1
|
Obara K, Yoshioka K, Tanaka Y. [Platelet-activating Factor (PAF) in Urinary Bladder Smooth Muscle (UBSM): Its Novel Role as a Potential Inducer of Detrusor Overactivity and the Mechanisms Underlying Its Effects in Enhancing UBSM Mechanical Activities]. YAKUGAKU ZASSHI 2024; 144:997-1007. [PMID: 39496432 DOI: 10.1248/yakushi.24-00136] [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: 11/06/2024]
Abstract
Platelet-activating factor (PAF), a phospholipid mediator, was discovered in 1972 as an inducer of platelet aggregation. Subsequent studies have revealed that PAF has a variety of biological functions, such as its role as a potent proinflammatory mediator. Additionally, PAF regulates the contractile functions of various types of smooth muscle (SM), such as the (1) endothelium-dependent relaxation of vascular SM; (2) contraction and epithelium-dependent relaxation of airway SM; (3) contraction of gastrointestinal SM; and (4) contraction of uterine SM, which occurs more strongly in pregnant females. PAF is produced in platelets, monocytes, neutrophils, and macrophages, which are cells related to thrombus formation and inflammation/immune responses. Furthermore, PAF is produced in various other cells throughout the body. Interestingly, recent studies have focused on the urinary bladder (UB) as a PAF-producing organ since the accumulation of this phospholipid is enhanced in patients with bladder cancer and interstitial cystitis/bladder pain syndrome, especially those who smoke. Therefore, in UB tissue, PAF may play a substantial role as an inducer or enhancer of cancers and inflammatory diseases. However, the effects of PAF on the immediate mechanical activities of UBSM have not been investigated to date. In this regard, we recently discovered that PAF strongly enhances mechanical activities (muscle tone and spontaneous contractile activity) in UBSM tissues isolated from guinea pigs and mice. In this review article, we present our data on these PAF effects together with the possible underlying mechanisms. We also discuss the potential pathophysiological roles of this phospholipid in UB diseases and disorders.
Collapse
Affiliation(s)
- Keisuke Obara
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Kento Yoshioka
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| | - Yoshio Tanaka
- Department of Chemical Pharmacology, Faculty of Pharmaceutical Sciences, Toho University
| |
Collapse
|
2
|
Rodriguez-Cuenca S, Pellegrinelli V, Campbell M, Oresic M, Vidal-Puig A. Sphingolipids and glycerophospholipids - The "ying and yang" of lipotoxicity in metabolic diseases. Prog Lipid Res 2017; 66:14-29. [PMID: 28104532 DOI: 10.1016/j.plipres.2017.01.002] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/30/2016] [Accepted: 01/05/2017] [Indexed: 12/14/2022]
Abstract
Sphingolipids in general and ceramides in particular, contribute to pathophysiological mechanisms by modifying signalling and metabolic pathways. Here, we present the available evidence for a bidirectional homeostatic crosstalk between sphingolipids and glycerophospholipids, whose dysregulation contributes to lipotoxicity induced metabolic stress. The initial evidence for this crosstalk originates from simulated models designed to investigate the biophysical properties of sphingolipids in plasma membrane representations. In this review, we reinterpret some of the original findings and conceptualise them as a sort of "ying/yang" interaction model of opposed/complementary forces, which is consistent with the current knowledge of lipid homeostasis and pathophysiology. We also propose that the dysregulation of the balance between sphingolipids and glycerophospholipids results in a lipotoxic insult relevant in the pathophysiology of common metabolic diseases, typically characterised by their increased ceramide/sphingosine pools.
Collapse
Affiliation(s)
- S Rodriguez-Cuenca
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK.
| | - V Pellegrinelli
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK
| | - M Campbell
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK
| | - M Oresic
- Turku Centre for Biotechnology, University of Turku and Åbo Akademi University, FI -20520 Turku, Finland
| | - A Vidal-Puig
- Metabolic Research Laboratories, Wellcome Trust MRC Institute of Metabolic Science, Addenbrooke's Hospital, University of Cambridge. Cambridge, UK; Wellcome Trust Sanger Institute, Hinxton, UK.
| |
Collapse
|
3
|
Hiob MA, Trane AE, Wise SG, Bernatchez PN, Weiss AS. Tropoelastin enhances nitric oxide production by endothelial cells. Nanomedicine (Lond) 2016; 11:1591-7. [PMID: 27175893 DOI: 10.2217/nnm-2016-0052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
AIMS This study aimed to characterize the role of tropoelastin in eliciting a nitric oxide response in endothelial cells. MATERIALS AND METHODS Nitric oxide production in cells was quantified following the addition of known nitric oxide synthase pathway inhibitors such as LNAME and 1400W. The effect of eNOS siRNA knockdowns was studied using western blotting and assessed in the presence of PI3K-inhibitor, wortmannin. RESULTS Tropoelastin-induced nitric oxide production was LNAME and wortmannin sensitive, while being unaffected by treatment with 1400W. CONCLUSION Tropoelastin acts through a PI3K-specific pathway that leads to the phosphorylation of eNOS to enhance nitric oxide production in endothelial cells. This result points to the benefit of the use of tropoelastin in vascular applications, where NO production is a characteristic marker of vascular health.
Collapse
Affiliation(s)
- Matti A Hiob
- School of Life & Environmental Biosciences, University of Sydney, Sydney, NSW 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Andy E Trane
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z1Y6, Canada.,Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, Canada
| | - Steven G Wise
- School of Life & Environmental Biosciences, University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, University of Sydney, NSW 2006, Australia.,The Heart Research Institute, Sydney, NSW, 2042, Australia
| | - Pascal N Bernatchez
- Centre for Heart Lung Innovation, St Paul's Hospital, Vancouver, BC, V6Z1Y6, Canada.,Department of Anesthesiology, Pharmacology & Therapeutics, University of British Columbia, Vancouver, Canada
| | - Anthony S Weiss
- School of Life & Environmental Biosciences, University of Sydney, Sydney, NSW 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.,Bosch Institute, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
4
|
Igarashi J, Hashimoto T, Kubota Y, Shoji K, Maruyama T, Sakakibara N, Takuwa Y, Ujihara Y, Katanosaka Y, Mohri S, Naruse K, Yamashita T, Okamoto R, Hirano K, Kosaka H, Takata M, Konishi R, Tsukamoto I. Involvement of S1P1 receptor pathway in angiogenic effects of a novel adenosine-like nucleic acid analog COA-Cl in cultured human vascular endothelial cells. Pharmacol Res Perspect 2014; 2:e00068. [PMID: 25505610 PMCID: PMC4186426 DOI: 10.1002/prp2.68] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Accepted: 07/11/2014] [Indexed: 12/13/2022] Open
Abstract
COA-Cl (2Cl-C.OXT-A) is a recently developed adenosine-like nucleic acid analog that promotes angiogenesis via the mitogen-activated protein (MAP) kinases ERK1/2. Endothelial S1P1 receptor plays indispensable roles in developmental angiogenesis. In this study, we examined the functions of S1P1 in COA-Cl-induced angiogenic responses. Antagonists for S1P1, W146, and VPC23019, substantially but still partly inhibited the effects of COA-Cl with regard to ERK1/2 activation and tube formation in cultured human umbilical vein endothelial cells (HUVEC). Antagonists for adenosine A1 receptor and purinergic P2Y1 receptor were without effect. Genetic knockdown of S1P1 with siRNA, but not that of S1P3, attenuated COA-Cl-elicited ERK1/2 responses. The signaling properties of COA-Cl showed significant similarities to those of sphingosine 1-phosphate, an endogenous S1P1 ligand, in that both induced responses sensitive to pertussis toxin (Gα i/o inhibitor), 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid tetrakis (acetoxymethyl ester) (BAPTA-AM), (calcium chelator), and PP2 (c-Src tyrosine kinase inhibitor). COA-Cl elevated intracellular Ca2+ concentration and induced tyrosine phosphorylation of p130Cas, a substrate of c-Src, in HUVEC. COA-Cl displaced [3H]S1P in a radioligand-binding competition assay in chem-1 cells overexpressing S1P1. However, COA-Cl activated ERK1/2 in CHO-K1 cells that lack functional S1P1 receptor, suggesting the presence of additional yet-to-be-defined COA-Cl target in these cells. The results thus suggest the major contribution of S1P1 in the angiogenic effects of COA-Cl. However, other mechanism such as that seen in CHO-K1 cells may also be partly involved. Collectively, these findings may lead to refinement of the design of this nucleic acid analog and ultimately to development of small molecule-based therapeutic angiogenesis.
Collapse
Affiliation(s)
- Junsuke Igarashi
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Takeshi Hashimoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Yasuo Kubota
- Department of Dermatology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Kazuyo Shoji
- Department of Dermatology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Tokumi Maruyama
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan
| | - Norikazu Sakakibara
- Kagawa School of Pharmaceutical Sciences, Tokushima Bunri University 1314-1 Shido, Sanuki, Kagawa, 769-2193, Japan
| | - Yoh Takuwa
- Department of Cardiovascular Physiology, Kanazawa University School of Medicine 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Yoshihiro Ujihara
- Department of Physiology, Kawasaki Medical School 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Yuki Katanosaka
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Shitada-Cho 2-5-1, Kita-Ku, Okayama, 700-0914, Japan
| | - Satoshi Mohri
- Department of Physiology, Kawasaki Medical School 577 Matsushima, Kurashiki, 701-0192, Japan
| | - Keiji Naruse
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University Shitada-Cho 2-5-1, Kita-Ku, Okayama, 700-0914, Japan
| | - Tetsuo Yamashita
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Ryuji Okamoto
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Katsuya Hirano
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Hiroaki Kosaka
- Department of Cardiovascular Physiology, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Maki Takata
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Ryoji Konishi
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| | - Ikuko Tsukamoto
- Department of Pharmaco-Bio-Informatics, Faculty of Medicine, Kagawa University 1750-1 Ikenobe, Miki-Cho, Kita-Gun, Kagawa, 761-0793, Japan
| |
Collapse
|
5
|
Farhat N, Mamarbachi AM, Thorin E, Allen BG. Cloning, expression and purification of functionally active human angiopoietin-like protein 2. SPRINGERPLUS 2014; 3:337. [PMID: 25077060 PMCID: PMC4112196 DOI: 10.1186/2193-1801-3-337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Accepted: 06/25/2014] [Indexed: 11/10/2022]
Abstract
Angiopoietin-like protein 2 (Angptl2) is a secreted glycoprotein that has been implicated in angiogenesis, inflammation and atherosclerosis as well as enhancing the survival of human hematopoietic stem cells. Glycosylation of Angptl2 is required for biological activity and studies of angiopoietin-like protein 2 have been hindered by the lack of a source for the mature form of this protein. We describe a system that permits purification of the glycosylated form of human Angptl2 from conditioned media of stably transfected HEK 293 cells. To facilitate purification while retaining the integrity of Angptl2's endogenous N-terminal secretion signal peptide, GST was fused downstream of the Angptl2 coding sequence. Secreted Angptl2-GST was purified using a one-step glutathione-affinity purification scheme. The purity and identity of the resulting protein were confirmed by SDS-PAGE, immunoblotting, and mass spectrometry. N-Glycosidase treatment reduced the apparent molecular mass of Angptl2-GST on SDS-PAGE, confirming its glycosylation state. Purified human Angptl2-GST stimulated both HUVEC migration and microtubule formation in vitro. The yield of Angptl2-GST obtained was in quantities suitable for multiple applications including functional in vitro and in vivo assays.
Collapse
Affiliation(s)
- Nada Farhat
- Departments of Pharmacology, Montreal, QC H3T 1J4 Canada ; Montreal Heart Institute, Montréal, Québec H1T 1C8 Canada ; Pharsight Corporation Canada, 2000 Peel, Montreal, Québec H3A 2W5 Canada
| | | | - Eric Thorin
- Departments of Pharmacology, Montreal, QC H3T 1J4 Canada ; Department of Surgery, Université de Montréal, Université de Montréal, 5000 Belanger St, Montréal, Québec H1T 1C8 Canada ; Montreal Heart Institute, Montréal, Québec H1T 1C8 Canada
| | - Bruce G Allen
- Departments of Biochemistry, Montreal, QC H3T 1J4 Canada ; Department of Medicine, Montreal Heart Institute, Université de Montréal, 5000 Belanger St, Montréal, Québec H1T 1C8 Canada ; Montreal Heart Institute, Montréal, Québec H1T 1C8 Canada ; Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6 Canada
| |
Collapse
|
6
|
Caballero S, Swaney J, Moreno K, Afzal A, Kielczewski J, Stoller G, Cavalli A, Garland W, Hansen G, Sabbadini R, Grant MB. Anti-sphingosine-1-phosphate monoclonal antibodies inhibit angiogenesis and sub-retinal fibrosis in a murine model of laser-induced choroidal neovascularization. Exp Eye Res 2008; 88:367-77. [PMID: 18723015 DOI: 10.1016/j.exer.2008.07.012] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 07/10/2008] [Accepted: 07/13/2008] [Indexed: 12/30/2022]
Abstract
The efficacy of novel monoclonal antibodies that neutralize the pro-angiogenic mediator, sphingosine-1-phosphate (S1P), were tested using in vitro and in vivo angiogenesis models, including choroidal neovascularization (CNV) induced by laser disruption of Bruch's membrane. S1P receptor levels in human brain choroid plexus endothelial cells (CPEC), human lung microvascular endothelial cells, human retinal vascular endothelial cells, and circulating endothelial progenitor cells were examined by semi-quantitative PCR. The ability of murine or humanized anti-S1P monoclonal antibodies (mAbs) to inhibit S1P-mediated microvessel tube formation by CPEC on Matrigel was evaluated and capillary density in subcutaneous growth factor-loaded Matrigel plugs was determined following anti-S1P treatment. S1P promoted in vitro capillary tube formation in CPEC consistent with the presence of cognate S1P(1-5) receptor expression by these cells and the S1P antibody induced a dose-dependent reduction in microvessel tube formation. In a murine model of laser-induced rupture of Bruch's membrane, S1P was detected in posterior cups of mice receiving laser injury, but not in uninjured controls. Intravitreous injection of anti-S1P mAbs dramatically inhibited CNV formation and sub-retinal collagen deposition in all treatment groups (p<0.05 compared to controls), thereby identifying S1P as a previously unrecognized mediator of angiogenesis and subretinal fibrosis in this model. These findings suggest that neutralizing S1P with anti-S1P mAbs may be a novel method of treating patients with exudative age-related macular degeneration by reducing angiogenesis and sub-retinal fibrosis, which are responsible for visual acuity loss in this disease.
Collapse
Affiliation(s)
- Sergio Caballero
- Department of Pharmacology & Therapeutics, University of Florida, Gainesville, FL, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Singh IN, Hall ED. Multifaceted roles of sphingosine-1-phosphate: How does this bioactive sphingolipid fit with acute neurological injury? J Neurosci Res 2008; 86:1419-33. [DOI: 10.1002/jnr.21586] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
8
|
Doublier S, Ceretto M, Lupia E, Bravo S, Bussolati B, Camussi G. The proangiogenic phenotype of tumor-derived endothelial cells is reverted by the overexpression of platelet-activating factor acetylhydrolase. Clin Cancer Res 2007; 13:5710-8. [PMID: 17908960 DOI: 10.1158/1078-0432.ccr-07-0412] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We previously reported that human tumor-derived endothelial cells (TEC) have an angiogenic phenotype related to the autocrine production of several angiogenic factors. The purpose of the present study was to evaluate whether an enhanced synthesis of platelet-activating factor (PAF) might contribute to the proangiogenic characteristics of TEC and whether its inactivation might inhibit angiogenesis. EXPERIMENTAL DESIGN To address the potential role of PAF in the proangiogenic characteristics of TEC, we engineered TEC to stably overexpress human plasma PAF-acetylhydrolase (PAF-AH), the major PAF-inactivating enzyme, and we evaluated in vitro and in vivo angiogenesis. RESULTS TECs were able to synthesize a significantly enhanced amount of PAF compared with normal human microvascular endothelial cells when stimulated with thrombin, vascular endothelial growth factor, or soluble CD154. Transfection of TEC with PAF-AH (TEC-PAF-AH) significantly inhibited apoptosis resistance and spontaneous motility of TEC. In addition, PAF and vascular endothelial growth factor stimulation enhanced the motility and adhesion of TEC but not of TEC-PAF-AH. In vitro, TEC-PAF-AH lost the characteristic ability of TEC to form vessel-like structures when plated on Matrigel. Finally, when cells were injected s.c. within Matrigel in severe combined immunodeficiency mice or coimplanted with a renal carcinoma cell line, the overexpression of PAF-AH induced a significant reduction of functional vessel formation. CONCLUSIONS These results suggest that inactivation of PAF, produced by TEC, by the overexpression of plasma PAF-AH affects survival, migration, and the angiogenic response of TEC both in vitro and in vivo.
Collapse
Affiliation(s)
- Sophie Doublier
- Department of Internal Medicine, Centre for Molecular Biotechnology, University of Turin, Turin, Italy
| | | | | | | | | | | |
Collapse
|
9
|
Farooqui AA, Horrocks LA, Farooqui T. Interactions between neural membrane glycerophospholipid and sphingolipid mediators: A recipe for neural cell survival or suicide. J Neurosci Res 2007; 85:1834-50. [PMID: 17393491 DOI: 10.1002/jnr.21268] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The neural membranes contain phospholipids, sphingolipids, cholesterol, and proteins. Glycerophospholipids and sphingolipids are precursors for lipid mediators involved in signal transduction processes. Degradation of glycerophospholipids by phospholipase A(2) (PLA(2)) generates arachidonic acid (AA) and docosahexaenoic acids (DHA). Arachidonic acid is metabolized to eicosanoids and DHA is metabolized to docosanoids. The catabolism of glycosphingolipids generates ceramide, ceramide 1-phosphate, sphingosine, and sphingosine 1-phosphate. These metabolites modulate PLA(2) activity. Arachidonic acid, a product derived from glycerophospholipid catabolism by PLA(2), modulates sphingomyelinase (SMase), the enzyme that generates ceramide and phosphocholine. Furthermore, sphingosine 1-phosphate modulates cyclooxygenase, an enzyme responsible for eicosanoid production in brain. This suggests that an interplay and cross talk occurs between lipid mediators of glycerophospholipid and glycosphingolipid metabolism in brain tissue. This interplay between metabolites of glycerophospholipid and sphingolipid metabolism may play an important role in initiation and maintenance of oxidative stress associated with neurologic disorders as well as in neural cell proliferation, differentiation, and apoptosis. Recent studies indicate that PLA(2) and SMase inhibitors can be used as neuroprotective and anti-apoptotic agents. Development of novel inhibitors of PLA(2) and SMase may be useful for the treatment of oxidative stress, and apoptosis associated with neurologic disorders such as stroke, Alzheimer disease, Parkinson disease, and head and spinal cord injuries.
Collapse
Affiliation(s)
- Akhlaq A Farooqui
- Department of Molecular and Cellular Biochemistry, Ohio State University, Columbus, Ohio 43210, USA
| | | | | |
Collapse
|
10
|
Zhao Y, Kalari SK, Usatyuk PV, Gorshkova I, He D, Watkins T, Brindley DN, Sun C, Bittman R, Garcia JGN, Berdyshev EV, Natarajan V. Intracellular generation of sphingosine 1-phosphate in human lung endothelial cells: role of lipid phosphate phosphatase-1 and sphingosine kinase 1. J Biol Chem 2007; 282:14165-77. [PMID: 17379599 PMCID: PMC2659598 DOI: 10.1074/jbc.m701279200] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) regulates diverse cellular functions through extracellular ligation to S1P receptors, and it also functions as an intracellular second messenger. Human pulmonary artery endothelial cells (HPAECs) effectively utilized exogenous S1P to generate intracellular S1P. We, therefore, examined the role of lipid phosphate phosphatase (LPP)-1 and sphingosine kinase1 (SphK1) in converting exogenous S1P to intracellular S1P. Exposure of (32)P-labeled HPAECs to S1P or sphingosine (Sph) increased the intracellular accumulation of [(32)P]S1P in a dose- and time-dependent manner. The S1P formed in the cells was not released into the medium. The exogenously added S1P did not stimulate the sphingomyelinase pathway; however, added [(3)H]S1P was hydrolyzed to [(3)H]Sph in HPAECs, and this was blocked by XY-14, an inhibitor of LPPs. HPAECs expressed LPP1-3, and overexpression of LPP-1 enhanced the hydrolysis of exogenous [(3)H]S1P to [(3)H]Sph and increased intracellular S1P production by 2-3-fold compared with vector control cells. Down-regulation of LPP-1 by siRNA decreased intracellular S1P production from extracellular S1P but had no effect on the phosphorylation of Sph to S1P. Knockdown of SphK1, but not SphK2, by siRNA attenuated the intracellular generation of S1P. Overexpression of wild type SphK1, but not SphK2 wild type, increased the accumulation of intracellular S1P after exposure to extracellular S1P. These studies provide the first direct evidence for a novel pathway of intracellular S1P generation. This involves the conversion of extracellular S1P to Sph by LPP-1, which facilitates Sph uptake, followed by the intracellular conversion of Sph to S1P by SphK1.
Collapse
Affiliation(s)
- Yutong Zhao
- Department of Medicine, Section of Pulmonary and Critical Care Medicine, University of Chicago, Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Barthomeuf C. Inhibition of S1P-induced angiogenesis, metastasis and inflammation by dietary polyphenols. Free Radic Biol Med 2007; 42:312-3. [PMID: 17189837 DOI: 10.1016/j.freeradbiomed.2006.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2006] [Accepted: 11/02/2006] [Indexed: 12/01/2022]
|
12
|
Kim KS, Sengupta S, Berk M, Kwak YG, Escobar PF, Belinson J, Mok SC, Xu Y. Hypoxia Enhances Lysophosphatidic Acid Responsiveness in Ovarian Cancer Cells and Lysophosphatidic Acid Induces Ovarian Tumor MetastasisIn vivo. Cancer Res 2006; 66:7983-90. [PMID: 16912173 DOI: 10.1158/0008-5472.can-05-4381] [Citation(s) in RCA: 118] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Lysophosphatidic acid (LPA) is elevated in ascites of ovarian cancer patients and stimulates growth and other activities of ovarian cancer cells in vitro. Tissue hypoxia is a critical factor for tumor aggressiveness and metastasis in cancers. We tested whether the ascites of ovarian cancer is hypoxic and whether hypoxia influences the effects of LPA on ovarian cancer cells. We found that ovarian ascitic fluids were hypoxic in vivo. Enhanced cellular responsiveness to LPA, including migration and/or invasion of ovarian cancer cells, was observed under hypoxic conditions. This enhancement could be completely blocked by geldanamycin or a small interfering RNA targeting hypoxia-inducible factor 1 alpha (HIF1 alpha). LPA-induced cell migration required cytosolic phospholipase A(2) (cPLA(2)) and LPA stimulates cPLA(2) phosphorylation in a HIF1 alpha-dependent manner under hypoxia conditions. Furthermore, we show for the first time that exogenous LPA enhances tumor metastasis in an orthotopic ovarian cancer model and HIF alpha expression in tumors. 17-Dimethylaminoethylamino-17-demethoxygeldanamycin (an inhibitor of the heat shock protein 90) effectively blocked LPA-induced tumor metastasis in vivo. Together, our data indicate that hypoxic conditions are likely to be pathologically important for ovarian cancer development. HIF1 alpha plays a critical role in enhancing and/or sensitizing the role of LPA on cell migration and invasion under hypoxic conditions, where cPLA(2) is required for LPA-induced cell migration.
Collapse
Affiliation(s)
- Kwan-Sik Kim
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Hemmings DG. Signal transduction underlying the vascular effects of sphingosine 1-phosphate and sphingosylphosphorylcholine. Naunyn Schmiedebergs Arch Pharmacol 2006; 373:18-29. [PMID: 16570136 DOI: 10.1007/s00210-006-0046-5] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Two related lysosphingolipids, sphingosine 1-phosphate (S1P) and sphingosylphosphorylcholine (SPC) mediate diverse cellular responses through signals transduced by either activation of G-protein coupled receptors or possibly by acting intracellularly. Vascular responses to S1P and SPC measured both in vivo and in dissected vessels show predominantly vasoconstriction with some evidence for vasodilation. Although stimulation with S1P or SPC generally leads to similar vascular responses, the signalling pathways stimulated to produce these responses are often distinct. Nevertheless, mobilization of Ca2+ from intracellular stores and influx of extracellular Ca2+, which both increase [Ca2+]i, occur in response to S1P and SPC. Both mobilization of Ca2+ from intracellular stores and influx of extracellular Ca2+ occur in response to S1P and SPC. As well, both S1P and SPC induce Ca2+-sensitization in vascular smooth muscle which is mediated through Rho kinase activation. In the endothelium, S1P and SPC stimulate the production of the vasodilator, nitric oxide through activation of endothelial nitric oxide synthase. This activation occurs through phosphorylation by Akt and through binding of Ca2+-calmodulin upon increased [Ca2+]i. These lysosphingolipids also activate cyclooxygenase-2 which produces prostaglandins with both vasoconstrictor and vasodilator properties. A balance between the signals inducing vasodilation versus the signals inducing vasoconstriction will determine the vascular outcome. Thus, perturbations in S1P and SPC concentrations, relative expression of receptors or downstream signalling pathways may provide a mechanism for pathophysiological conditions such as hypertension. Given this background, recent studies examining a potential role for S1P and SPC in hypertension and vascular dysfunction in aging are discussed.
Collapse
Affiliation(s)
- Denise G Hemmings
- Department Obstetrics and Gynecology, Perinatal Research Centre, University of Alberta, 227 Heritage Medical Research Center, T6G 2S2, Edmonton, Alberta, Canada.
| |
Collapse
|
14
|
Czyborra C, Bischoff A, Michel MC. Indomethacin differentiates the renal effects of sphingosine-1-phosphate and sphingosylphosphorylcholine. Naunyn Schmiedebergs Arch Pharmacol 2006; 373:37-44. [PMID: 16521006 DOI: 10.1007/s00210-006-0037-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 01/10/2006] [Indexed: 12/26/2022]
Abstract
The sphingomyelin breakdown products sphingosine-1-phosphate (S1P) and sphingosylphosphorylcholine (SPC) constrict intrarenal microvessels in vitro in a pertussis toxin (PTX) sensitive manner, and S1P also reduces renal blood flow in vivo. Nevertheless, both S1P and SPC have been reported to enhance diuresis and natriuresis. This pattern is similar to that of neuropeptide Y, which also reduces renal blood flow and enhances diuresis and natriuresis. The latter effects are inhibited by the cyclooxygenase inhibitor indomethacin, and various S1P and SPC responses have also been linked to the cyclooxygenase pathway. Therefore, we have investigated whether indomethacin can alter the renal effects of S1P and SPC in anaesthetised rats in vivo. In line with earlier experiments S1P bolus injections dose-dependently reduced renal blood flow (by up to 4.8 +/- 0.5 ml min(-1)), and this was not significantly affected by indomethacin treatment (5 mg kg(-1) i.p.). Infusion of S1P but not of SPC (30 microg kg(-1) min(-1) each) for 60 min reduced renal blood flow by up to 0.8 +/- 0.2 ml min(-1), and this was not markedly altered by indomethacin. Despite the differential renovascular effect, both S1P and SPC enhanced diuresis by up to 215 +/- 65 and 201 +/- 58 microl 15 min(-1) respectively, and natriuresis by up to 25 +/- 9 and 29 +/- 11 micromol 15 min(-1) respectively. While indomethacin abolished the SPC-induced diuresis and natriuresis, it, if anything, slightly enhanced the diuretic and natriuretic effect of S1P. To determine whether tubular SPC effects are receptor-mediated, PTX experiments were performed. SPC-induced enhancements of diuresis and natriuresis were abolished by PTX. We conclude that S1P, SPC and neuropeptide Y exhibit distinct patterns of modulation of renal function and that indomethacin allows such effects to be differentiated.
Collapse
Affiliation(s)
- Claudia Czyborra
- Department of Medicine, University of Duisburg-Essen, Essen, Germany
| | | | | |
Collapse
|
15
|
Ozbay T, Rowan A, Leon A, Patel P, Sewer MB. Cyclic adenosine 5'-monophosphate-dependent sphingosine-1-phosphate biosynthesis induces human CYP17 gene transcription by activating cleavage of sterol regulatory element binding protein 1. Endocrinology 2006; 147:1427-37. [PMID: 16306078 DOI: 10.1210/en.2005-1091] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In the human adrenal cortex, ACTH activates steroid hormone biosynthesis by acutely increasing cholesterol delivery to the mitochondrion and chronically increasing the transcription of steroidogenic genes (including CYP17) via a cAMP-dependent pathway. In the present study, we characterized the role of sphingolipids in ACTH-dependent steroidogenesis. H295R human adrenocortical cells were treated with ACTH or dibutyryl cAMP (Bt2cAMP) and the content of several sphingolipid species quantified by mass spectrometry. Both ACTH and Bt2cAMP decreased cellular amounts of several sphingolipids, including sphingomyelin, ceramides, and sphingosine and stimulating the activity of sphingosine kinase and increasing the release of sphingosine-1-phosphate (S1P) into the media. S1P increased CYP17 mRNA expression by promoting the cleavage and nuclear localization of sterol regulatory element binding protein (SREBP) 1. Chromatin immunoprecipitation assays revealed that Bt2cAMP and S1P increased acetylation of histone H3 and promoted binding of SREBP1 to the -520/-331 region of the CYP17 promoter. In summary, our studies demonstrate a role for sphingolipid metabolism and SREBP1 in ACTH-dependent CYP17 regulation and steroidogenesis.
Collapse
Affiliation(s)
- Tuba Ozbay
- School of Biology, Georgia Institute of Technology, 310 Ferst Drive, Atlanta, Georgia 30332-0230, USA
| | | | | | | | | |
Collapse
|
16
|
Barthomeuf C, Lamy S, Blanchette M, Boivin D, Gingras D, Béliveau R. Inhibition of sphingosine-1-phosphate- and vascular endothelial growth factor-induced endothelial cell chemotaxis by red grape skin polyphenols correlates with a decrease in early platelet-activating factor synthesis. Free Radic Biol Med 2006; 40:581-90. [PMID: 16458188 DOI: 10.1016/j.freeradbiomed.2005.09.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2005] [Revised: 08/25/2005] [Accepted: 09/09/2005] [Indexed: 11/20/2022]
Abstract
Vascular endothelial growth factor (VEGF) and platelet-derived lipid sphingosine-1-phosphate (S1P) are two proinflammatory mediators which contribute to angiogenesis, in part through the synthesis of platelet-activating factor (PAF). The red grape skin polyphenolic extract (SGE) both prevents and inhibits angiogenesis in the Matrigel model, decreases the basal motility of endothelial and cancer cells, and reverses the chemotactic effect of S1P and VEGF on bovine aortic endothelial cells (BAECs) as well as the chemotactic effect of conditioned medium on human HT-1080 fibrosarcoma, human U-87 glioblastoma, and human DAOY medulloblastoma cells. Inhibition of VEGF- and S1P-mediated chemotaxis by SGE is associated with a down-regulation of ERK and p38/MAPK phosphorylation and a decreased in acute PAF synthesis. Notably, as do extracellular inhibitors of PAF receptor, SGE prevents S1P-induced PAF synthesis and the resulting activation of the S1P/endothelial differentiation gene-1 cascade. Given the key role of VEGF and S1P in inflammation, angiogenesis, and tumor invasion, SGE may therefore contribute to prevent (or to delay) the development of diseases associated with angiogenesis dysregulation, including cancer. The dual inhibition of S1P- and VEGF-mediated migration of endothelial cell and of serum-stimulated migration of U-87 cells suggests a usefulness of SGE against highly invasive human glioblastoma.
Collapse
Affiliation(s)
- Chantal Barthomeuf
- INSERM U-484, Laboratoire de Pharmacognosie et Biotechnologies, Université d'Auvergne, Faculté de Pharmacie, Place H. Dunant, 63001 Clermont-Fd, France.
| | | | | | | | | | | |
Collapse
|
17
|
Bartolmäs T, Heyn T, Mickeleit M, Fischer A, Reutter W, Danker K. Glucosamine-glycerophospholipids that activate cell-matrix adhesion and migration. J Med Chem 2005; 48:6750-5. [PMID: 16220990 DOI: 10.1021/jm050558n] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Two new analogues derived from the platelet activating factor (PAF), containing glucosamine instead of the acetyl group, were synthesized, and their effect on the human keratinocyte cell line HaCaT was evaluated with respect to cytotoxicity, proliferation, adhesion, and migration. Starting with (R)-1,2-isopropylideneglycerol (3), the glycosylation acceptor 1-O-octadecyl-3-O-tert-butyldimethylsilyl-sn-glycerol (6) was synthesized in three steps. Glycosylation of 6 with the already known O-(3,4,6-tri-O-acetyl-2-deoxy-2-dimethylmaleimido-beta-D-glycopyranosyl)trichloracetimidate gave 1-O-octadecyl-2-O-(3',4',6'-tri-O-acetyl-2'-deoxy-2'-dimethylmaleimido-beta-D-glucopyranosyl)-3-O-tert-butyldimethylsilyl-sn-glycerol (7). After removing the (tert-butyldimethyl)silyl (TBDMS) group with FeCl3x6H2O, phosphoryl choline was introduced, yielding [1-O-octadecyl-2-O-(2'-deoxy-2'-dimethylmaleimido-beta-D-glucopyranosyl)-sn-glycero(3)]phosphorylcholine (2) (glucosimide-PAF). pH controlled cleavage of the amino protection group gave [1-O-octadecyl-2-O-(2'-deoxy-2'-amino-beta-D-glucopyranosyl)-sn-glycero(3)]phosphorylcholine hydrochloride (1) (glucosamine-PAF). 2 inhibited proliferation of HaCaT cells by 26% at nontoxic concentrations, while 1 increased the proliferation rate by 30% at low concentrations. At higher concentrations, both compounds showed cytotoxic properties with LD50 = 30 micromol/L (1) and LD50 = 5-6 micromol/L (2). Both 1 and 2 were potent promoters of cell adhesion and migration of HaCaT cells.
Collapse
Affiliation(s)
- Thilo Bartolmäs
- Institut für Molekularbiologie und Biochemie, Campus Benjamin Franklin, Charité-Universitaetsmedizin Berlin, Arnimallee 22, D-14195 Berlin-Dahlem, Germany
| | | | | | | | | | | |
Collapse
|
18
|
Schmitz G, Heimerl S, Langmann T. Zinc finger protein ZNF202 structure and function in transcriptional control of HDL metabolism. Curr Opin Lipidol 2004; 15:199-208. [PMID: 15017363 DOI: 10.1097/00041433-200404000-00013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW The zinc finger protein ZNF202 is a transcriptional repressor controlling promoter elements predominantly found in genes involved in lipid metabolism and energy homeostasis. Here we summarize the structure, regulation and modulation of ZNF202 function by protein interactions. RECENT FINDINGS We review recent data and discuss the importance of the steadily growing list of ZNF202 target genes, defining a central role for ZNF202 as a key transcriptional regulator in metabolic disorders. Furthermore, we provide an interlink between transcriptional repression by ZNF202 and enhancement of gene activation via nuclear receptor coactivation by SCAN domain protein 1. SUMMARY The novel findings suggest that ZNF202 together with other SCAN domain proteins orchestrates a complex transcriptional regulatory network, which justifies a further exploration of its potential as a therapeutic target in lipid disorders such as atherosclerosis and associated metabolic syndromes.
Collapse
Affiliation(s)
- Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, University of Regensburg, Regensburg, Germany.
| | | | | |
Collapse
|
19
|
|