA2's may exert different impact on VSMC. The influence of sPLA2's, IIA, V, X, HDL, LDL, and hydrolysis products was tested on mitogenesis of VSMC, i.e., the early effect on the cell membrane phospholipids, and on PGE2 and LTB4 release, i.e., late effect of Cyclooxygenase and 5-lipooxygenase activity in VSMC. Mitogenesis was significantly enhanced by HDL and LDL, and by products of sPLA2 hydrolysis. Hydrolysis of HDL or LDL enhanced mitogenic activity in order V>X>IIA. The release of PGE2 was enhanced by group X sPLA2 and by HDL hydrolyzed by groups V and X. LDL and its hydrolysis products enhanced the release of PGE2 in order X>V>IIA. The release of LTB4 was markedly increased by LDL and HDL, and by hydrolytic products of group V and X, but not group IIA sPLA2. Our study demonstrates a diverse interaction of pro-inflammatory sPLA2's with HDL and LDL affecting both mitogenesis and eicosanoid release from VSMC, therefore potentially enhancing their pro-atherogenic activity.
Collapse
Affiliation(s)
- Waldemar Pruzanski
- St. Michael's Hopital, Toronto, Canada; University of Toronto, Toronto, Canada.
| | | | - Arnis Kuksis
- The Banting and Best Department of Medical Research, Toronto, Canada; University of Toronto, Toronto, Canada
| |
Collapse
|
17
|
Solà-Villà D, Dilmé JF, Rodríguez C, Soto B, Vila L, Escudero JR, Martínez-González J, Camacho M. Expression and Cellular Localization of 15-Hydroxy-Prostaglandin-Dehydrogenase in Abdominal Aortic Aneurysm. PLoS One 2015; 10:e0136201. [PMID: 26287481 PMCID: PMC4545606 DOI: 10.1371/journal.pone.0136201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 07/30/2015] [Indexed: 01/07/2023] Open
Abstract
PGE2 has been implicated in abdominal aortic aneurysm (AAA) associated hypervascularization. PGE2-metabolism involves 15-hydroxyprostaglandin-dehydrogenase (15-PGDH) the expression of which in AAA is unknown. The aim of this study was to examine the expression and cell distribution of 15-PGDH in AAA. Here, we show that 15-PGDH mRNA levels were significantly higher in aorta samples from patients undergoing AAA repair than in those from healthy multiorgan donors. Consequently, the ratio of metabolized PGE2 secreted by aortic samples was significantly higher in AAA. AAA production of total PGE2 and PGE2 metabolites correlated positively with PGI2 production, while the percentage of metabolized PGE2 correlated negatively with the total amount of PGE2 and with PGI2. Transcript levels of 15-PGDH were statistically associated with leukocyte markers but did not correlate with microvascular endothelial cell markers. Immunohistochemistry revealed 15-PGDH in the areas of leukocyte infiltration in AAA samples, mainly associated with CD45-positive cells, but not in normal aorta samples. We provide new data concerning 15-PGDH expression in human AAA, showing that 15-PGDH is upregulated in AAA and mainly expressed in infiltrating leukocytes. Our data suggest that microvasculature was not involved in PGE2 catabolism, reinforcing the potential role of microvasculature derived PGE2 in AAA-associated hypervascularization.
Collapse
Affiliation(s)
- David Solà-Villà
- Autonomous University of Barcelona, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - Jaime-Félix Dilmé
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
- Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - Cristina Rodríguez
- Cardiovascular Research Center (CSIC-ICCC), Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - Begoña Soto
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
- Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - Luis Vila
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - José-Román Escudero
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
- Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - José Martínez-González
- Cardiovascular Research Center (CSIC-ICCC), Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - Mercedes Camacho
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
- * E-mail:
| |
Collapse
|
18
|
Akasaka H, So SP, Ruan KH. Relationship of the Topological Distances and Activities between mPGES-1 and COX-2 versus COX-1: Implications of the Different Post-Translational Endoplasmic Reticulum Organizations of COX-1 and COX-2. Biochemistry 2015; 54:3707-15. [PMID: 25988363 DOI: 10.1021/acs.biochem.5b00339] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vascular inflammation, prostaglandin E2 (PGE₂) is largely biosynthesized by microsomal PGE₂ synthase-1 (mPGES-1), competing with other downstream eicosanoid-synthesizing enzymes, such as PGIS, a synthase of a vascular protector prostacyclin (PGI₂), to isomerize the cyclooxygenase (COX)-2-derived prostaglandin H2 (PGH₂). In this study, we found that a majority of the product from the cells co-expressing human COX-2, mPGES-1, and PGIS was PGE₂. We hypothesize that the molecular and cellular mechanisms are related to the post-translational endoplasmic reticulum (ER) arrangement of those enzymes. A set of fusion enzymes, COX-2-linker [10 amino acids (aa)]-PGIS and COX-2-linker (22 amino acids)-PGIS, were created as "The Bioruler", in which the 10 and 22 amino acids are defined linkers with known helical structures and distances (14.4 and 30.8 Å, respectively). Our experiments have shown that the efficiency of PGI₂ biosynthesis was reduced when the separation distance increased from 10 to 22 amino acids. When COX-2-10aa-PGIS (with a 14.4 Å separation) was co-expressed with mPGES-1 on the ER membrane, a major product was PGE₂, but not PGI₂. However, expression of COX-2-10aa-PGIS and mPGES-1 on a separated ER with a distance of ≫30.8 Å reduced the level of PGE₂ production. These data indicated that the mPGES-1 is "complex-likely" colocalized with COX-2 within a distance of 14.4 Å. In addition, the cells co-expressing COX-1-10aa-PGIS and mPGES-1 produced PGI₂ mainly, but not PGE₂. This indicates that mPGES-1 is expressed much farther from COX-1. These findings have led to proposed models showing the different post-translational ER organization between COX-2 and COX-1 with respect to the topological arrangement of the mPGES-1 during vascular inflammation.
Collapse
Affiliation(s)
- Hironari Akasaka
- Department of Pharmacological and Pharmaceutical Sciences, Center for Experimental Therapeutics and Pharmacoinformatics, College of Pharmacy, University of Houston, Houston, Texas 77204-5037, United States
| | - Shui-Ping So
- Department of Pharmacological and Pharmaceutical Sciences, Center for Experimental Therapeutics and Pharmacoinformatics, College of Pharmacy, University of Houston, Houston, Texas 77204-5037, United States
| | - Ke-He Ruan
- Department of Pharmacological and Pharmaceutical Sciences, Center for Experimental Therapeutics and Pharmacoinformatics, College of Pharmacy, University of Houston, Houston, Texas 77204-5037, United States
| |
Collapse
|
19
|
Chen Y, Liu H, Xu S, Wang T, Li W. Targeting microsomal prostaglandin E2synthase-1 (mPGES-1): the development of inhibitors as an alternative to non-steroidal anti-inflammatory drugs (NSAIDs). MEDCHEMCOMM 2015. [DOI: 10.1039/c5md00278h] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
AA cascade and several key residues in the 3D structure of mPGES-1.
Collapse
Affiliation(s)
- Yuqing Chen
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | | | - Shuang Xu
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Tianlin Wang
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| | - Wei Li
- Department of Medicinal Chemistry, School of Pharmacy
- Nanjing University of Chinese Medicine
- Nanjing
- China
| |
Collapse
|
20
|
Rehal S, von der Weid PY. Experimental ileitis alters prostaglandin biosynthesis in mesenteric lymphatic and blood vessels. Prostaglandins Other Lipid Mediat 2015; 116-117:37-48. [DOI: 10.1016/j.prostaglandins.2014.11.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/09/2014] [Accepted: 11/11/2014] [Indexed: 01/22/2023]
|
21
|
Camacho M, Piñeiro Z, Alcolea S, García J, Balart J, Terra X, Avilés-Jurado FX, Soler M, Quer M, León X, Vila L. Prostacyclin-synthase expression in head and neck carcinoma patients and its prognostic value in the response to radiotherapy. J Pathol 2014; 235:125-35. [DOI: 10.1002/path.4453] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 09/04/2014] [Accepted: 09/23/2014] [Indexed: 01/09/2023]
Affiliation(s)
- Mercedes Camacho
- Laboratory of Angiology, Vascular Biology and Inflammation; Institute of Biomedical Research (IIB Sant Pau) and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Zenaida Piñeiro
- Laboratory of Angiology, Vascular Biology and Inflammation; Institute of Biomedical Research (IIB Sant Pau) and Universitat Autònoma de Barcelona; Barcelona Spain
- Otorhinolaryngology Department; Hospital de la Santa Creu i Sant Pau and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Sonia Alcolea
- Laboratory of Angiology, Vascular Biology and Inflammation; Institute of Biomedical Research (IIB Sant Pau) and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Jacinto García
- Otorhinolaryngology Department; Hospital de la Santa Creu i Sant Pau and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Josep Balart
- Radiation Oncology Department; Hospital de la Santa Creu i Sant Pau and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Ximena Terra
- Otorhinolaryngology Department; Hospital Universitari de Tarragona Joan XXIII, ISPV, Universitat Rovira i Virgili; Tarragona Spain
| | - Francesc-Xavier Avilés-Jurado
- Otorhinolaryngology Department; Hospital Universitari de Tarragona Joan XXIII, ISPV, Universitat Rovira i Virgili; Tarragona Spain
| | - Marta Soler
- Scientific and Technical Services Platform of the Institute of Biomedical Research (II-B Sant Pau); Barcelona Spain
| | - Miquel Quer
- Otorhinolaryngology Department; Hospital de la Santa Creu i Sant Pau and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Xavier León
- Otorhinolaryngology Department; Hospital de la Santa Creu i Sant Pau and Universitat Autònoma de Barcelona; Barcelona Spain
| | - Luis Vila
- Laboratory of Angiology, Vascular Biology and Inflammation; Institute of Biomedical Research (IIB Sant Pau) and Universitat Autònoma de Barcelona; Barcelona Spain
| |
Collapse
|
22
|
Chen X, Jiang D, Wang J, Chen X, Xu X, Xi P, Fan Y, Zhang X, Guan Y. Prostaglandin E2 EP1 receptor enhances TGF-β1-induced mesangial cell injury. Int J Mol Med 2014; 35:285-93. [PMID: 25352206 DOI: 10.3892/ijmm.2014.1979] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 10/15/2014] [Indexed: 11/05/2022] Open
Abstract
Increasing evidence indicates that transforming growth factor-β1 (TGF-β1) is a pivotal mediator in the pathogenesis of renal fibrosis. Mesangial cells (MCs) are important for glomerular function under both physiological and pathological conditions. Studies have found that the expression level of prostaglandin E2 (PGE2) in MCs increases under high glucose conditions, that PGE2 affects the proliferation and hypertrophy of MCs mainly through the EP1 pathway, and that the proliferation of MCs and the accumulation of extracellular matrix are the main events leading to glomerular fibrosis. In this study, we investigated the effects and mechanisms of action of the EP1 receptor, which is induced by transforming growth factor (TGF)-β1, on the proliferation of mouse MCs, the accumulation of extracellular matrix and the expression of PGE2 synthase. Primary mouse glomerular MCs were isolated from EP1 receptor-deficient mice (EP1-/- mice, in which the EP1 receptor was knocked down) and wild-type (WT) mice (WT MCs). In our preliminary experiments, we found that cell proliferation, as well as the mRNA and protein expression of cyclin D1, proliferating cell nuclear antigen (PCNA), fibronectin (FN), collagen I (ColI), membrane-associated PGE2 synthase-1 (mPGES-1) and cyclooxygenase-2 (COX-2) in the WT MCs were significantly increased following treatment with 10 ng/ml TGF-β1 for 24 h. Compared with the WT MCs, following the knockdown of the EP1 gene, the TGF-β1-induced MC injury was markedly suppressed. The aforementioned changes were notably enhanced following treatment with the EP1 agonist, 17-phenyl trinor PGE2 ethyl amide. Additionally, TGF-β1 induced extracellular signal-regulated kinase (ERK) phosphorylation. We found that the TGF-β1-induced ERK phosphorylation was alleviated by EP1 knockdown and promoted by EP1 expression. These results suggest that the EP1 receptor plays a role in the proliferation of mouse MCs, in the accumulation of extracellular matrix and in the expression of mPGES-1 induced by TGF-β1. Its mechanisms of action are possibly related to the reinforcement of ERK phosphorylation.
Collapse
Affiliation(s)
- Xu Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Daishan Jiang
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Jing Wang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Xiaolan Chen
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Xiaolin Xu
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Peipei Xi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Yaping Fan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Xiaoyan Zhang
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| | - Youfei Guan
- Department of Nephrology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, P.R. China
| |
Collapse
|
23
|
Dilmé JF, Bellmunt S, Camacho M, Solà-Villà D, Romero JM, Escudero JR, Vila L. Influence of cardiovascular risk factors on levels of matrix metalloproteinases 2 and 9 in human abdominal aortic aneurysms. Eur J Vasc Endovasc Surg 2014; 48:374-81. [PMID: 24980077 DOI: 10.1016/j.ejvs.2014.05.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 05/27/2014] [Indexed: 10/25/2022]
Abstract
OBJECTIVE To evaluate the influence of cardiovascular risk factors on levels of matrix metalloproteinases (MMP) 2 and 9 in human abdominal aortic aneurysms (AAA). METHODS Aortic samples were collected from patients who underwent AAA repair (n = 89). Patients were stratified according to the maximum transverse aorta diameter: small diameter (<55 mm), moderate diameter (55-69.9 mm) and large diameter (≥70 mm). Aortic walls were studied using real-time PCR and immunohistochemistry. MMP-2, MMP-9, α-actin, CD45, and CD68 transcript levels were determined relative to β-actin. Quantitative data were expressed as median (IQ-range). RESULTS No differences were found in MMP-2 expression between the patient groups, which was mainly associated with vascular smooth muscle cells (VSMC); however, MMP-9 displayed the maximum level in the moderate-diameter group, associated with infiltrating macrophages. Current smoking (CS) and renal insufficiency (RI) significantly increased local levels of MMP-2 (CS 349.5 [219.5-414.1] vs. no-CS 184.4 [100.0-320.5]; p < .008; RI 286.8 [189.6-410.8] vs. no-RI 177.3 [99.3-326.9]; p = .047). Nevertheless, after stepwise linear regression analysis only CS remained as an independent variable predicting local levels of MMP-2 (p = .002). No risk factors influenced local levels of MMP-9. CONCLUSIONS The results show that local levels of MMP-2, an important factor for AAA development, were increased in current smoking AAA patients. MMP-2 was mainly associated with VSMC. It is suggested that MMP-2 could contribute significantly to the increased AAA growth rate observed in current smoking patients. These findings support inclusion of smokers in screening for aneurysmal disease, and emphasize the need for more aggressive monitoring of aneurysmal disease outside the surgical range in patients who smoke at the time of diagnosis and in those who continue to smoke during follow-up.
Collapse
Affiliation(s)
- J-F Dilmé
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain; Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - S Bellmunt
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain; Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - M Camacho
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain.
| | - D Solà-Villà
- Autonomous University of Barcelona, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - J-M Romero
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain; Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - J-R Escudero
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain; Vascular Surgery Department, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | - L Vila
- Angiology, Vascular Biology and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| |
Collapse
|
24
|
Active smoking increases microsomal PGE2-synthase-1/PGE-receptor-4 axis in human abdominal aortic aneurysms. Mediators Inflamm 2014; 2014:316150. [PMID: 24876670 PMCID: PMC4021751 DOI: 10.1155/2014/316150] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 04/10/2014] [Indexed: 01/19/2023] Open
Abstract
Background. The cyclooxygenase- (COX-) 2/microsomal PGE-synthase- (mPGES-) 1/PGE-receptor- (EP-) 4 axis could play a key role in the physiopathology of abdominal aortic aneurysm (AAA) in humans. In this study, we investigated the influence of cardiovascular risk factors on the expression of the PGE2 pathway in human AAA. Methods. Aortic (n = 89) and plasma (n = 79) samples from patients who underwent AAA repair were collected. Patients were grouped according to risk factors. COX-isoenzymes, mPGES-1, EPs, α-actin, and CD45 and CD68 transcripts levels were quantified by QRT-PCR and plasma PGE2 metabolites by EIA. Results. Current smoking (CS) patients compared to no-CS had significantly higher local levels of mPGES-1 (P = 0.009), EP-4 (P = 0.007), and PGE2 metabolites plasma levels (P = 0.008). In the multiple linear regression analysis, these parameters remained significantly enhanced in CS after adding confounding factors. Results from association studies with cell type markers suggested that the increased mPGES-1/EP-4 levels were mainly associated with microvascular endothelial cells. Conclusions. This study shows that elements of the PGE2 pathway, which play an important role in AAA development, are increased in CS. These results provide insight into the relevance of tobacco smoking in AAA development and reinforce the potential of mPGES-1 and EP-4 as targets for therapy in AAA patients.
Collapse
|
25
|
Mangal D, Uboh CE, Jiang Z, Soma LR. Interleukin-1β inhibits synthesis of 5-lipooxygenase in lipopolysaccharide-stimulated equine whole blood. Prostaglandins Other Lipid Mediat 2014; 108:9-22. [PMID: 24530239 DOI: 10.1016/j.prostaglandins.2014.01.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2013] [Revised: 01/07/2014] [Accepted: 01/28/2014] [Indexed: 12/25/2022]
Abstract
Interleukin-1β (IL-1β) is a pro-inflammatory cytokine. It induces the synthesis of prostaglandin E2 (PGE2) catalyzed by cyclooxygenase (COX) and microsomal prostaglandin E synthase (m-PGES). Besides its pro-inflammatory properties, PGE2 also exhibits anti-inflammatory properties by inhibiting synthesis of 5-lipooxygenase (5-LO) products which are in themselves, pro-inflammatory mediators. Thus, inhibition of 5-LO products is beneficial in regulating immune-responses and pro-inflammatory processes. To investigate the hypothesis that IL-1β is responsible for the increase in the synthesis of PGE2 and in the reduction of 5-LO products, equine whole blood (EWB) was treated with lipopolysaccharide (LPS). In vitro treatment of EWB with LPS resulted in increased expression of IL-1β while expression of 5-LO was suppressed. Quantification of eicosanoids using liquid-chromatography-mass spectrometry/multiple reaction monitoring (LC-MS/MRM) showed increased concentrations of prostaglandins and decreased 5-LO products in LPS-treated EWB. Pretreatment of EWB with IL-1β followed by calcium ionophore A23187 (CI) reduced synthesis of 5-LO products. However, pretreatment of EWB with COX-2 inhibitor (NS-398) or m-PGES-1 inhibitor (CAY 10526) and IL-1β followed with CI resulted in a significant (p<0.0001) increase in 5-LO products. Pretreatment of EWB with phospholipase C inhibitor (U73122) followed with LPS reduced PGE2 production but increased 5-LO products. The result of this study indicated that increased PGE2 production led to reduction in 5-LO products in LPS-treated EWB via IL-1β. However, other pathways, cytokines and mediators may be involved in inhibiting 5-LO products but the present study did not include those other potential pathways. Inhibition of 5-LO products by PGE2 in EWB may regulate the initiation and pathogenesis of inflammatory responses in the horse.
Collapse
Affiliation(s)
- Dipti Mangal
- University of Pennsylvania School of Veterinary Medicine, Department of Clinical Studies, New Bolton Center Campus, 382 West Street Road, Kennett Square, PA 19348, USA
| | - Cornelius E Uboh
- University of Pennsylvania School of Veterinary Medicine, Department of Clinical Studies, New Bolton Center Campus, 382 West Street Road, Kennett Square, PA 19348, USA; PA Equine Toxicology & Research Center, West Chester University, Department of Chemistry, 220 East Rosedale Avenue, West Chester, PA 19382, USA.
| | - Zibin Jiang
- University of Pennsylvania School of Veterinary Medicine, Department of Clinical Studies, New Bolton Center Campus, 382 West Street Road, Kennett Square, PA 19348, USA
| | - Lawrence R Soma
- University of Pennsylvania School of Veterinary Medicine, Department of Clinical Studies, New Bolton Center Campus, 382 West Street Road, Kennett Square, PA 19348, USA
| |
Collapse
|
26
|
Gomez I, Benyahia C, Louedec L, Leséche G, Jacob MP, Longrois D, Norel X. Decreased PGE₂ content reduces MMP-1 activity and consequently increases collagen density in human varicose vein. PLoS One 2014; 9:e88021. [PMID: 24505358 PMCID: PMC3914898 DOI: 10.1371/journal.pone.0088021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 01/02/2014] [Indexed: 11/19/2022] Open
Abstract
UNLABELLED Varicose veins are elongated and dilated saphenous veins. Despite the high prevalence of this disease, its pathogenesis remains unclear. AIMS In this study, we investigated the control of matrix metalloproteinases (MMPs) expression by prostaglandin (PG)E₂ during the vascular wall remodeling of human varicose veins. METHODS AND RESULTS Varicose (small (SDv) and large diameter (LDv)) and healthy saphenous veins (SV) were obtained after surgery. Microsomal and cytosolic PGE-synthases (mPGES and cPGES) protein and mRNA responsible for PGE₂ metabolism were analyzed in all veins. cPGES protein was absent while its mRNA was weakly expressed. mPGES-2 expression was similar in the different saphenous veins. mPGES-1 mRNA and protein were detected in healthy veins and a significant decrease was found in LDv. Additionally, 15-hydroxyprostaglandin dehydrogenase (15-PGDH), responsible for PGE₂ degradation, was over-expressed in varicose veins. These variations in mPGES-1 and 15-PGDH density account for the decreased PGE₂ level observed in varicose veins. Furthermore, a significant decrease in PGE₂ receptor (EP4) levels was also found in SDv and LDv. Active MMP-1 and total MMP-2 concentrations were significantly decreased in varicose veins while the tissue inhibitors of metalloproteinases (TIMP -1 and -2), were significantly increased, probably explaining the increased collagen content found in LDv. Finally, the MMP/TIMP ratio is restored by exogenous PGE₂ in varicose veins and reduced in presence of an EP4 receptor antagonist in healthy veins. CONCLUSIONS In conclusion, PGE₂ could be responsible for the vascular wall thickening in human varicose veins. This mechanism could be protective, strengthening the vascular wall in order to counteract venous stasis.
Collapse
Affiliation(s)
- Ingrid Gomez
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | - Chabha Benyahia
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
| | | | - Guy Leséche
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Vascular and Thoracic Surgery, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | | | - Dan Longrois
- INSERM, U698, Paris, France
- AP-HP CHU X. Bichat, Department of Anesthesia and Intensive Care, University Paris Diderot, Sorbonne Paris-Cité, UMR-S698, Paris, France
| | - Xavier Norel
- INSERM, U698, Paris, France
- University Paris Nord, UMR-S698, Paris, France
- * E-mail:
| |
Collapse
|
27
|
Camacho M, Dilmé J, Solà-Villà D, Rodríguez C, Bellmunt S, Siguero L, Alcolea S, Romero JM, Escudero JR, Martínez-González J, Vila L. Microvascular COX-2/mPGES-1/EP-4 axis in human abdominal aortic aneurysm. J Lipid Res 2013; 54:3506-15. [PMID: 24133193 DOI: 10.1194/jlr.m042481] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We investigated the prostaglandin (PG)E2 pathway in human abdominal aortic aneurysm (AAA) and its relationship with hypervascularization. We analyzed samples from patients undergoing AAA repair in comparison with those from healthy multiorgan donors. Patients were stratified according to maximum aortic diameter: low diameter (LD) (<55 mm), moderate diameter (MD) (55-69.9 mm), and high diameter (HD) (≥70 mm). AAA was characterized by abundant microvessels in the media and adventitia with perivascular infiltration of CD45-positive cells. Like endothelial cell markers, cyclooxygenase (COX)-2 and the microsomal isoform of prostaglandin E synthase (mPGES-1) transcripts were increased in AAA (4.4- and 1.4-fold, respectively). Both enzymes were localized in vascular cells and leukocytes, with maximal expression in the LD group, whereas leukocyte markers display a maximum in the MD group, suggesting that the upregulation of COX-2/mPGES-1 precedes maximal leukocyte infiltration. Plasma and in vitro tissue secreted levels of PGE2 metabolites were higher in AAA than in controls (plasma-controls, 19.9 ± 2.2; plasma-AAA, 38.8 ± 5.5 pg/ml; secretion-normal aorta, 16.5 ± 6.4; secretion-AAA, 72.9 ± 6.4 pg/mg; mean ± SEM). E-prostanoid receptor (EP)-2 and EP-4 were overexpressed in AAA, EP-4 being the only EP substantially expressed and colocalized with mPGES-1 in the microvasculature. Additionally, EP-4 mediated PGE2-induced angiogenesis in vitro. We provide new data concerning mPGES-1 expression in human AAA. Our findings suggest the potential relevance of the COX-2/mPGES-1/EP-4 axis in the AAA-associated hypervascularization.
Collapse
Affiliation(s)
- Mercedes Camacho
- Angiology, Vascular Biology, and Inflammation Laboratory, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gomez I, Foudi N, Longrois D, Norel X. The role of prostaglandin E2 in human vascular inflammation. Prostaglandins Leukot Essent Fatty Acids 2013; 89:55-63. [PMID: 23756023 DOI: 10.1016/j.plefa.2013.04.004] [Citation(s) in RCA: 116] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 10/26/2022]
Abstract
Prostaglandins (PG) are the product of a cascade of enzymes such as cyclooxygenases and PG synthases. Among PG, PGE2 is produced by 3 isoforms of PGE synthase (PGES) and through activation of its cognate receptors (EP1-4), this PG is involved in the pathophysiology of vascular diseases. Some anti-inflammatory drugs (e.g. glucocorticoids, nonsteroidal anti-inflammatory drugs) interfere with its metabolism or effects. Vascular cells can initiate many of the responses associated with inflammation. In human vascular tissue, PGE2 is involved in many physiological processes, such as increasing vascular permeability, cell proliferation, cell migration and control of vascular smooth muscle tone. PGE2 has been shown to contribute to the pathogenesis of atherosclerosis, abdominal aortic aneurysm but also in physiologic/adaptive processes such as angiogenesis. Understanding the roles of PGE2 and its cognate receptors in vascular diseases could help to identify diagnostic and prognostic biomarkers. In addition, from these recent studies new promising therapeutic approaches like mPGES-1 inhibition and/or EP4-antagonism should be investigated.
Collapse
Affiliation(s)
- I Gomez
- INSERM, U698, Paris F-75018, France; University Paris Nord, UMR-S698, Paris F-75018, France
| | | | | | | |
Collapse
|
29
|
Alcolea S, Antón R, Camacho M, Soler M, Alfranca A, Avilés-Jurado FX, Redondo JM, Quer M, León X, Vila L. Interaction between head and neck squamous cell carcinoma cells and fibroblasts in the biosynthesis of PGE2. J Lipid Res 2012; 53:630-42. [PMID: 22308510 DOI: 10.1194/jlr.m019695] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Prostaglandin (PG)E(2) is relevant in tumor biology, and interactions between tumor and stroma cells dramatically influence tumor progression. We tested the hypothesis that cross-talk between head and neck squamous cell carcinoma (HNSCC) cells and fibroblasts could substantially enhance PGE(2) biosynthesis. We observed an enhanced production of PGE(2) in cocultures of HNSCC cell lines and fibroblasts, which was consistent with an upregulation of COX-2 and microsomal PGE-synthase-1 (mPGES-1) in fibroblasts. In cultured endothelial cells, medium from fibroblasts treated with tumor cell-conditioned medium induced in vitro angiogenesis, and in tumor cell induced migration and proliferation, these effects were sensitive to PGs inhibition. Proteomic analysis shows that tumor cells released IL-1, and tumor cell-induced COX-2 and mPGES-1 were suppressed by the IL-1-receptor antagonist. IL-1α levels were higher than those of IL-1β in the tumor cell-conditioning medium and in the secretion from samples obtained from 20 patients with HNSCC. Fractionation of tumor cell-conditioning media indicated that tumor cells secreted mature and unprocessed forms of IL-1. Our results support the concept that tumor-associated fibroblasts are a relevant source of PGE(2) in the tumor mass. Because mPGES-1 seems to be essential for a substantial biosynthesis of PGE(2), these findings also strengthen the concept that mPGES-1 may be \a target for therapeutic intervention in patients with HNSCC.
Collapse
Affiliation(s)
- Sonia Alcolea
- Laboratory of Angiology, Vascular Biology and Inflammation, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Angiotensin II differentially modulates cyclooxygenase-2, microsomal prostaglandin E2 synthase-1 and prostaglandin I2 synthase expression in adventitial fibroblasts exposed to inflammatory stimuli. J Hypertens 2011; 29:529-36. [PMID: 21169864 DOI: 10.1097/hjh.0b013e328342b271] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
AIMS To assess whether angiotensin II (Ang II) modulates key enzymes of the cyclooxygenase (COX)-2/prostanoid pathway, including prostaglandin E synthase-1 (mPGES-1) and prostacyclin synthase (PGIS) in rat aortic adventitial fibroblasts in the presence or absence of an inflammatory stimulus [interleukin (IL)-1β]. METHODS AND RESULTS Fibroblasts stimulated with IL-1β (10 ng/ml, 24 h) and/or Ang II (0.1 μmol/l, 24 h) were used. IL-1β up-regulated COX-2 and mPGES-1 (protein and mRNA) and increased PGI2 and PGE2 release, without altering PGIS protein expression. Ang II did modify neither COX-2 and mPGES-1 expression nor prostanoid levels, but it induced PGIS expression. Interestingly, Ang II further enhanced IL-1β-induced COX-2 expression and PGI2 release and concomitantly reduced IL-1β-induced mPGES-1 expression. The AT1 receptor antagonist losartan prevented the effects of Ang II on IL-1β-induced COX-2 or mPGES-1 expression. IL-1β activated p38 mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinase (ERK)1/2 pathways, and coincubation with Ang II resulted in a higher and more sustained phosphorylation of both MAPK. Inhibition of either p38 MAPK (SB203580) or ERK1/2 (PD98059) reduced COX-2 and mPGES-1 expression in cells treated with IL-1β or the combination of IL-1β and Ang II. Ang II did not modify COX-2 transcriptional activity but increased COX-2 mRNA stability in IL-1β-treated cells; by contrast, it increased PGIS mRNA levels through a transcriptional mechanism. CONCLUSION Ang II differentially modulates key enzymes involved in prostanoid biosynthesis thereby altering the balance between PGI2/PGE2 in vascular cells exposed to inflammatory stimuli.
Collapse
|
31
|
Zhu S, Xue R, Zhao P, Fan FL, Kong X, Zheng S, Han Q, Zhu Y, Wang N, Yang J, Guan Y. Targeted disruption of the prostaglandin E2 E-prostanoid 2 receptor exacerbates vascular neointimal formation in mice. Arterioscler Thromb Vasc Biol 2011; 31:1739-47. [PMID: 21636806 DOI: 10.1161/atvbaha.111.226142] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Restenosis after angioplasty remains a major clinical problem. Prostaglandin E(2) (PGE(2)) plays an important role in vascular homeostasis. The PGE(2) receptor E-prostanoid 2 (EP2) is involved in the proliferation and migration of various cell types. We aimed to determine the role of EP2 in the pathogenesis of neointimal formation after vascular injury. METHODS AND RESULTS Wire-mediated vascular injury was induced in the femoral arteries of male wild-type (EP2+/+) and EP2 gene-deficient (EP2-/-) mice. In EP2+/+ mice, EP2 mRNA expression was increased in injured vessels for at least 4 weeks after vascular injury. Neointimal hyperplasia was markedly accelerated in EP2-/- mice, which was associated with increased proliferation and migration of vascular smooth muscle cells (VSMCs) and increased cyclin D1 expression in the neointima layer. Platelet-derived growth factor-BB (PDGF-BB) treatment resulted in more significant cell proliferation and migration in VSMCs of EP2-/- mice than in those of EP2+/+ mice. Activation and overexpression of EP2 attenuated PDGF-BB-elicited cell proliferation and migration, induced G(1)→S-phase arrest and reduced PDGF-BB-stimulated extracellular signal-regulated kinase phosphorylation in EP2+/+ VSMCs. CONCLUSIONS These findings reveal a novel role of the EP2 receptor in neointimal hyperplasia after arterial injury. The EP2 receptor may represent a potential therapeutic target for restenosis after angioplasty.
Collapse
Affiliation(s)
- Sen Zhu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Båge T, Kats A, Lopez BS, Morgan G, Nilsson G, Burt I, Korotkova M, Corbett L, Knox AJ, Pino L, Jakobsson PJ, Modéer T, Yucel-Lindberg T. Expression of prostaglandin E synthases in periodontitis immunolocalization and cellular regulation. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1676-88. [PMID: 21435451 PMCID: PMC3078457 DOI: 10.1016/j.ajpath.2010.12.048] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2010] [Revised: 12/10/2010] [Accepted: 12/22/2010] [Indexed: 12/17/2022]
Abstract
The inflammatory mediator prostaglandin E(2) (PGE(2)) is implicated in the pathogenesis of chronic inflammatory diseases including periodontitis; it is synthesized by cyclooxygenases (COX) and the prostaglandin E synthases mPGES-1, mPGES-2, and cPGES. The distribution of PGES in gingival tissue of patients with periodontitis and the contribution of these enzymes to inflammation-induced PGE(2) synthesis in different cell types was investigated. In gingival biopsies, positive staining for PGES was observed in fibroblasts and endothelial, smooth muscle, epithelial, and immune cells. To further explore the contribution of PGES to inflammation-induced PGE(2) production, in vitro cell culture experiments were performed using fibroblasts and endothelial, smooth muscle, and mast cells. All cell types expressed PGES and COX-2, resulting in basal levels of PGE(2) synthesis. In response to tumor necrosis factor (TNF-α), IL-1β, and cocultured lymphocytes, however, mPGES-1 and COX-2 protein expression increased in fibroblasts and smooth muscle cells, accompanied by increased PGE(2), whereas mPGES-2 and cPGES were unaffected. In endothelial cells, TNF-α increased PGE(2) production only via COX-2 expression, whereas in mast cells the cytokines did not affect PGE(2) enzyme expression or PGE(2) production. Furthermore, PGE(2) production was diminished in gingival fibroblasts derived from mPGES-1 knockout mice, compared with wild-type fibroblasts. These results suggest that fibroblasts and smooth muscle cells are important sources of mPGES-1, which may contribute to increased PGE(2) production in the inflammatory condition periodontitis.
Collapse
Affiliation(s)
- Tove Båge
- Division of Pediatric Dentistry, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anna Kats
- Division of Pediatric Dentistry, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Blanca Silva Lopez
- Division of Pediatric Dentistry, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gareth Morgan
- Division of Pathology, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Nilsson
- Clinical Immunology and Allergy Unit, Karolinska Institutet, Stockholm, Sweden
| | - Idil Burt
- Department of Orthodontics, Eastman Institute, Stockholm, Sweden
| | | | - Lisa Corbett
- Division of Respiratory Medicine, City Hospital, University of Nottingham, Nottingham, United Kingdom
| | - Alan J. Knox
- Division of Respiratory Medicine, City Hospital, University of Nottingham, Nottingham, United Kingdom
| | - Leonardo Pino
- Örebro City Council, Postgraduate Dental Education Center, Örebro, Sweden
| | | | - Thomas Modéer
- Division of Pediatric Dentistry, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Tülay Yucel-Lindberg
- Division of Pediatric Dentistry, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
33
|
Camacho M, Rodríguez C, Guadall A, Alcolea S, Orriols M, Escudero JR, Martínez-González J, Vila L. Hypoxia upregulates PGI-synthase and increases PGI₂ release in human vascular cells exposed to inflammatory stimuli. J Lipid Res 2011; 52:720-31. [PMID: 21296955 DOI: 10.1194/jlr.m011007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hypoxia affects vascular function and cell metabolism, survival, growth, and motility; these processes are partially regulated by prostanoids. We analyzed the effect of hypoxia and inflammation on key enzymes involved in prostanoid biosynthesis in human vascular cells. In human vascular smooth muscle cells (VSMC), hypoxia and interleukin (IL)-1β synergistically increased prostaglandin (PG)I₂ but not PGE₂ release, thereby increasing the PGI₂/PGE₂ ratio. Concomitantly, these stimuli upregulated cyclooxygenase-2 (COX-2) expression (mRNA and protein) and COX activity. Interestingly, hypoxia enhanced PGI-synthase (PGIS) expression and activity in VSMC and human endothelial cells. Hypoxia did not significantly modify the inducible microsomal-PGE-synthase (mPGES)-1. Hypoxia-inducible factor (HIF)-1α-silencing abrogated hypoxia-induced PGIS upregulation. PGIS transcriptional activity was enhanced by hypoxia; however, the minimal PGIS promoter responsive to hypoxia (-131 bp) did not contain any putative hypoxia response element (HRE), suggesting that HIF-1 does not directly drive PGIS transcription. Serial deletion and site-directed mutagenesis studies suggested several transcription factors participate cooperatively. Plasma levels of the stable metabolite of PGI₂ and PGIS expression in several tissues were also upregulated in mice exposed to hypoxia. These data suggest that PGIS upregulation is part of the adaptive response of vascular cells to hypoxic stress and could play a role in counteracting the deleterious effect of inflammatory stimuli.
Collapse
Affiliation(s)
- Mercedes Camacho
- Angiology, Vascular Biology, and Inflammation Laboratory, Institute of Biomedical Research (IIB-Sant Pau), Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Casós K, Siguero L, Fernández-Figueras MT, León X, Sardá MP, Vila L, Camacho M. Tumor cells induce COX-2 and mPGES-1 expression in microvascular endothelial cells mainly by means of IL-1 receptor activation. Microvasc Res 2011; 81:261-8. [PMID: 21277871 DOI: 10.1016/j.mvr.2011.01.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Revised: 01/18/2011] [Accepted: 01/18/2011] [Indexed: 01/13/2023]
Abstract
Prostaglandin (PG) E(2) plays a key role in immune response, tumor progression and metastasis. We previously showed that macrovessel-derived endothelial cells do not produce PGE(2) enzymatically because they do not express the inducible microsomal PGE-synthase-1 (mPGES-1). Nevertheless, differences between macro- and micro-vessel-derived endothelial cells regarding arachidonic acid (AAc) metabolism profile have been reported. The present work was conducted to evaluate the expression of PGE(2)-pathway-related enzymes in human microvascular endothelial cells (HMVEC) in culture and to test the hypothesis that the tumor cell-HMVEC cross talk could increase mPGES-1 expression in HMVEC. We treated HMVEC in culture with human recombinant IL-1β. IL-1β induced PGE(2) release and COX-2 and mPGES-1 expression in terms of mRNA and protein, determined by real-time PCR and immunoblotting, respectively. HMVEC constitutively expressed mPGES-2 and cytosolic PGES (cPGES) and the IL-1β treatment did not modify their expression. PGE(2) synthesized by HMVEC from exogenous AAc was linked to mPGES-1 expression. Immunohistochemistry analysis confirmed mPGES-1 expression in microvessels in vivo. COX-2 and mPGES-1 were also induced in HMVEC by the conditioned medium from two squamous head and neck carcinoma cell lines. Conditioned medium from tumor cell cultures contained several cytokines including the IL-1β and IL-1α. Tumor cell-induced COX-2 and mPGES-1 in HMVEC was strongly inhibited by the IL-1-receptor antagonist, indicating the important implication of IL-1 in this effect. HMVEC could therefore contribute directly to PGE(2) formed in the tumor. Our findings support the concept that mPGES-1 could be a target for therapeutic intervention in patients with cancer.
Collapse
Affiliation(s)
- Kelly Casós
- Laboratory of Angiology, Vascular Biology and Inflammation, Institute of Biomedical Research (II-B Sant Pau), Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
35
|
Díaz-Muñoz MD, Osma-García IC, Cacheiro-Llaguno C, Fresno M, Íñiguez MA. Coordinated up-regulation of cyclooxygenase-2 and microsomal prostaglandin E synthase 1 transcription by nuclear factor kappa B and early growth response-1 in macrophages. Cell Signal 2010; 22:1427-36. [DOI: 10.1016/j.cellsig.2010.05.011] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 05/15/2010] [Accepted: 05/15/2010] [Indexed: 01/19/2023]
|
36
|
Enhanced pressor response to acute Ang II infusion in mice lacking membrane-associated prostaglandin E2 synthase-1. Acta Pharmacol Sin 2010; 31:1284-92. [PMID: 20871624 DOI: 10.1038/aps.2010.99] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
AIM To examine the contribution of vascular membrane-associated prostaglandin E2 synthase-1 (mPGES-1) to acute blood pressure homeostasis. METHODS Angiotensin II (AngII, 75 pmol·kg⁻¹·min⁻¹) was continuously infused via the jugular vein into wild-type and mPGES-1(-/-) mice for 30 min, and blood pressure was measured by carotid arterial catheterization. RT-PCR and immunohistochemistry were performed to detect the expression and localization of mPGES-1 in the mouse arterial vessels. Mesenteric arteries were dissected from mice of both genotypes to study vessel tension and measure vascular PGE2 levels. RESULTS Wild-type and mPGES-1(-/-) mice showed similar blood pressure levels at baseline, and the acute intravenous infusion of AngII caused a greater increase in mean arterial pressure in the mPGES-1(-/-) group, with a similar diuretic and natriuretic response in both groups. mPGES-1 was constitutively expressed in the aortic and mesenteric arteries and vascular smooth muscle cells of wild-type mice. Strong staining was detected in the smooth muscle layer of arterial vessels. Ex vivo treatment of mesenteric arteries with AngII produced more vasodilatory PGE2 in wild-type than in mPGES-1(-/-) mice. In vitro tension assays further revealed that the mesenteric arteries of mPGES-1(-/-) mice exhibited a greater vasopressor response to AngII than those arteries of wild-type mice. CONCLUSION Vascular mPGES-1 acts as an important tonic vasodilator, contributing to acute blood pressure regulation.
Collapse
|
37
|
Vila L, Martinez-Perez A, Camacho M, Buil A, Alcolea S, Pujol-Moix N, Soler M, Antón R, Souto JC, Fontcuberta J, Soria JM. Heritability of thromboxane A2 and prostaglandin E2 biosynthetic machinery in a Spanish population. Arterioscler Thromb Vasc Biol 2009; 30:128-34. [PMID: 19850905 DOI: 10.1161/atvbaha.109.193219] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Prostanoids play a critical role in clinical areas such as inflammation, thrombosis, immune response, and cancer. Although some studies suggest that there are genes that determine variability of some prostanoid-related phenotypes, the genetic influence on these traits has not been evaluated. METHODS AND RESULTS The relative contributions of genetic and environmental influences to the prostanoid biosynthetic pathway-related phenotypes, cyclooxygenase isoenzymes, microsomal-PGE-synthase-1 and TxA-synthase expression, and thromboxane-A(2) and prostaglandin-E(2) production by stimulated whole blood, were assessed in a sample of 308 individuals in 15 extended families. The effects of measured covariates (such as sex, age, and smoking), genes, and environmental variables shared by members of a household were quantified. Heritabilities ranging from 0.406 to 0.634 for enzyme expression and from 0.283 to 0. 751 for prostanoid production were found. CONCLUSIONS These results demonstrate clearly the importance of genetic factors in determining variation in phenotypes that are components of the prostanoid biosynthetic pathways. The presence of such strong genetic effects suggest that it will be possible to localize previously unknown genes that influence quantitative variation in these phenotypes, some of which affect multiple aspects of cell biology, with important clinical implications.
Collapse
Affiliation(s)
- Luis Vila
- H.S. Creu i S. Pau, S. Antonio M Claret 167, 08025-Barcelona, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Iyer JP, Srivastava PK, Dev R, Dastidar SG, Ray A. Prostaglandin E(2) synthase inhibition as a therapeutic target. Expert Opin Ther Targets 2009; 13:849-65. [PMID: 19530988 DOI: 10.1517/14728220903018932] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Most NSAIDs function by inhibiting biosynthesis of PGE(2) by inhibition of COX-1 and/or COX-2. Since COX-1 has a protective function in the gastro-intestinal tract (GIT), non-selective inhibition of both cycloxy genases leads to moderate to severe gastro-intestinal intolerance. Attempts to identify selective inhibitors of COX-2, led to the identification of celecoxib and rofecoxib. However, long-term use of these drugs has serious adverse effects of sudden myocardial infarction and thrombosis. Drug-mediated imbalance in the levels of prostaglandin I(2) (PGI(2)) and thromboxane A(2) (TXA(2)) with a bias towards TXA(2) may be the primary reason for these events. This resulted in the drugs being withdrawn from the market, leaving a need for an effective and safe anti-inflammatory drug. METHODS Recently, the focus of research has shifted to enzymes downstream of COX in the prosta glandin biosynthetic pathway such as prostaglandin E(2) synthases. Microsomal prostaglandin E(2) synthase-1 (mPGES-1) specifically isomerizes PGH(2) to PGE(2), under inflammatory conditions. In this review, we examine the biology of mPGES-1 and its role in disease. Progress in designing molecules that can selectively inhibit mPGES-1 is reviewed. CONCLUSION mPGES-1 has the potential to be a target for anti-inflammatory therapy, devoid of adverse GIT and cardiac effects and warrants further investigation.
Collapse
Affiliation(s)
- Jitesh P Iyer
- Department of Pharmacology, New Drug Discovery Research, Ranbaxy Research Laboratories, Plot No-20, Sector-18, Udyog Vihar, Gurgaon, Haryana, India-122015
| | | | | | | | | |
Collapse
|
39
|
Increased Human Vascular Reactivity Via Cyclooxygenase-2 Inhibition During Acute Inflammation: Role of Prostaglandins E2 and I2. Inflamm Res 2009. [DOI: 10.1007/bf03354230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
40
|
Foudi N, Louedec L, Cachina T, Brink C, Norel X. Selective cyclooxygenase-2 inhibition directly increases human vascular reactivity to norepinephrine during acute inflammation. Cardiovasc Res 2008; 81:269-77. [DOI: 10.1093/cvr/cvn287] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
41
|
Camacho M, León X, Fernández-Figueras MT, Quer M, Vila L. Prostaglandin E2pathway in head and neck squamous cell carcinoma. Head Neck 2008; 30:1175-81. [DOI: 10.1002/hed.20850] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
|
42
|
Hsieh HL, Sun CC, Wang TS, Yang CM. PKC-δ/c-Src-mediated EGF receptor transactivation regulates thrombin-induced COX-2 expression and PGE2 production in rat vascular smooth muscle cells. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:1563-75. [DOI: 10.1016/j.bbamcr.2008.03.016] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2007] [Revised: 03/12/2008] [Accepted: 03/27/2008] [Indexed: 01/25/2023]
|
43
|
Camacho M, Rodríguez C, Salazar J, Martínez-González J, Ribalta J, Escudero JR, Masana L, Vila L. Retinoic acid induces PGI synthase expression in human endothelial cells. J Lipid Res 2008; 49:1707-14. [DOI: 10.1194/jlr.m700559-jlr200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
44
|
Foudi N, Kotelevets L, Louedec L, Leséche G, Henin D, Chastre E, Norel X. Vasorelaxation induced by prostaglandin E2 in human pulmonary vein: role of the EP4 receptor subtype. Br J Pharmacol 2008; 154:1631-9. [PMID: 18516068 DOI: 10.1038/bjp.2008.214] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND AND PURPOSE PGE2 has been shown to induce relaxations in precontracted human pulmonary venous preparations, while in pulmonary arteries this response was not observed. We investigated and characterized the prostanoid receptors which are activated by PGE2 in the human pulmonary veins. EXPERIMENTAL APPROACH Human pulmonary arteries and veins were cut as rings and set up in organ baths in presence of a TP antagonist. A pharmacological study was performed using selective EP1-4 ligands. The cellular localization of the EP4 receptors by immunohistochemistry and their corresponding transcripts were also investigated in these vessels. KEY RESULTS PGE2 and the EP4 agonists (L-902688, ONO-AE1-329) induced potent vasodilatation of the human pulmonary vein, pEC50 values: <7.22+/-0.20, 8.06+/-0.12 and 7.80+/-0.09, respectively. These relaxations were inhibited by the EP(4) antagonist GW627368X and not modified in presence of the DP antagonist L-877499. Higher concentrations (>or=1 microM) of the EP2 agonist ONO-AE1-259 induced relaxations of the veins. The EP4 agonists had no effect on the precontracted arteries. Finally, the EP(1) antagonists ONO-8713 and SC-51322 potentiated the relaxation of the veins induced by PGE2. EP4 and EP1 receptors were detected by immunohistochemistry in the veins but not in the arteries. EP4 mRNA accumulation was also greater in the veins when compared with the arterial preparations. CONCLUSIONS AND IMPLICATIONS Of the 4 EP receptor subtypes, smooth muscle cells in the human pulmonary vein express the EP4 and EP1 receptor subtypes. The relaxations induced by PGE2 in this vessel result from the activation of the EP4 receptor.
Collapse
|
45
|
Hamza A, AbdulHameed MDM, Zhan CG. Understanding Microscopic Binding of Human Microsomal Prostaglandin E Synthase-1 with Substrates and Inhibitors by Molecular Modeling and Dynamics Simulation. J Phys Chem B 2008; 112:7320-9. [DOI: 10.1021/jp8007688] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Adel Hamza
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, Kentucky 40536
| | - Mohamed Diwan M. AbdulHameed
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, Kentucky 40536
| | - Chang-Guo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, Kentucky 40536
| |
Collapse
|
46
|
Hétu PO, Ouellet M, Falgueyret JP, Ramachandran C, Robichaud J, Zamboni R, Riendeau D. Photo-crosslinking of proteins in intact cells reveals a dimeric structure of cyclooxygenase-2 and an inhibitor-sensitive oligomeric structure of microsomal prostaglandin E2 synthase-1. Arch Biochem Biophys 2008; 477:155-62. [PMID: 18498757 DOI: 10.1016/j.abb.2008.04.038] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2008] [Revised: 04/25/2008] [Accepted: 04/26/2008] [Indexed: 10/22/2022]
Abstract
We have characterized the structures of cyclooxygenase-2 (COX-2) and microsomal prostaglandin E(2) synthase-1 (mPGES-1) in intact cells using bifunctional and photo-activatable crosslinking agents. A dimeric complex was detected for COX-2 by both crosslinking approaches, consistent with the crystal structure of the enzyme. For mPGES-1, treatment of A549 cells with disuccinimidyl suberate yielded immunoreactive protein bands corresponding to a dimer (33 kDa) and a trimer (45 kDa), as observed for the isolated enzyme. Photo-crosslinking with photoactivatable methionine in intact cells generated complexes with molecular weights corresponding to the dimer (33 kDa) and two putative trimer forms (50 and 55 kDa). Treatment with the selective mPGES-1 inhibitor MF63 prevented the formation of the 50 and 55 kDa crosslinked complexes, while an inactive structural analogue had no effect. Our data indicate that COX-2 forms a dimer in intact cells and that mPGES-1 has an oligomeric structure that can be disrupted by a selective inhibitor.
Collapse
Affiliation(s)
- Pierre-Olivier Hétu
- Department of Biochemistry & Molecular Biology, Merck Frosst Centre for Therapeutic Research, Kirkland, Que., Canada
| | | | | | | | | | | | | |
Collapse
|
47
|
Wang M, Song WL, Cheng Y, Fitzgerald GA. Microsomal prostaglandin E synthase-1 inhibition in cardiovascular inflammatory disease. J Intern Med 2008; 263:500-5. [PMID: 18410593 DOI: 10.1111/j.1365-2796.2008.01938.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Prostaglandins (PGs), particularly PGE2 and prostacyclin (PGI2), are potent mediators of pain and inflammation. Both atherosclerosis and aortic aneurysm exhibit the hallmarks of inflammation. However, randomized trials of inhibitors of PG synthesis--nonsteroidal anti-inflammatory drugs--reveal that they predispose to cardiovascular risk. This appears to be consequent to inhibition of PGI2 and PGE2 formed by cyclooxygenase-2 (COX-2). Inhibitors of microsomal PGE synthase-1 (mPGES-1) are being developed for relief of pain and interest has focused on their potential impact on the cardiovascular system. Deletion of mPGES-1 retards atherogenesis and limits aortic aneurysm formation in hyperlipidaemic mice. However, it does not predispose to thrombogenesis and has a limited impact on blood pressure compared to inhibition of COX-2. This occurs despite the potential of the suppressed PGE2 in affording cardioprotection via its EP2 and EP4 receptors. However, deletion of mPGES-1 permits rediversion of the PGH2 substrate to other PG synthases and augmented formation of PGI2 and PGD2 mitigates this effect. However, increased PGI2 may also attenuate relief of pain. Pain relief seems likely to be a nuanced indication for mPGES-1 inhibitors, but they have therapeutic potential in syndromes of cardiovascular inflammation, cancer and perhaps in neurodegenerative disease. However, as the products of substrate rediversion vary according to cell type, these drugs may have contrasting impact amongst individuals at varied stages of disease evolution.
Collapse
Affiliation(s)
- M Wang
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | |
Collapse
|
48
|
Lee IY, Cho W, Kim J, Park CS, Choe J. Human Follicular Dendritic Cells Interact with T Cells via Expression and Regulation of Cyclooxygenases and Prostaglandin E and I Synthases. THE JOURNAL OF IMMUNOLOGY 2008; 180:1390-7. [DOI: 10.4049/jimmunol.180.3.1390] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
49
|
AbdulHameed MDM, Hamza A, Liu J, Huang X, Zhan CG. Human microsomal prostaglandin E synthase-1 (mPGES-1) binding with inhibitors and the quantitative structure-activity correlation. J Chem Inf Model 2007; 48:179-85. [PMID: 18052142 DOI: 10.1021/ci700315c] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The detailed structures of microsomal prostaglandin E synthase-1 (mPGES-1) binding with inhibitors have been studied, for the first time, by using a newly developed computational three-dimensional (3D) structural model of mPGES-1 along with a 3D-quantitative structure--activity relationship (3D-QSAR) analysis. The obtained satisfactory binding structures and 3D-QSAR models strongly suggest that the 3D structural model of mPGES-1 is reasonable for study of mPGES-1 binding with inhibitors and for future design of novel mPGES-1 inhibitors.
Collapse
Affiliation(s)
- Mohamed Diwan M AbdulHameed
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, 725 Rose Street, Lexington, Kentucky 40536, USA
| | | | | | | | | |
Collapse
|
50
|
Krötz F. Preserving vascular prostacyclin levels by microsomal prostaglandin E(2) synthase isoform 1 inhibition: a new strategy for vasoprotection? J Thromb Haemost 2007; 5:1409-10. [PMID: 17488348 DOI: 10.1111/j.1538-7836.2007.02614.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- F Krötz
- Cardiology Division, Medizinische Poliklinik, Ludwig-Maximilians-Universität, München, Germany.
| |
Collapse
|