1
|
Tourkochristou E, Mouzaki A, Triantos C. Unveiling the biological role of sphingosine-1-phosphate receptor modulators in inflammatory bowel diseases. World J Gastroenterol 2023; 29:110-125. [PMID: 36683721 PMCID: PMC9850947 DOI: 10.3748/wjg.v29.i1.110] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/16/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is chronic inflammation of the gastrointestinal tract that has a high epidemiological prevalence worldwide. The increasing disease burden worldwide, lack of response to current biologic therapeutics, and treatment-related immunogenicity have led to major concerns regarding the clinical management of IBD patients and treatment efficacy. Understanding disease pathogenesis and disease-related molecular mechanisms is the most important goal in developing new and effective therapeutics. Sphingosine-1-phosphate (S1P) receptor (S1PR) modulators form a class of oral small molecule drugs currently in clinical development for IBD have shown promising effects on disease improvement. S1P is a sphingosine-derived phospholipid that acts by binding to its receptor S1PR and is involved in the regulation of several biological processes including cell survival, differentiation, migration, proliferation, immune response, and lymphocyte trafficking. T lymphocytes play an important role in regulating inflammatory responses. In inflamed IBD tissue, an imbalance between T helper (Th) and regulatory T lymphocytes and Th cytokine levels was found. The S1P/S1PR signaling axis and metabolism have been linked to inflammatory responses in IBD. S1P modulators targeting S1PRs and S1P metabolism have been developed and shown to regulate inflammatory responses by affecting lymphocyte trafficking, lymphocyte number, lymphocyte activity, cytokine production, and contributing to gut barrier function.
Collapse
Affiliation(s)
- Evanthia Tourkochristou
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | - Athanasia Mouzaki
- Division of Hematology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, Medical School, University of Patras, Patras 26504, Greece
| |
Collapse
|
2
|
The Role of Obesity, Inflammation and Sphingolipids in the Development of an Abdominal Aortic Aneurysm. Nutrients 2022; 14:nu14122438. [PMID: 35745168 PMCID: PMC9229568 DOI: 10.3390/nu14122438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 06/03/2022] [Accepted: 06/11/2022] [Indexed: 02/06/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is a local dilatation of the vessel equal to or exceeding 3 cm. It is a disease with a long preclinical period commonly without any symptoms in its initial stage. Undiagnosed for years, aneurysm often leads to death due to vessel rupture. The basis of AAA pathogenesis is inflammation, which is often associated with the excess of adipose tissue, especially perivascular adipose tissue, which synthesizes adipocytokines that exert a significant influence on the formation of aneurysms. Pro-inflammatory cytokines such as resistin, leptin, and TNFα have been shown to induce changes leading to the formation of aneurysms, while adiponectin is the only known compound that is secreted by adipose tissue and limits the development of aneurysms. However, in obesity, adiponectin levels decline. Moreover, inflammation is associated with an increase in the amount of macrophages infiltrating adipose tissue, which are the source of matrix metalloproteinases (MMP) involved in the degradation of the extracellular matrix, which are an important factor in the formation of aneurysms. In addition, an excess of body fat is associated with altered sphingolipid metabolism. It has been shown that among sphingolipids, there are compounds that play an opposite role in the cell: ceramide is a pro-apoptotic compound that mediates the development of inflammation, while sphingosine-1-phosphate exerts pro-proliferative and anti-inflammatory effects. It has been shown that the increase in the level of ceramide is associated with a decrease in the concentration of adiponectin, an increase in the concentration of TNFα, MMP-9 and reactive oxygen species (which contribute to the apoptosis of vascular smooth muscle cell). The available data indicate a potential relationship between obesity, inflammation and disturbed sphingolipid metabolism with the formation of aneurysms; therefore, the aim of this study was to systematize the current knowledge on the role of these factors in the pathogenesis of abdominal aortic aneurysm.
Collapse
|
3
|
Crosstalk between ORMDL3, serine palmitoyltransferase, and 5-lipoxygenase in the sphingolipid and eicosanoid metabolic pathways. J Lipid Res 2021; 62:100121. [PMID: 34560079 PMCID: PMC8527048 DOI: 10.1016/j.jlr.2021.100121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 08/23/2021] [Accepted: 09/08/2021] [Indexed: 11/21/2022] Open
Abstract
Leukotrienes (LTs) and sphingolipids are critical lipid mediators participating in numerous cellular signal transduction events and developing various disorders, such as bronchial hyperactivity leading to asthma. Enzymatic reactions initiating production of these lipid mediators involve 5-lipoxygenase (5-LO)-mediated conversion of arachidonic acid to LTs and serine palmitoyltransferase (SPT)-mediated de novo synthesis of sphingolipids. Previous studies have shown that endoplasmic reticulum membrane protein ORM1-like protein 3 (ORMDL3) inhibits the activity of SPT and subsequent sphingolipid synthesis. However, the role of ORMDL3 in the synthesis of LTs is not known. In this study, we used peritoneal-derived mast cells isolated from ORMDL3 KO or control mice and examined their calcium mobilization, degranulation, NF-κB inhibitor-α phosphorylation, and TNF-α production. We found that peritoneal-derived mast cells with ORMDL3 KO exhibited increased responsiveness to antigen. Detailed lipid analysis showed that compared with WT cells, ORMDL3-deficient cells exhibited not only enhanced production of sphingolipids but also of LT signaling mediators LTB4, 6t-LTB4, LTC4, LTB5, and 6t-LTB5. The crosstalk between ORMDL3 and 5-LO metabolic pathways was supported by the finding that endogenous ORMDL3 and 5-LO are localized in similar endoplasmic reticulum domains in human mast cells and that ORMDL3 physically interacts with 5-LO. Further experiments showed that 5-LO also interacts with the long-chain 1 and long-chain 2 subunits of SPT. In agreement with these findings, 5-LO knockdown increased ceramide levels, and silencing of SPTLC1 decreased arachidonic acid metabolism to LTs to levels observed upon 5-LO knockdown. These results demonstrate functional crosstalk between the LT and sphingolipid metabolic pathways, leading to the production of lipid signaling mediators.
Collapse
|
4
|
Machida T, Endo TH, Oyoshi R, Yutani M, Machida M, Shiga S, Murakami H, Hiraide S, Hirafuji M, Iizuka K. Abnormal Pressure Stress Reduces Interleukin-1β-Induced Cyclooxygenase-2 Expression in Cultured Rat Vascular Smooth Muscle Cells. Biol Pharm Bull 2021; 44:853-860. [PMID: 34078818 DOI: 10.1248/bpb.b21-00078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Elevated mechanical stress on blood vessels associated with hypertension has a direct effect on the function of vascular endothelial cells and vascular smooth muscle cells (VSMCs). In the present study, we have identified the effect of pulsatile pressure stress on cyclooxygenase-2 (COX-2) expression induced by interleukin (IL)-1β in cultured rat VSMCs. VSMCs were isolated from aortic media of Wistar rats and cultured. Pulsatile pressure applied to VSMCs was repeatedly given between either 80 and 160 mmHg, which simulates systolic hypertension, or 80 and 120 mmHg, which simulates normal blood pressure, at a frequency of 4 cycles per min using our original apparatus. Pressure loading that simulates systolic hypertension reduced IL-1β-induced COX-2 expression. The pressure also inhibited the rapid and transient phosphorylation of extracellular signal-regulated kinase (ERK) induced by IL-1β. IL-1β-induced COX-2 expression was significantly inhibited by a specific conventional protein kinase C (PKC) inhibitor. Pressure loading that simulates systolic hypertension also reduced phorbol myristate 13-acetate (PMA) (a PKC activator)-induced COX-2 expression and the rapid and transient phosphorylation of ERK. Pressure loading that simulates normal blood pressure had no effect on IL-1β- and PMA-induced COX-2 expression. The present study shows that pressure stress between 80 and 160 mmHg, which simulates systolic hypertension reduces IL-1β-induced COX-2 expression by affecting a mechanism involving PKC and ERK signaling pathways. Downregulation of COX-2 expression in VSMCs by abnormal pressure stress may further worsen local vascular injury associated with hypertension.
Collapse
Affiliation(s)
- Takuji Machida
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Tomoko Hinse Endo
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Riho Oyoshi
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Mikiko Yutani
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Maiko Machida
- Division of Pharmacotherapy, Faculty of Pharmaceutical Sciences, Hokkaido University of Science
| | - Saki Shiga
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Hina Murakami
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Sachiko Hiraide
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Masahiko Hirafuji
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| | - Kenji Iizuka
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido
| |
Collapse
|
5
|
Yang CC, Hsiao LD, Su MH, Yang CM. Sphingosine 1-Phosphate Induces Cyclooxygenase-2/Prostaglandin E 2 Expression via PKCα-dependent Mitogen-Activated Protein Kinases and NF-κB Cascade in Human Cardiac Fibroblasts. Front Pharmacol 2020; 11:569802. [PMID: 33192511 PMCID: PMC7662885 DOI: 10.3389/fphar.2020.569802] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 09/22/2020] [Indexed: 12/12/2022] Open
Abstract
In the regions of tissue injuries and inflammatory diseases, sphingosine 1-phosphate (S1P), a proinflammatory mediator, is increased. S1P may induce the upregulation of cyclooxygenase-2 (COX-2)/prostaglandin E2 (PGE2) system in various types of cells to exacerbate heart inflammation. However, the detailed molecular mechanisms by which S1P induces COX-2 expression in human cardiac fibroblasts (HCFs) remain unknown. HCFs were incubated with S1P and analyzed by Western blotting, real time-Polymerase chain reaction (RT-PCR), and immunofluorescent staining. Our results indicated that S1P activated S1PR1/3-dependent transcriptional activity to induce COX-2 expression and PGE2 production. S1P recruited and activated PTX-sensitive Gi or -insensitive Gq protein-coupled S1PR and then stimulated PKCα-dependent phosphorylation of p42/p44 MAPK, p38 MAPK, and JNK1/2, leading to activating transcription factor NF-κB. Moreover, S1P-activated NF-κB was translocated into the nucleus and bound to its corresponding binding sites on COX-2 promoters determined by chromatin immunoprecipitation (ChIP) and promoter-reporter assays, thereby turning on COX-2 gene transcription associated with PGE2 production in HCFs. These results concluded that in HCFs, activation of NF-κB by PKCα-mediated MAPK cascades was essential for S1P-induced up-regulation of the COX-2/PGE2 system. Understanding the mechanisms of COX-2 expression and PGE2 production regulated by the S1P/S1PRs system on cardiac fibroblasts may provide rationally therapeutic interventions for heart injury or inflammatory diseases.
Collapse
Affiliation(s)
- Chien-Chung Yang
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital at Tao-Yuan, Tao-Yuan, Taiwan.,School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Mei-Hsiu Su
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan
| | - Chuen-Mao Yang
- Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan.,Department of Post-Baccalaureate Veterinary Medicine, College of Medical and Health Science, Asia University, Wufeng, Taichung, Taiwan
| |
Collapse
|
6
|
Sukocheva OA, Furuya H, Ng ML, Friedemann M, Menschikowski M, Tarasov VV, Chubarev VN, Klochkov SG, Neganova ME, Mangoni AA, Aliev G, Bishayee A. Sphingosine kinase and sphingosine-1-phosphate receptor signaling pathway in inflammatory gastrointestinal disease and cancers: A novel therapeutic target. Pharmacol Ther 2020; 207:107464. [PMID: 31863815 DOI: 10.1016/j.pharmthera.2019.107464] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 12/10/2019] [Indexed: 02/07/2023]
Abstract
Inflammatory gastrointestinal (GI) diseases and malignancies are associated with growing morbidity and cancer-related mortality worldwide. GI tumor and inflammatory cells contain activated sphingolipid-metabolizing enzymes, including sphingosine kinase 1 (SphK1) and SphK2, that generate sphingosine-1-phosphate (S1P), a highly bioactive compound. Many inflammatory responses, including lymphocyte trafficking, are directed by circulatory S1P, present in high concentrations in both the plasma and the lymph of cancer patients. High fat and sugar diet, disbalanced intestinal flora, and obesity have recently been linked to activation of inflammation and SphK/S1P/S1P receptor (S1PR) signaling in various GI pathologies, including cancer. SphK1 overexpression and activation facilitate and enhance the development and progression of esophageal, gastric, and colon cancers. SphK/S1P axis, a mediator of inflammation in the tumor microenvironment, has recently been defined as a target for the treatment of GI disease states, including inflammatory bowel disease and colitis. Several SphK1 inhibitors and S1PR antagonists have been developed as novel anti-inflammatory and anticancer agents. In this review, we analyze the mechanisms of SphK/S1P signaling in GI tissues and critically appraise recent studies on the role of SphK/S1P/S1PR in inflammatory GI disorders and cancers. The potential role of SphK/S1PR inhibitors in the prevention and treatment of inflammation-mediated GI diseases, including GI cancer, is also evaluated.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, South Australia 5042, Australia
| | - Hideki Furuya
- Department of Surgery, Samuel Oschin Cancer Center Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Mei Li Ng
- Advanced Medical and Dental Institute, University Sains 13200 Kepala Batas, Pulau Pinang, Malaysia
| | - Markus Friedemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Vadim V Tarasov
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - Vladimir N Chubarev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia
| | - Sergey G Klochkov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Margarita E Neganova
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia
| | - Arduino A Mangoni
- Discipline of Clinical Pharmacology, College of Medicine and Public Health, Flinders University and Flinders Medical Centre, Bedford Park, South Australia 5042, Australia
| | - Gjumrakch Aliev
- Sechenov First Moscow State Medical University (Sechenov University), Moscow 119991, Russia; Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka 142432, Russia; GALLY International Research Institute, San Antonio, TX 78229, USA; Research Institute of Human Morphology, Moscow 117418, Russia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL 34211, USA.
| |
Collapse
|
7
|
Sukocheva OA, Lukina E, McGowan E, Bishayee A. Sphingolipids as mediators of inflammation and novel therapeutic target in inflammatory bowel disease. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2020; 120:123-158. [PMID: 32085881 DOI: 10.1016/bs.apcsb.2019.11.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Morbidity of inflammatory gastrointestinal (GI) diseases continues to grow resulting in worsen quality of life and increased burden on public medical systems. Complex and heterogenous illnesses, inflammatory bowel diseases (IBDs) encompass several inflammation -associated pathologies including Crohn's disease and ulcerative colitis. IBD is often initiated by a complex interplay between host genetic and environmental factors, lifestyle and diet, and intestinal bacterial components. IBD inflammatory signature was linked to the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) signaling pathway that is currently targeted by IBD therapies. Sphingolipid signaling was identified as one of the key mediators and regulators of pro-inflammatory conditions, and, specifically, TNF-α related signaling. All GI tissues and circulating immune/blood cells contain activated sphingolipid-metabolizing enzymes, including sphingosine kinases (SphK1 and SphK2) that generate sphingosine-1-phosphate (S1P), a bioactive lipid and ligand for five G-protein coupled membrane S1P receptors (S1PRs). Numerous normal and pathogenic inflammatory responses are mediated by SphK/S1P/S1PRs signaling axis including lymphocyte trafficking and activation of cytokine signaling machinery. SphK1/S1P/S1PRs axis has recently been defined as a target for the treatment of GI diseases including IBD/colitis. Several SphK1 inhibitors and S1PRs antagonists have been developed as novel anti-inflammatory agents. In this review, we discuss the mechanisms of SphK/S1P signaling in inflammation-linked GI disorders. The potential role of SphK/S1PRs inhibitors in the prevention and treatment of IBD/colitis is critically evaluated.
Collapse
Affiliation(s)
- Olga A Sukocheva
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Elena Lukina
- Discipline of Health Sciences, College of Nursing and Health Sciences, Flinders University, Bedford Park, SA, Australia
| | - Eileen McGowan
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Anupam Bishayee
- Lake Erie College of Osteopathic Medicine, Bradenton, FL, United States
| |
Collapse
|
8
|
Yu X, Bao Y, Meng X, Wang S, Li T, Chang X, Xu W, Yang G, Bo T. Multi-pathway integrated adjustment mechanism of licorice flavonoids presenting anti-inflammatory activity. Oncol Lett 2019; 18:4956-4963. [PMID: 31612007 DOI: 10.3892/ol.2019.10793] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 11/08/2017] [Indexed: 12/29/2022] Open
Abstract
Glycyrrhiza, commonly known as licorice, is a herbal medicine that has been used for thousands of years. Licorice contains multiple flavonoids, which possess a variety of biological activities. On the basis of the anti-inflammatory effects of licorice flavonoids, the potential mechanism of action was investigated via a plasma metabolomics approach. A total of 9 differential endogenous metabolites associated with the therapeutic effect of licorice flavonoids were identified, including linoleic acid, sphingosine, tryptophanamide, corticosterone and leukotriene B4. Besides classical arachidonic acid metabolism, metabolism of sphingolipids, tryptophan and fatty acids, phospholipids synthesis, and other pathways were also involved. The multi-pathway integrated adjustment mechanism of licorice flavonoid action may reduce side effects in patients, along with any anti-inflammatory functions, which provides a foundation for identifying and developing novel, high-potential natural drugs with fewer side effects for clinical application.
Collapse
Affiliation(s)
- Xiaomeng Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Yongrui Bao
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, Liaoning 116600, P.R. China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, Liaoning 116600, P.R. China.,Liaoning University of Traditional Chinese Medicine-Agilent Technologies Modern TCM and Multi-omics Research Collaboration Laboratory, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Xiansheng Meng
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, Liaoning 116600, P.R. China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, Liaoning 116600, P.R. China.,Liaoning University of Traditional Chinese Medicine-Agilent Technologies Modern TCM and Multi-omics Research Collaboration Laboratory, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Shuai Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, Liaoning 116600, P.R. China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, Liaoning 116600, P.R. China.,Liaoning University of Traditional Chinese Medicine-Agilent Technologies Modern TCM and Multi-omics Research Collaboration Laboratory, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Tianjiao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China.,Component Medicine Engineering Research Center of Liaoning Province, Dalian, Liaoning 116600, P.R. China.,Liaoning Province Modern Chinese Medicine Research Engineering Laboratory, Dalian, Liaoning 116600, P.R. China.,Liaoning University of Traditional Chinese Medicine-Agilent Technologies Modern TCM and Multi-omics Research Collaboration Laboratory, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Xin Chang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Weifeng Xu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Guanlin Yang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| | - Tao Bo
- Liaoning University of Traditional Chinese Medicine-Agilent Technologies Modern TCM and Multi-omics Research Collaboration Laboratory, Liaoning University of Traditional Chinese Medicine, Dalian, Liaoning 116600, P.R. China
| |
Collapse
|
9
|
Rumzhum NN, Ammit AJ. Cyclooxygenase 2: its regulation, role and impact in airway inflammation. Clin Exp Allergy 2016; 46:397-410. [PMID: 26685098 DOI: 10.1111/cea.12697] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Cyclooxygenase 2 (COX-2: official gene symbol - PTGS2) has long been regarded as playing a pivotal role in the pathogenesis of airway inflammation in respiratory diseases including asthma. COX-2 can be rapidly and robustly expressed in response to a diverse range of pro-inflammatory cytokines and mediators. Thus, increased levels of COX-2 protein and prostanoid metabolites serve as key contributors to pathobiology in respiratory diseases typified by dysregulated inflammation. But COX-2 products may not be all bad: prostanoids can exert anti-inflammatory/bronchoprotective functions in airways in addition to their pro-inflammatory actions. Herein, we outline COX-2 regulation and review the diverse stimuli known to induce COX-2 in the context of airway inflammation. We discuss some of the positive and negative effects that COX-2/prostanoids can exert in in vitro and in vivo models of airway inflammation, and suggest that inhibiting COX-2 expression to repress airway inflammation may be too blunt an approach; because although it might reduce the unwanted effects of COX-2 activation, it may also negate the positive effects. Evidence suggests that prostanoids produced via COX-2 upregulation show diverse actions (and herein we focus on prostaglandin E2 as a key example); these can be either beneficial or deleterious and their impact on respiratory disease can be dictated by local concentration and specific interaction with individual receptors. We propose that understanding the regulation of COX-2 expression and associated receptor-mediated functional outcomes may reveal number of critical steps amenable to pharmacological intervention. These may prove invaluable in our quest towards future development of novel anti-inflammatory pharmacotherapeutic strategies for the treatment of airway diseases.
Collapse
Affiliation(s)
- N N Rumzhum
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| | - A J Ammit
- Faculty of Pharmacy, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
10
|
Rahman MM, Prünte L, Lebender LF, Patel BS, Gelissen I, Hansbro PM, Morris JC, Clark AR, Verrills NM, Ammit AJ. The phosphorylated form of FTY720 activates PP2A, represses inflammation and is devoid of S1P agonism in A549 lung epithelial cells. Sci Rep 2016; 6:37297. [PMID: 27849062 PMCID: PMC5110966 DOI: 10.1038/srep37297] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/27/2016] [Indexed: 12/21/2022] Open
Abstract
Protein phosphatase 2A (PP2A) activity can be enhanced pharmacologically by PP2A-activating drugs (PADs). The sphingosine analog FTY720 is the best known PAD and we have shown that FTY720 represses production of pro-inflammatory cytokines responsible for respiratory disease pathogenesis. Whether its phosphorylated form, FTY720-P, also enhances PP2A activity independently of the sphingosine 1-phosphate (S1P) pathway was unknown. Herein, we show that FTY720-P enhances TNF-induced PP2A phosphatase activity and significantly represses TNF-induced interleukin 6 (IL-6) and IL-8 mRNA expression and protein secretion from A549 lung epithelial cells. Comparing FTY720 and FTY720-P with S1P, we show that unlike S1P, the sphingosine analogs do not induce cytokine production on their own. In fact, FTY720 and FTY720-P significantly repress S1P-induced IL-6 and IL-8 production. We then examined their impact on expression of cyclooxygenase 2 (COX-2) and resultant prostaglandin E2 (PGE2) production. S1P did not increase production of this pro-inflammatory enzyme because COX-2 mRNA gene expression is NF-κB-dependent, and unlike TNF, S1P did not activate NF-κB. However, TNF-induced COX-2 mRNA expression and PGE2 secretion is repressed by FTY720 and FTY720-P. Hence, FTY720-P enhances PP2A activity and that PADs can repress production of pro-inflammatory cytokines and enzymes in A549 lung epithelial cells in a manner devoid of S1P agonism.
Collapse
Affiliation(s)
| | - Laura Prünte
- Faculty of Pharmacy, University of Sydney, NSW, 2006, Australia
| | | | | | - Ingrid Gelissen
- Faculty of Pharmacy, University of Sydney, NSW, 2006, Australia
| | - Philip M. Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and the University of Newcastle, NSW, 2308, Australia
| | | | - Andrew R. Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston B15 2TT, United Kingdom
| | - Nicole M. Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, NSW, 2308, Australia
| | - Alaina J. Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, NSW, Australia
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia
| |
Collapse
|
11
|
Anguita E, Villalobo A. Src-family tyrosine kinases and the Ca 2+ signal. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1864:915-932. [PMID: 27818271 DOI: 10.1016/j.bbamcr.2016.10.022] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 10/25/2016] [Accepted: 10/30/2016] [Indexed: 01/08/2023]
Abstract
In this review, we shall describe the rich crosstalk between non-receptor Src-family kinases (SFKs) and the Ca2+ transient generated in activated cells by a variety of extracellular and intracellular stimuli, resulting in diverse signaling events. The exchange of information between SFKs and Ca2+ is reciprocal, as it flows in both directions. These kinases are main actors in pathways leading to the generation of the Ca2+ signal, and reciprocally, the Ca2+ signal modulates SFKs activity and functions. We will cover how SFKs participate in the generation of the cytosolic Ca2+ rise upon activation of a series of receptors and the mechanism of clearance of this Ca2+ signal. The role of SFKs modulating Ca2+-translocating channels participating in these events will be amply discussed. Finally, the role of the Ca2+ sensor protein calmodulin on the activity of c-Src, and potentially on other SFKs, will be outlined as well. This article is part of a Special Issue entitled: ECS Meeting edited by Claus Heizmann, Joachim Krebs and Jacques Haiech.
Collapse
Affiliation(s)
- Estefanía Anguita
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain
| | - Antonio Villalobo
- Department of Cancer Biology, Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, c/ Arturo Duperier 4, E-28029 Madrid, Spain.
| |
Collapse
|
12
|
Filipenko I, Schwalm S, Reali L, Pfeilschifter J, Fabbro D, Huwiler A, Zangemeister-Wittke U. Upregulation of the S1P 3 receptor in metastatic breast cancer cells increases migration and invasion by induction of PGE 2 and EP 2/EP 4 activation. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:1840-1851. [PMID: 27616330 DOI: 10.1016/j.bbalip.2016.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 09/02/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
Breast cancer is one of the most common and devastating malignancies among women worldwide. Recent evidence suggests that malignant progression is also driven by processes involving the sphingolipid molecule sphingosine 1-phosphate (S1P) and its binding to cognate receptor subtypes on the cell surface. To investigate the effect of this interaction on the metastatic phenotype, we used the breast cancer cell line MDA-MB-231 and the sublines 4175 and 1833 derived from lung and bone metastases in nude mice, respectively. In both metastatic cell lines expression of the S1P3 receptor was strongly upregulated compared to the parental cells and correlated with higher S1P-induced intracellular calcium ([Ca2+]i), higher cyclooxygenase (COX)-2 and microsomal prostaglandin (PG) E2 synthase expression, and consequently with increased PGE2 synthesis. PGE2 synthesis was decreased by antagonists and siRNA against S1P3 and S1P2. Moreover, in parental MDA-MB-231 cells overexpression of S1P3 by cDNA transfection also increased PGE2 synthesis, but only after treatment with the DNA methyltransferase inhibitor 5-aza-2-deoxycytidine, indicating reversible silencing of the COX-2 promoter. Functionally, the metastatic sublines showed enhanced migration and Matrigel invasion in adapted Boyden chamber assays, which further increased by S1P stimulation. This response was abrogated by either S1P3 antagonism, COX-2 inhibition or PGE2 receptor 2 (EP2) and 4 (EP4) antagonism, but not by S1P2 antagonism. Our data demonstrate that in breast cancer cells overexpression of S1P3 and its activation by S1P has pro-inflammatory and pro-metastatic potential by inducing COX-2 expression and PGE2 signaling via EP2 and EP4.
Collapse
Affiliation(s)
- Iuliia Filipenko
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe Universität Frankfurt am Main, Theodor Stern Kai 7, D-60590 Frankfurt am Main, Germany
| | - Luca Reali
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe Universität Frankfurt am Main, Theodor Stern Kai 7, D-60590 Frankfurt am Main, Germany
| | - Doriano Fabbro
- PIQUR Therapeutics AG, Hochbergstrasse 60C, CH-4057 Basel, Switzerland
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland.
| | - Uwe Zangemeister-Wittke
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3011 Bern, Switzerland.
| |
Collapse
|
13
|
Machida T, Matamura R, Iizuka K, Hirafuji M. Cellular function and signaling pathways of vascular smooth muscle cells modulated by sphingosine 1-phosphate. J Pharmacol Sci 2016; 132:211-217. [PMID: 27581589 DOI: 10.1016/j.jphs.2016.05.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/25/2016] [Accepted: 05/26/2016] [Indexed: 01/21/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) plays important roles in cardiovascular pathophysiology. S1P1 and/or S1P3, rather than S1P2 receptors, seem to be predominantly expressed in vascular endothelial cells, while S1P2 and/or S1P3, rather than S1P1 receptors, seem to be predominantly expressed in vascular smooth muscle cells (VSMCs). S1P has multiple actions, such as proliferation, inhibition or stimulation of migration, and vasoconstriction or release of vasoactive mediators. S1P induces an increase of the intracellular Ca2+ concentration in many cell types, including VSMCs. Activation of S1P3 seems to play an important role in Ca2+ mobilization. S1P induces cyclooxygenase-2 expression in VSMCs via both S1P2 and S1P3 receptors. S1P2 receptor activation in VSMCs inhibits inducible nitric oxide synthase (iNOS) expression. At the local site of vascular injury, vasoactive mediators such as prostaglandins and NO produced by VSMCs are considered primarily as a defensive and compensatory mechanism for the lack of endothelial function to prevent further pathology. Therefore, selective S1P2 receptor antagonists may have the potential to be therapeutic agents, in view of their antagonism of iNOS inhibition by S1P. Further progress in studies of the precise mechanisms of S1P may provide useful knowledge for the development of new S1P-related drugs for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Takuji Machida
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan.
| | - Ryosuke Matamura
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Kenji Iizuka
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | - Masahiko Hirafuji
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| |
Collapse
|
14
|
Cheng JC, Chang HM, Liu PP, Leung PC. Sphingosine-1-phosphate induces COX-2 expression and PGE2 production in human granulosa cells through a S1P1/3-mediated YAP signaling. Cell Signal 2016; 28:643-51. [DOI: 10.1016/j.cellsig.2016.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 03/15/2016] [Accepted: 03/15/2016] [Indexed: 01/07/2023]
|
15
|
Mahajan-Thakur S, Böhm A, Jedlitschky G, Schrör K, Rauch BH. Sphingosine-1-Phosphate and Its Receptors: A Mutual Link between Blood Coagulation and Inflammation. Mediators Inflamm 2015; 2015:831059. [PMID: 26604433 PMCID: PMC4641948 DOI: 10.1155/2015/831059] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/26/2015] [Accepted: 09/30/2015] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a versatile lipid signaling molecule and key regulator in vascular inflammation. S1P is secreted by platelets, monocytes, and vascular endothelial and smooth muscle cells. It binds specifically to a family of G-protein-coupled receptors, S1P receptors 1 to 5, resulting in downstream signaling and numerous cellular effects. S1P modulates cell proliferation and migration, and mediates proinflammatory responses and apoptosis. In the vascular barrier, S1P regulates permeability and endothelial reactions and recruitment of monocytes and may modulate atherosclerosis. Only recently has S1P emerged as a critical mediator which directly links the coagulation factor system to vascular inflammation. The multifunctional proteases thrombin and FXa regulate local S1P availability and interact with S1P signaling at multiple levels in various vascular cell types. Differential expression patterns and intracellular signaling pathways of each receptor enable S1P to exert its widespread functions. Although a vast amount of information is available about the functions of S1P and its receptors in the regulation of physiological and pathophysiological conditions, S1P-mediated mechanisms in the vasculature remain to be elucidated. This review summarizes recent findings regarding the role of S1P and its receptors in vascular wall and blood cells, which link the coagulation system to inflammatory responses in the vasculature.
Collapse
Affiliation(s)
- Shailaja Mahajan-Thakur
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Andreas Böhm
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Gabriele Jedlitschky
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| | - Karsten Schrör
- Institut für Pharmakologie und Klinische Pharmakologie, Universitätsklinikum Düsseldorf, Universitätsstrasse 1, 40225 Düsseldorf, Germany
| | - Bernhard H. Rauch
- Institut für Pharmakologie, Universitätsmedizin Greifswald, Felix-Hausdorf Strasse 3, 17487 Greifswald, Germany
| |
Collapse
|
16
|
Hsu CK, Lin CC, Hsiao LD, Yang CM. Mevastatin ameliorates sphingosine 1-phosphate-induced COX-2/PGE2-dependent cell migration via FoxO1 and CREB phosphorylation and translocation. Br J Pharmacol 2015; 172:5360-76. [PMID: 26359950 DOI: 10.1111/bph.13326] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 08/19/2015] [Accepted: 09/03/2015] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine 1-phosphate (S1P), an important inflammatory mediator, has been shown to regulate COX-2 production and promote various cellular responses such as cell migration. Mevastatin, an inhibitor of 3-hydroxy-3-methylglutaryl-CoA reductase (HMG-CoA), effectively inhibits inflammatory responses. However, the mechanisms underlying S1P-evoked COX-2-dependent cell migration, which is modulated by mevastatin in human tracheal smooth muscle cells (HTSMCs) remain unclear. EXPERIMENTAL APPROACH The expression of COX-2 was determined by Western blotting, real time-PCR and promoter analyses. The signalling molecules were investigated by pretreatment with respective pharmacological inhibitors or transfection with siRNAs. The interaction between COX-2 promoter and transcription factors was determined by chromatin immunoprecipitation assay. Finally, the effect of mevastatin on HTSMC migration and leukocyte counts in BAL fluid and COX-2 expression induced by S1P was determined by a cell migration assay, cell counting and Western blot. KEY RESULTS S1P stimulated mTOR activation through the Nox2/ROS and PI3K/Akt pathways, which can further stimulate FoxO1 phosphorylation and translocation to the cytosol. We also found that S1P induced CREB activation and translocation via an mTOR-independent signalling pathway. Finally, we showed that pretreatment with mevastatin markedly reduced S1P-induced cell migration and COX-2/PGE2 production via a PPARγ-dependent signalling pathway. CONCLUSIONS AND IMPLICATIONS Mevastatin attenuates the S1P-induced increased expression of COX-2 and cell migration via the regulation of FoxO1 and CREB phosphorylation and translocation by PPARγ in HTSMCs. Mevastatin could be beneficial for prevention of airway inflammation in the future.
Collapse
Affiliation(s)
- Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anaesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anaesthetics, Chang Gung Memorial Hospital at Lin-Kou and College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.,Research Center for Industry of Human Ecology and Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan
| |
Collapse
|
17
|
Xing XQ, Li YL, Zhang YX, Xiao Y, Li ZD, Liu LQ, Zhou YS, Zhang HY, Liu YH, Zhang LH, Zhuang M, Chen YP, Ouyang SR, Wu XW, Yang J. Sphingosine kinase 1/sphingosine 1-phosphate signalling pathway as a potential therapeutic target of pulmonary hypertension. Int J Clin Exp Med 2015; 8:11930-11935. [PMID: 26550106 DOI: pmid/26550106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 05/28/2015] [Indexed: 02/08/2023]
Abstract
Pulmonary hypertension is characterized by extensive vascular remodelling, leading to increased pulmonary vascular resistance and eventual death due to right heart failure. The pathogenesis of pulmonary hypertension involves vascular endothelial dysfunction and disordered vascular smooth muscle cell (VSMC) proliferation and migration, but the exact processes remain unknown. Sphingosine 1-phosphate (S1P) is a bioactive lysophospholipid involved in a wide spectrum of biological processes. S1P has been shown to regulate VSMC proliferation and migration and vascular tension via a family of five S1P G-protein-coupled receptors (S1P1-SIP5). S1P has been shown to have both a vasoconstrictive and vasodilating effect. The S1P receptors S1P1 and S1P3 promote, while S1P2 inhibits VSMC proliferation and migration in vitro in response to S1P. Moreover, it has been reported recently that sphingosine kinase 1 and S1P2 inhibitors might be useful therapeutic agents in the treatment of empirical pulmonary hypertension. The sphingosine kinase 1/S1P signalling pathways may play a role in the pathogenesis of pulmonary hypertension. Modulation of this pathway may offer novel therapeutic strategies.
Collapse
Affiliation(s)
- Xi-Qian Xing
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Li Li
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yu-Xuan Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yi Xiao
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Zhi-Dong Li
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Li-Qiong Liu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yu-Shan Zhou
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Hong-Yan Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Hong Liu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Li-Hui Zhang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Min Zhuang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Yan-Ping Chen
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Sheng-Rong Ouyang
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Xu-Wei Wu
- First Department of Respiratory Medicine, Yan'an Hospital Affiliated to Kunming Medical University Kunming, Yunnan, China
| | - Jiao Yang
- First Department of Respiratory Medicine, First Affiliated Hospital of Kunming Medical University Kunming, Yunnan, China
| |
Collapse
|
18
|
Ong WY, Herr DR, Farooqui T, Ling EA, Farooqui AA. Role of sphingomyelinases in neurological disorders. Expert Opin Ther Targets 2015; 19:1725-42. [DOI: 10.1517/14728222.2015.1071794] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
19
|
Chien HY, Lu CS, Chuang KH, Kao PH, Wu YL. Attenuation of LPS-induced cyclooxygenase-2 and inducible NO synthase expression by lysophosphatidic acid in macrophages. Innate Immun 2015; 21:635-46. [DOI: 10.1177/1753425915576345] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/12/2015] [Indexed: 01/09/2023] Open
Abstract
LPS can activate the inflammatory cascades by inducing various inflammatory mediators, such as prostaglandin E2 (PGE2) resulting from cyclooxygenase-2 (COX-2), and NO produced by inducible NO synthase (iNOS). Lysophosphatidic acid (LPA) has been demonstrated to participate in inflammation. This study aimed to clarify the impact and the involving mechanisms of LPA on LPS-incurred inflammation in macrophages. First, LPA appeared to attenuate LPS-induced protein and mRNA expression of COX-2 and iNOS genes, as well as production of PGE2 and NO. By using selective inhibitors targeting various signaling players, the inhibitory G protein alpha subunit (Gαi) seemed to be involved in the effect of LPA; p38, ERK and NF-κB were involved in the LPS-mediated COX-2/PGE2 pathway; and p38, JNK, phosphoinositide-3-kinase and NF-κB were involved in the LPS-mediated iNOS/NO pathway. LPA was able to diminish LPS-induced phosphorylation of p38 and Akt, as well as NF-κB p65 nuclear translocation. By utilization of inhibitors of COX-2 and iNOS, there appeared to be no modulation between the COX-2/PGE2 and the iNOS/NO signaling pathways. Our findings demonstrate a clear anti-inflammatory role of LPA acting via Gαi in LPS-mediated inflammatory response in macrophages, owing, at least in part, to its suppressive effect on LPS-induced activation of p38, Akt and NF-κB.
Collapse
Affiliation(s)
- Han-Yuan Chien
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Chih-Shen Lu
- Department of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan
| | - Kun-Han Chuang
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Pu-Hong Kao
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Lin Wu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
20
|
Hsu CK, Lee IT, Lin CC, Hsiao LD, Yang CM. Sphingosine-1-Phosphate Mediates COX-2 Expression and PGE2/IL-6 Secretion via c-Src-Dependent AP-1 Activation. J Cell Physiol 2014; 230:702-15. [DOI: 10.1002/jcp.24795] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 09/05/2014] [Indexed: 11/08/2022]
Affiliation(s)
- Chih-Kai Hsu
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - I-Ta Lee
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics; Chang Gung Memorial Hospital at Lin-Kou and College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics; Chang Gung Memorial Hospital at Lin-Kou and College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| | - Chuen-Mao Yang
- Department of Physiology and Pharmacology and Health Aging Research Center; College of Medicine; Chang Gung University; Kwei-San Tao-Yuan Taiwan
| |
Collapse
|
21
|
Fujii K, Machida T, Iizuka K, Hirafuji M. Sphingosine 1-phosphate increases an intracellular Ca(2+) concentration via S1P3 receptor in cultured vascular smooth muscle cells. J Pharm Pharmacol 2014; 66:802-10. [PMID: 24450400 DOI: 10.1111/jphp.12214] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 12/07/2013] [Indexed: 12/29/2022]
Abstract
OBJECTIVE We investigated the effect of sphingosine 1-phosphate (S1P) on intracellular Ca(2+) dynamics in rat vascular smooth muscle cells (VSMCs). METHODS Intracellular Ca(2+) concentration ([Ca(2+) ]i) was determined using a fluorescence dye fura-2/AM. Small interfering RNAs (siRNA) were transfected into VSMCs to deplete the expression of S1P2 and S1P3 receptors. KEY FINDINGS S1P induced a rapid and transient elevation in [Ca(2+) ]i, which was maximal 1 min after the stimulation, followed by a sustained increase. When extracellular Ca(2+) was removed, a decrease in resting level and a small and transient increase in [Ca(2+) ]i by S1P stimulation were observed. siRNA targeted for the S1P3 receptor almost completely inhibited the S1P-induced increase in [Ca(2+) ]i. The rapid and transient increase in [Ca(2+) ]i was significantly inhibited by diltiazem at a high concentration. Pertussis toxin and a phospholipase C (PLC) inhibitor inhibited the S1P-induced increase in [Ca(2+) ]i regardless of the presence of extracellular Ca(2+) . Furthermore, S1P activated store-operated and receptor-operated Ca(2+) entry. CONCLUSIONS These results suggest that S1P increases [Ca(2+) ]i via the S1P3 receptor by inducing an influx of extracellular Ca(2+) partially through the voltage-dependent Ca(2+) channels, as well as by mobilizing Ca(2+) from its intracellular stores. S1P3 receptor-coupled Gi/o protein and PLC activation mediate the mechanisms.
Collapse
Affiliation(s)
- Kazumi Fujii
- Department of Pharmacological Sciences, School of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido, Japan
| | | | | | | |
Collapse
|
22
|
Wan Z, Woodward DF, Stamer WD. Endogenous Bioactive Lipids and the Regulation of Conventional Outflow Facility. EXPERT REVIEW OF OPHTHALMOLOGY 2014; 3:457-470. [PMID: 19381354 DOI: 10.1586/17469899.3.4.457] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Perturbation of paracrine signaling within the human conventional outflow pathway influences tissue homeostasis and outflow function. For example, exogenous introduction of the bioactive lipids, sphingosine-1-phosphate, anandamide or prostaglandin F(2α), to conventional outflow tissues alters the rate of drainage of aqueous humor through the trabecular meshwork, and into Schlemm's canal. This review summarizes recent data that characterizes endogenous bioactive lipids, their receptors and associated signaling partners in the conventional outflow tract. We also discuss the potential of targeting such signaling pathways as a strategy for the development of therapeutics to treat ocular hypertension and glaucoma.
Collapse
Affiliation(s)
- Zhou Wan
- Department of Ophthalmology and Vision Science, University of Arizona, Tucson, Arizona
| | | | | |
Collapse
|
23
|
Völzke A, Koch A, Meyer Zu Heringdorf D, Huwiler A, Pfeilschifter J. Sphingosine 1-phosphate (S1P) induces COX-2 expression and PGE2 formation via S1P receptor 2 in renal mesangial cells. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:11-21. [PMID: 24064301 DOI: 10.1016/j.bbalip.2013.09.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Revised: 08/27/2013] [Accepted: 09/17/2013] [Indexed: 12/21/2022]
Abstract
Understanding the mechanisms of sphingosine 1-phosphate (S1P)-induced cyclooxygenase (COX)-2 expression and prostaglandin E2 (PGE2) formation in renal mesangial cells may provide potential therapeutic targets to treat inflammatory glomerular diseases. Thus, we evaluated the S1P-dependent signaling mechanisms which are responsible for enhanced COX-2 expression and PGE2 formation in rat mesangial cells under basal conditions. Furthermore, we investigated whether these mechanisms are operative in the presence of angiotensin II (Ang II) and of the pro-inflammatory cytokine interleukin-1β (IL-1β). Treatment of rat and human mesangial cells with S1P led to concentration-dependent enhanced expression of COX-2. Pharmacological and molecular biology approaches revealed that the S1P-dependent increase of COX-2 mRNA and protein expression was mediated via activation of S1P receptor 2 (S1P2). Further, inhibition of Gi and p42/p44 MAPK signaling, both downstream of S1P2, abolished the S1P-induced COX-2 expression. In addition, S1P/S1P2-dependent upregulation of COX-2 led to significantly elevated PGE2 levels, which were further potentiated in the presence of Ang II and IL-1β. A functional consequence downstream of S1P/S1P2 signaling is mesangial cell migration that is stimulated by S1P. Interestingly, inhibition of COX-2 by celecoxib and SC-236 completely abolished the migratory response. Overall, our results demonstrate that extracellular S1P induces COX-2 expression via activation of S1P2 and subsequent Gi and p42/p44 MAPK-dependent signaling in renal mesangial cells leading to enhanced PGE2 formation and cell migration that essentially requires COX-2. Thus, targeting S1P/S1P2 signaling pathways might be a novel strategy to treat renal inflammatory diseases.
Collapse
Affiliation(s)
- Anja Völzke
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Johann Wolfgang Goethe-Universität, Theodor-Stern-Kai 7, D-60590 Frankfurt am Main, Germany.
| | | | | | | | | |
Collapse
|
24
|
Kim YD, Han KT, Lee J, Park CG, Kim MY, Shahi PK, Zuo DC, Choi S, Jun JY. Effects of sphingosine-1-phosphate on pacemaker activity of interstitial cells of Cajal from mouse small intestine. Mol Cells 2013; 35:79-86. [PMID: 23307289 PMCID: PMC3887851 DOI: 10.1007/s10059-013-2282-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/03/2012] [Accepted: 12/03/2012] [Indexed: 01/28/2023] Open
Abstract
Interstitial cells of Cajal (ICC) are the pacemaker cells that generate the rhythmic oscillation responsible for the production of slow waves in gastrointestinal smooth muscle. Spingolipids are known to present in digestive system and are responsible for multiple important physiological and pathological processes. In this study, we are interested in the action of sphingosine 1-phosphate (S1P) on ICC. S1P depolarized the membrane and increased tonic inward pacemaker currents. FTY720 phosphate (FTY720P, an S1P(1,3,4,5) agonist) and SEW 2871 (an S1P(1) agonist) had no effects on pacemaker activity. Suramin (an S1P(3) antagonist) did not block the S1P-induced action on pacemaker currents. However, JTE-013 (an S1P(2) antagonist) blocked the S1P-induced action. RT-PCR revealed the presence of the S1P(2) in ICC. Calphostin C (a protein kinase C inhibitor), NS-398 (a cyclooxygenase-2 inhibitor), PD 98059 (a p42/44 inhibitor), or SB 203580 (a p38 inhibitor) had no effects on S1P-induced action. However, c-jun NH(2)-terminal kinase (JNK) inhibitor II suppressed S1P-induced action. External Ca(2+)-free solution or thapsigargin (a Ca(2+)-ATPase inhibitor of endoplasmic reticulum) suppressed action of S1P on ICC. In recording of intracellular Ca(2+) ([Ca(2+)](i)) concentration using fluo-4/AM S1P increased intensity of spontaneous [Ca(2+)](i) oscillations in ICC. These results suggest that S1P can modulate pacemaker activity of ICC through S1P(2) via regulation of external and internal Ca(2+) and mitogenactivated protein kinase activation.
Collapse
Affiliation(s)
- Young Dae Kim
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Kyoung Taek Han
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Jun Lee
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Chan Guk Park
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | - Man Yoo Kim
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| | | | | | | | - Jae Yeoul Jun
- Department of Physiology, Department of Internal Medicine, College of Medicine, Chosun University, Gwangju 501-759,
Korea
| |
Collapse
|
25
|
Differential expression of sphingosine-1-phosphate receptors in abdominal aortic aneurysms. Mediators Inflamm 2012; 2012:643609. [PMID: 22547907 PMCID: PMC3323867 DOI: 10.1155/2012/643609] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 01/19/2012] [Indexed: 11/17/2022] Open
Abstract
Objective. Inflammation plays a key role in the pathophysiology of abdominal aortic aneurysms (AAAs). Newly discovered Sphingosine-1-Phosphate Receptors (S1P receptors) are critical in modulating inflammatory response via prostaglandin production. The aim of the current study was to investigate the expression of different S1P receptors in AAAs and compared with normal aortas at the protein level. Materials and Methods. Aortic specimens were harvested during aortic reconstructive surgery for the AAA group or during organ transplant for the control group. The protein expression of S1P1, 2 and 3 in AAAs and normal aortas was assessed by Western blotting and immunohistochemical analysis. Results. There were 40 AAAs and 20 control aortas collected for the receptor analysis. For Western blot analysis, S1P1 expression was not detected in either group; S1P2 protein was constitutively detected in both types of aortas but its expression level was significantly decreased
by 73% (P < 0.05) in AAAs compared with the control group. In contrast, strong S1P3 expression was detected in AAAs aortas but not in normal aortas. Immumohistochemical staining showed
similar results, except a weak S1P3 signal was detectable in normal aortas. Conclusions. Western blot and staining results consistently showed the down-regulation of the S1P2 protein with simultaneous up-regulation of the S1P3 protein in AAAs. Since those newly discovered receptors play an important role in the inflammatory cascade, the modulating of S1P signaling, particularly via S1P2 and S1P3, could represent novel therapeutic targets in future AAA treatments.
Collapse
|
26
|
Kue CS, Jung MY, Cho D, Kim TS. C6-ceramide enhances Interleukin-12-mediated T helper type 1 cell responses through a cyclooxygenase-2-dependent pathway. Immunobiology 2011; 217:601-9. [PMID: 22112438 DOI: 10.1016/j.imbio.2011.10.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 10/25/2011] [Indexed: 12/24/2022]
Abstract
Ceramides, lipid molecules located predominantly within the plasma membrane of a cell, can function as second messengers, and have been known to carry out a number of cellular functions. T helper type 1 (Th1) immune responses are known to be involved in the cellular immunity, which is crucial in the cancer and allergy immunotherapy. This study was designed to evaluate the effects of ceramides on T helper cell responses and their underlying mechanisms. We demonstrated that a cell-permeable C6-ceramide (C6) together with IL-12 enhanced Th1 cell differentiation, whereas C6 alone had no effects, as demonstrated by the increased populations of IFN-γ expressing CD4(+) T cells and the up-regulation of IFN-γ production from CD4(+) T cells. In contrast, C2-ceramide and long chain ceramides (C16 and C24) did not affect the Th1 responses. C6 treatment was shown to increase the expression of T-bet, a master transcription factor of Th1 responses, in a dose-dependent fashion. Furthermore, C6 increased the expression of cyclooxygenase-2 (COX-2) in CD4(+) T cells. The C6-mediated increase of IFN-γ production and IFN-γ expressing CD4(+) T cell populations were significantly suppressed by a COX-2 specific inhibitor (NS-398) in a dose-dependent manner. T-bet expression was also decreased by NS-398 treatment, thereby indicating that C6 ceramide enhances Th1 responses via a COX-2 dependent pathway. This result demonstrates that C6 may be utilized in therapies for the treatment of immune diseases such cancer and allergy by enhancing the Th1 activity.
Collapse
Affiliation(s)
- Chin Siang Kue
- Division of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | |
Collapse
|
27
|
Machida T, Ohta M, Onoguchi A, Iizuka K, Sakai M, Minami M, Hirafuji M. 5-Hydroxytryptaime induces cyclooxygenase-2 in rat vascular smooth muscle cells: Mechanisms involving Src, PKC and MAPK activation. Eur J Pharmacol 2011; 656:19-26. [DOI: 10.1016/j.ejphar.2010.12.044] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 12/02/2010] [Accepted: 12/22/2010] [Indexed: 11/16/2022]
|
28
|
Spijkers LJA, Alewijnse AE, Peters SLM. Sphingolipids and the orchestration of endothelium-derived vasoactive factors: when endothelial function demands greasing. Mol Cells 2010; 29:105-11. [PMID: 20127284 DOI: 10.1007/s10059-010-0042-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2010] [Accepted: 01/08/2010] [Indexed: 02/02/2023] Open
Abstract
Vasomotor tone is regulated by a complex interplay of a variety of extrinsic neurohumoral and intrinsic factors. It is the endothelium that has a major influence on smooth muscle cell tone via the release of intrinsic vasoactive factors and is therefore an important regulator of vasomotor tone. Sphingolipids are an emerging class of lipid mediators with important physiological properties. In the last two decades it has not only become increasingly clear that sphingolipid signaling plays a pivotal role in immune function, but also its role in the vascular system is now becoming more recognized. In this mini-review we will highlight the possible cross-talk between sphingolipids and intrinsic vasoactive factors released by the endothelium. Via this cross-talk sphingolipids can orchestrate vasomotor tone and may therefore also be involved in the pathophysiology of disease states associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Léon J A Spijkers
- Department Pharmacology and Pharmacotherapy, Academic Medical Center, Amsterdam, The Netherlands
| | | | | |
Collapse
|
29
|
Kawabe JI, Ushikubi F, Hasebe N. Prostacyclin in Vascular Diseases - Recent Insights and Future Perspectives -. Circ J 2010; 74:836-43. [DOI: 10.1253/circj.cj-10-0195] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Jun-ichi Kawabe
- Departments of Cardiovascular Regeneration and Innovation, Asahikawa Medical College
| | | | - Naoyuki Hasebe
- Departments of Cardiovascular Regeneration and Innovation, Asahikawa Medical College
- Department of Internal Medicine, Cardiovascular, Respiratory and Neurology Division, Asahikawa Medical College
| |
Collapse
|
30
|
Li C, Jiang X, Yang L, Liu X, Yue S, Li L. Involvement of sphingosine 1-phosphate (SIP)/S1P3 signaling in cholestasis-induced liver fibrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1464-72. [PMID: 19729475 DOI: 10.2353/ajpath.2009.090037] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Bioactive sphingosine 1-phosphate (S1P) and S1P receptors (S1PRs) have been implicated in many critical cellular events, including inflammation, cancer, and angiogenesis. However, the role of S1P/S1PR signaling in the pathogenesis of liver fibrosis has not been well documented. In this study, we found that S1P levels and S1P(3) receptor expression in liver tissue were markedly up-regulated in a mouse model of cholestasis-induced liver fibrosis. In addition, the S1P(3) receptor was also expressed in green fluorescent protein transgenic bone marrow (BM)-derived cells found in the damaged liver of transplanted chimeric mice that underwent bile duct ligation. Silencing of S1P(3) expression significantly inhibited S1P-induced BM cell migration in vitro. Furthermore, a selective S1P(3) receptor antagonist, suramin, markedly reduced the number of BM-derived cells during cholestasis. Interestingly, suramin administration clearly ameliorated bile duct ligation-induced hepatic fibrosis, as demonstrated by attenuated deposition of collagen type I and III, reduced smooth muscle alpha-actin expression, and decreased total hydroxyproline content. In conclusion, our data suggest that S1P/S1P(3) signaling plays an important role in cholestasis-induced liver fibrosis through mediating the homing of BM cells. Modulation of S1PR activity may therefore represent a new antifibrotic strategy.
Collapse
Affiliation(s)
- Changyong Li
- Department of Cell Biology, Municipal Laboratory for Liver Protection and Regulation of Regeneration, Capital Medical University, Beijing, China
| | | | | | | | | | | |
Collapse
|
31
|
Sattler K, Levkau B. Sphingosine-1-phosphate as a mediator of high-density lipoprotein effects in cardiovascular protection. Cardiovasc Res 2009; 82:201-11. [PMID: 19233866 DOI: 10.1093/cvr/cvp070] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) has gained special attention in the high-density lipoprotein (HDL) field because HDL is the most prominent plasma carrier of S1P and because the S1P content of HDL may be responsible for many of the pleiotropic functions of HDL. This revelation has come from the evidence that HDL employ S1P receptors and signalling pathways to implement several HDL-ascribed biological effects as diverse as endothelial nitric oxide production, vasodilation, survival, and cardioprotection. This review focuses on HDL effects that are completely or partially mediated by the S1P content of the HDL particle and differentiates them from genuine HDL effects that are S1P-independent. In addition, the functional properties of 'free', HDL-unbound S1P are sometimes different from or even contrary to those of HDL-associated S1P. The nature of the physical interactions between HDL and local and systemic S1P production will be discussed as well as their consequences for organ function. Finally, we will elucidate the potential benefits and limitations of S1P analogues as a new class of functional HDL mimetics for cardiovascular therapy.
Collapse
Affiliation(s)
- Katherine Sattler
- Institute of Pathophysiology, Zentrum für Innere Medizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122 Essen, Germany
| | | |
Collapse
|
32
|
Ohama T, Okada M, Murata T, Brautigan DL, Hori M, Ozaki H. Sphingosine-1-phosphate enhances IL-1{beta}-induced COX-2 expression in mouse intestinal subepithelial myofibroblasts. Am J Physiol Gastrointest Liver Physiol 2008; 295:G766-75. [PMID: 18703638 DOI: 10.1152/ajpgi.90423.2008] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Intestinal subepithelial myofibroblasts (SEMFs) is a specific population of cells involved in intestinal inflammation and carcinogenesis via an elaborate network of cytokines, chemokines and other inflammatory factors, including PGE(2). Sphingosine-1-phosphate (S1P) has been implicated as an important mediator of inflammation and cancer and in certain cell types increases cyclooxygenase-2 (COX-2) expression. In the present study, we aimed to assess involvement of S1P in COX-2 expression by SEMFs. Primary SEMFs were obtained from C57BL/6J mouse and their identity was verified by fluorescent staining of specific marker proteins. Expression of S1P receptors 1, 2, 3 and sphingosine kinases 1 and 2 in SEMFs were determined by RT-PCR analysis. COX-2 expression and PGE(2) production were assayed by Western blotting and ELISA, respectively. COX-2 mRNA stability was assayed by Northern blotting. S1P produced dose-dependent increase in COX-2 expression, resulting in increased PGE(2) release from SEMFs. Using specific inhibitors, we show that actions of p38, ERK, IKK, and PKC were involved in S1P-induced COX-2 expression. On the other hand, p38 and PKC had lesser roles in IL-1beta-induced COX-2 expression. Inhibition of sphingosine kinase to block S1P production did not affect IL-1beta-induced COX-2 expression, but S1P amplified IL-1beta-induced p38 activation and COX-2 expression. PKC inhibition blocked S1P amplified COX-2 expression. S1P addition increased COX-2 mRNA stability. In SEMFs, S1P amplifies IL-1beta-induced COX-2 expression through increased mRNA stability. These observations point to involvement of S1P in activation of SEMFs that may contribute to intestinal inflammation and carcinogenesis.
Collapse
Affiliation(s)
- Takashi Ohama
- Dept. of Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The Univ. of Tokyo, Yayoi 1-1-1, Bunkyo-ku, Tokyo 113-8657, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Machida T, Hamaya Y, Izumi S, Hamaya Y, Iizuka K, Igarashi Y, Minami M, Levi R, Hirafuji M. Sphingosine 1-phosphate inhibits nitric oxide production induced by interleukin-1beta in rat vascular smooth muscle cells. J Pharmacol Exp Ther 2008; 325:200-9. [PMID: 18171908 DOI: 10.1124/jpet.107.127290] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2025] Open
Abstract
Sphingosine 1-phosphate (S1P) is a lipid mediator that exerts potent and diverse biological effects on several cardiovascular cells. We investigated the effect of S1P on interleukin (IL)-1beta-induced nitric oxide (NO) production and inducible NO synthase (iNOS) expression in rat vascular smooth muscle cells (VSMCs). S1P inhibited NO production at concentrations higher than 0.1 muM; this was associated with the inhibition of iNOS protein and mRNA expression. S1P also inhibited IL-1beta-induced GTP cyclohydrolase I (GTPCH) mRNA expression. Pertussis toxin (PTX) partially attenuated the inhibitory effects of S1P on NO production and iNOS protein induction, whereas it completely blocked the inhibitory effects on iNOS and GTPCH mRNA expression. S1P inhibited iNOS expression in Ca(2+)-depleted conditions; PTX did not modify this effect. The Rho kinase inhibitor Y 27632 partially but significantly attenuated the inhibitory effect of S1P on iNOS expression in Ca(2+)-depleted condition but did not affect it in the presence of Ca(2+). S1P significantly inhibited IL-1beta-induced persistent activation of extracellular signal-regulated kinase (ERK) but had no effect in Ca(2+)-depleted conditions. Thus, S1P inhibits IL-1beta induction of NO production and iNOS expression in rat VSMCs through multiple mechanisms involving both PTX-sensitive and -insensitive G proteins coupled to S1P receptors. Furthermore, Ca(2+)-dependent ERK inhibition and Ca(2+)-independent Rho kinase activation might be involved in the inhibitory mechanism of iNOS expression. Through its action on NO production by VSMCs, S1P may play an important role in the progression of local vascular injury associated with thrombosis, atherosclerosis, and hypertension.
Collapse
Affiliation(s)
- Takuji Machida
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Health Sciences University of Hokkaido, Ishikari-Tobetsu, Hokkaido 061-0293, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Ikeda Y, Ohashi K, Shibata R, Pimentel DR, Kihara S, Ouchi N, Walsh K. Cyclooxygenase-2 induction by adiponectin is regulated by a sphingosine kinase-1 dependent mechanism in cardiac myocytes. FEBS Lett 2008; 582:1147-50. [PMID: 18339320 DOI: 10.1016/j.febslet.2008.03.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2007] [Revised: 02/29/2008] [Accepted: 03/03/2008] [Indexed: 12/30/2022]
Abstract
The adipose-derived plasma protein, adiponectin (APN), has various protective effects on cardiovascular diseases. In this study, we show that endogenous APN is required for full cyclooxygenase-2 (COX-2) induction by ischemia-reperfusion injury in the heart in vivo. In rat neonatal cardiac myocytes, APN-induced COX-2 expression was reduced by treatment with a sphingosine kinase-1 (SphK-1) inhibitor or siRNA targeting SphK-1. Treatment with a sphingosine-1-phosphate (S1P) receptor antagonist also diminished COX-2 expression in response to APN stimulation. These findings suggest that APN is a physiological regulator of COX-2 signaling in the heart and that this regulation occurs in part via a SphK-1-S1P receptor dependent mechanism in cardiac myocytes.
Collapse
Affiliation(s)
- Yasumasa Ikeda
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, W611, Boston, MA 02118, USA
| | | | | | | | | | | | | |
Collapse
|
35
|
Balakumar P, Kaur T, Singh M. Potential target sites to modulate vascular endothelial dysfunction: Current perspectives and future directions. Toxicology 2008; 245:49-64. [DOI: 10.1016/j.tox.2007.12.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2007] [Revised: 12/13/2007] [Accepted: 12/13/2007] [Indexed: 12/26/2022]
|