1
|
Karam M, Auclair C. Sphingosine-1-Phosphate as Lung and Cardiac Vasculature Protecting Agent in SARS-CoV-2 Infection. Int J Mol Sci 2023; 24:13088. [PMID: 37685894 PMCID: PMC10488186 DOI: 10.3390/ijms241713088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 08/16/2023] [Accepted: 08/19/2023] [Indexed: 09/10/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) may cause severe respiratory illness with high mortality. SARS-CoV-2 infection results in a massive inflammatory cell infiltration into the infected lungs accompanied by excessive pro-inflammatory cytokine production. The lung histology of dead patients shows that some areas are severely emphysematous, with enormously dilated blood vessels and micro-thromboses. The inappropriate inflammatory response damaging the pulmonary interstitial arteriolar walls suggests that the respiratory distress may come in a large part from lung vasculature injuries. It has been recently observed that low plasmatic sphingosine-1-phosphate (S1P) is a marker of a worse prognosis of clinical outcome in severe coronavirus disease (COVID) patients. S1P is an angiogenic molecule displaying anti-inflammatory and anti-apoptotic properties, that promote intercellular interactions between endothelial cells and pericytes resulting in the stabilization of arteries and capillaries. In this context, it can be hypothesized that the benefit of a normal S1P level is due to its protective effect on lung vasculature functionality. This paper provides evidence supporting this concept, opening the way for the design of a pharmacological approach involving the use of an S1P lyase inhibitor to increase the S1P level that in turn will rescue the lung vasculature functionality.
Collapse
Affiliation(s)
| | - Christian Auclair
- AC BioTech, Villejuif Biopark, Cancer Campus, 1 mail du Professeur Georges Mathé, 94800 Villejuif, France;
| |
Collapse
|
2
|
Liu H, Li L, Chen Z, Song Y, Liu W, Gao G, Li L, Jiang J, Xu C, Yan G, Cui H. S1PR2 Inhibition Attenuates Allergic Asthma Possibly by Regulating Autophagy. Front Pharmacol 2021; 11:598007. [PMID: 33643037 PMCID: PMC7902893 DOI: 10.3389/fphar.2020.598007] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/30/2020] [Indexed: 11/18/2022] Open
Abstract
This study is to investigate the role of Sphingosine-1-phosphate (S1P) in the asthma progression, and the involvement of autophagy. Airway remodeling mice were subjected to the HE, PAS, and Masson staining. Protein expression levels in the tissues, samples and model cells were detected with ELISA, Western blot analysis, and immunohistochemical/immunofluorescent analysis. The S1P2 receptor antagonist JTE-013 decreased the inflammatory cell infiltration and goblet cell production in asthmatic mice tissues. The IL-1, IL-4, IL-5 and serum IgE contents were decreased in bronchoalveolar lavage fluid, while the Beclin1 expression in lung tissues was decreased. The LC3B1 to LC-3B2 conversion was decreased, with increased P62 accumulation and decreased p-P62 expression. In airway remodeling mice, JTE-013 significantly decreased collagen deposition in lung tissues and decreased smooth muscle cell smooth muscle activating protein expression. In lung tissue, the expression levels of Beclin1 were decreased, with decreased LC3B1 to LC-3B2 conversion, as well as the increased P62 accumulation and decreased p-P62 expression. However, these effects were reversed by the RAC1 inhibitor EHT 1864. Similar results were observed for the silencing of S1P2 receptor in the cells, as shown by the decreased Beclin1 expression, decreased LC3B1 to LC-3B2 conversion, increased P62 accumulation, and decreased p-P62 expression. The smooth muscle activators were significantly decreased in the JTE-013 and EHT1864 groups, and the EHT 1864 + S1P2-SiRNA expression level was increased. S1P is involved in the progression of asthma and airway remodeling, which may be related to the activation of S1PR2 receptor and inhibition of autophagy through RAC1.
Collapse
Affiliation(s)
- Hanye Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Pharmacology, Yanbian University College of Medicine, Yanji, China.,Center of Medical Functional Experiment, Yanbian University College of Medicine, Yanji, China
| | - Liangchang Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Zhengai Chen
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Pharmacology, Yanbian University College of Medicine, Yanji, China
| | - Yilan Song
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Weidong Liu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Center of Medical Functional Experiment, Yanbian University College of Medicine, Yanji, China
| | - Ge Gao
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Center of Medical Functional Experiment, Yanbian University College of Medicine, Yanji, China
| | - Li Li
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Jingzhi Jiang
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Chang Xu
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Guanghai Yan
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Department of Anatomy, Histology and Embryology, Yanbian University College of Medicine, Yanji, China
| | - Hong Cui
- Jilin Key Laboratory for Immune and Targeting Research on Common Allergic Diseases, Yanbian University, Yanji, China.,Center of Medical Functional Experiment, Yanbian University College of Medicine, Yanji, China
| |
Collapse
|
3
|
D'Aprile C, Prioni S, Mauri L, Prinetti A, Grassi S. Lipid rafts as platforms for sphingosine 1-phosphate metabolism and signalling. Cell Signal 2021; 80:109929. [PMID: 33493577 DOI: 10.1016/j.cellsig.2021.109929] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/06/2023]
Abstract
Spontaneous segregation of cholesterol and sphingolipids as a liquid-ordered phase leads to their clustering in selected membrane areas, the lipid rafts. These specialized membrane domains enriched in gangliosides, sphingomyelin, cholesterol and selected proteins involved in signal transduction, organize and determine the function of multiprotein complexes involved in several aspects of signal transduction, thus regulating cell homeostasis. Sphingosine 1-phosphate, an important biologically active mediator, is involved in several signal transduction processes regulating a plethora of cell functions and, not only several of its downstream effectors tend to localize in lipid rafts, some of the enzymes involved in its pathway, of receptors involved in its signalling and its transporters have been often found in these membrane microdomains. Considering this, in this review we address what is currently known regarding the relationship between sphingosine 1-phosphate metabolism and signalling and plasma membrane lipid rafts.
Collapse
Affiliation(s)
- Chiara D'Aprile
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
4
|
Fang T, Jiang YX, Chen L, Huang L, Tian XH, Zhou YD, Nagle DG, Zhang DD. Coix Seed Oil Exerts an Anti-Triple-Negative Breast Cancer Effect by Disrupting miR-205/S1PR1 Axis. Front Pharmacol 2020; 11:529962. [PMID: 33101013 PMCID: PMC7556270 DOI: 10.3389/fphar.2020.529962] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 09/04/2020] [Indexed: 12/13/2022] Open
Abstract
Coix Seed Oil (CSO) possesses a wide range of pharmacological activities. Kanglaite Injection, a commercial product of CSO, has been used clinically as an anticancer drug in China for decades. However, its molecular mechanisms on triple-negative breast cancer (TNBC) remains to be elucidated. In this study, the effect of CSO was evaluated on murine TNBC 4T1 cells and the orthotopic tumor-bearing mouse model and underlying mechanisms were explored. CSO suppressed cell proliferation, colony formation in vitro, and tumor growth in vivo. miR-205-5p was substantially altered in CSO treated tumor tissues compared to the control group by miRNA-sequencing analysis. Sphingomyelin metabolism (SM) decreased in serum in model group compared to the control group, while it increased by CSO administration by lipid metabolomics analysis. The expression of sphingosine 1 phosphate receptor 1 (S1PR1), the critical effector of SM, was downregulated upon CSO treatment. Mechanically, miRNA-205 directly targeted S1PR1 to regulate SM and cell proliferation. CSO reduced the expression of S1PR1, cyclinD1, and phosphorylation levels of STAT3, MAPK, and AKT while upregulated p27. These results revealed that CSO exerted an anti-TNBC effect via the miR-205/S1PR1 axis to regulate sphingomyelin metabolism, and the downstream STAT3/MAPK/AKT signal pathways were partly involved.
Collapse
Affiliation(s)
- Ting Fang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,School of Pharmacy, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yi-Xin Jiang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Long Chen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ling Huang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xin-Hui Tian
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu-Dong Zhou
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Chemistry and Biochemistry, College of Liberal Arts, University of Mississippi, University, Misissippi, MS, United States
| | - Dale G Nagle
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of BioMolecular Sciences and Research Institute of Pharmaceutical Sciences (RIPS), School of Pharmacy, University of Mississippi, University, Mississippi, MS, United States
| | - Dan-Dan Zhang
- Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
5
|
Figueiredo AM, Villacampa P, Diéguez-Hurtado R, José Lozano J, Kobialka P, Cortazar AR, Martinez-Romero A, Angulo-Urarte A, Franco CA, Claret M, Aransay AM, Adams RH, Carracedo A, Graupera M. Phosphoinositide 3-Kinase-Regulated Pericyte Maturation Governs Vascular Remodeling. Circulation 2020; 142:688-704. [PMID: 32466671 DOI: 10.1161/circulationaha.119.042354] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Pericytes regulate vessel stabilization and function, and their loss is associated with diseases such as diabetic retinopathy or cancer. Despite their physiological importance, pericyte function and molecular regulation during angiogenesis remain poorly understood. METHODS To decipher the transcriptomic programs of pericytes during angiogenesis, we crossed Pdgfrb(BAC)-CreERT2 mice into RiboTagflox/flox mice. Pericyte morphological changes were assessed in mural cell-specific R26-mTmG reporter mice, in which low doses of tamoxifen allowed labeling of single-cell pericytes at high resolution. To study the role of phosphoinositide 3-kinase (PI3K) signaling in pericyte biology during angiogenesis, we used genetic mouse models that allow selective inactivation of PI3Kα and PI3Kβ isoforms and their negative regulator phosphate and tensin homolog deleted on chromosome 10 (PTEN) in mural cells. RESULTS At the onset of angiogenesis, pericytes exhibit molecular traits of cell proliferation and activated PI3K signaling, whereas during vascular remodeling, pericytes upregulate genes involved in mature pericyte cell function, together with a remarkable decrease in PI3K signaling. Immature pericytes showed stellate shape and high proliferation, and mature pericytes were quiescent and elongated. Unexpectedly, we demonstrate that PI3Kβ, but not PI3Kα, regulates pericyte proliferation and maturation during vessel formation. Genetic PI3Kβ inactivation in pericytes triggered early pericyte maturation. Conversely, unleashing PI3K signaling by means of PTEN deletion delayed pericyte maturation. Pericyte maturation was necessary to undergo vessel remodeling during angiogenesis. CONCLUSIONS Our results identify new molecular and morphological traits associated with pericyte maturation and uncover PI3Kβ activity as a checkpoint to ensure appropriate vessel formation. In turn, our results may open new therapeutic opportunities to regulate angiogenesis in pathological processes through the manipulation of pericyte PI3Kβ activity.
Collapse
Affiliation(s)
- Ana M Figueiredo
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Pilar Villacampa
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Rodrigo Diéguez-Hurtado
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, Germany (R.D.-H., R.H.A.)
| | - Juan José Lozano
- Bioinformatics Platform, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Barcelona, Spain (J.J.L.)
| | - Piotr Kobialka
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Ana Rosa Cortazar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain (A.R.C., A.M.A., A.C.)
| | - Anabel Martinez-Romero
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Ana Angulo-Urarte
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| | - Claudio A Franco
- CIBERONC (A.R.C., A.M.A., A.C., M.G.) and CIBERehd (A.M.A.), Instituto de Salud Carlos III, Madrid, Spain. Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal (C.A.F.)
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (M.C.)
| | - Ana María Aransay
- CIBERONC (A.R.C., A.M.A., A.C., M.G.) and CIBERehd (A.M.A.), Instituto de Salud Carlos III, Madrid, Spain. Instituto de Medicina Molecular-João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal (C.A.F.)
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, and Faculty of Medicine, University of Münster, Germany (R.D.-H., R.H.A.)
| | - Arkaitz Carracedo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Spain (A.R.C., A.M.A., A.C.)
| | - Mariona Graupera
- Vascular Biology and Signalling Group, ProCURE, Oncobell Program, Institut d´Investigació Biomèdica de Bellvitge (IDIBELL), Gran Via de l'Hospitalet 199, 08908 L´Hospitalet de Llobregat, Barcelona, Spain (A.M.F., P.V., P.K., A.M.-R., A.A.-U., M.G.)
| |
Collapse
|
6
|
Abstract
Sphingosine-1-phosphate (S1P) can regulate several physiological and pathological processes. S1P signaling via its cell surface receptor S1PR1 has been shown to enhance tumorigenesis and stimulate growth, expansion, angiogenesis, metastasis, and survival of cancer cells. S1PR1-mediated tumorigenesis is supported and amplified by activation of downstream effectors including STAT3, interleukin-6, and NF-κB networks. S1PR1 signaling can also trigger various other signaling pathways involved in carcinogenesis including activation of PI3K/AKT, MAPK/ERK1/2, Rac, and PKC/Ca, as well as suppression of cyclic adenosine monophosphate (cAMP). It also induces immunological tolerance in the tumor microenvironment, while the immunosuppressive function of S1PR1 can also lead to the generation of pre-metastatic niches. Some tumor cells upregulate S1PR1 signaling pathways, which leads to drug resistant cancer cells, mainly through activation of STAT3. This signaling pathway is also implicated in some inflammatory conditions leading to the instigation of inflammation-driven cancers. Furthermore, it can also increase survival via induction of anti-apoptotic pathways, for instance, in breast cancer cells. Therefore, S1PR1 and its signaling pathways can be considered as potential anti-tumor therapeutic targets, alone or in combination therapies. Given the oncogenic nature of S1PR1 and its distribution in a variety of cancer cell types along with its targeting advantages over other molecules of this family, S1PR1 should be considered a favorable target in therapeutic approaches to cancer. This review describes the role of S1PR1 in cancer development and progression, specifically addressing breast cancer, glioma, and hematopoietic malignancies. We also discuss the potential use of S1P signaling modulators as therapeutic targets in cancer therapy.
Collapse
|
7
|
S1P 1 receptor phosphorylation, internalization, and interaction with Rab proteins: effects of sphingosine 1-phosphate, FTY720-P, phorbol esters, and paroxetine. Biosci Rep 2018; 38:BSR20181612. [PMID: 30366961 PMCID: PMC6294635 DOI: 10.1042/bsr20181612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 10/19/2018] [Accepted: 10/26/2018] [Indexed: 01/04/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) and FTY720-phosphate (FTYp) increased intracellular calcium in cells expressing S1P1 mCherry-tagged receptors; the synthetic agonist was considerably less potent. Activation of protein kinase C by phorbol myristate acetate (PMA) blocked these effects. The three agents induced receptor phosphorylation and internalization, with the action of FTYp being more intense. S1P1 receptor–Rab protein (GFP-tagged) interaction was studied using FRET. The three agents were able to induce S1P1 receptor–Rab5 interaction, although with different time courses. S1P1 receptor–Rab9 interaction was mainly increased by the phorbol ester, whereas S1P1 receptor–Rab7 interaction was only increased by FTYp and after a 30-min incubation. These actions were not observed using dominant negative (GDP-bound) Rab protein mutants. The data suggested that the three agents induce interaction with early endosomes, but that the natural agonist induced rapid receptor recycling, whereas activation of protein kinase C favored interaction with late endosome and slow recycling and FTYp triggered receptor interaction with vesicles associated with proteasomal/lysosomal degradation. The ability of bisindolylmaleimide I and paroxetine to block some of these actions suggested the activation of protein kinase C was associated mainly with the action of PMA, whereas G protein-coupled receptor kinase (GRK) 2 (GRK2) was involved in the action of the three agents.
Collapse
|
8
|
van Gastel J, Hendrickx JO, Leysen H, Santos-Otte P, Luttrell LM, Martin B, Maudsley S. β-Arrestin Based Receptor Signaling Paradigms: Potential Therapeutic Targets for Complex Age-Related Disorders. Front Pharmacol 2018; 9:1369. [PMID: 30546309 PMCID: PMC6280185 DOI: 10.3389/fphar.2018.01369] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022] Open
Abstract
G protein coupled receptors (GPCRs) were first characterized as signal transducers that elicit downstream effects through modulation of guanine (G) nucleotide-binding proteins. The pharmacotherapeutic exploitation of this signaling paradigm has created a drug-based field covering nearly 50% of the current pharmacopeia. Since the groundbreaking discoveries of the late 1990s to the present day, it is now clear however that GPCRs can also generate productive signaling cascades through the modulation of β-arrestin functionality. β-Arrestins were first thought to only regulate receptor desensitization and internalization - exemplified by the action of visual arrestin with respect to rhodopsin desensitization. Nearly 20 years ago, it was found that rather than controlling GPCR signal termination, productive β-arrestin dependent GPCR signaling paradigms were highly dependent on multi-protein complex formation and generated long-lasting cellular effects, in contrast to G protein signaling which is transient and functions through soluble second messenger systems. β-Arrestin signaling was then first shown to activate mitogen activated protein kinase signaling in a G protein-independent manner and eventually initiate protein transcription - thus controlling expression patterns of downstream proteins. While the possibility of developing β-arrestin biased or functionally selective ligands is now being investigated, no additional research has been performed on its possible contextual specificity in treating age-related disorders. The ability of β-arrestin-dependent signaling to control complex and multidimensional protein expression patterns makes this therapeutic strategy feasible, as treating complex age-related disorders will likely require therapeutics that can exert network-level efficacy profiles. It is our understanding that therapeutically targeting G protein-independent effectors such as β-arrestin will aid in the development of precision medicines with tailored efficacy profiles for disease/age-specific contextualities.
Collapse
Affiliation(s)
- Jaana van Gastel
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Jhana O Hendrickx
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Hanne Leysen
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| | - Paula Santos-Otte
- Institute of Biophysics, Humboldt University of Berlin, Berlin, Germany
| | - Louis M Luttrell
- Division of Endocrinology, Diabetes and Medical Genetics, Medical University of South Carolina, Charleston, SC, United States
| | - Bronwen Martin
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.,Translational Neurobiology Group, Centre for Molecular Neuroscience, VIB, Antwerp, Belgium
| |
Collapse
|
9
|
Ng ML, Yarla NS, Menschikowski M, Sukocheva OA. Regulatory role of sphingosine kinase and sphingosine-1-phosphate receptor signaling in progenitor/stem cells. World J Stem Cells 2018; 10:119-133. [PMID: 30310531 PMCID: PMC6177561 DOI: 10.4252/wjsc.v10.i9.119] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 07/27/2018] [Accepted: 08/05/2018] [Indexed: 02/06/2023] Open
Abstract
Balanced sphingolipid signaling is important for the maintenance of homeostasis. Sphingolipids were demonstrated to function as structural components, second messengers, and regulators of cell growth and survival in normal and disease-affected tissues. Particularly, sphingosine kinase 1 (SphK1) and its product sphingosine-1-phosphate (S1P) operate as mediators and facilitators of proliferation-linked signaling. Unlimited proliferation (self-renewal) within the regulated environment is a hallmark of progenitor/stem cells that was recently associated with the S1P signaling network in vasculature, nervous, muscular, and immune systems. S1P was shown to regulate progenitor-related characteristics in normal and cancer stem cells (CSCs) via G-protein coupled receptors S1Pn (n = 1 to 5). The SphK/S1P axis is crucially involved in the regulation of embryonic development of vasculature and the nervous system, hematopoietic stem cell migration, regeneration of skeletal muscle, and development of multiple sclerosis. The ratio of the S1P receptor expression, localization, and specific S1P receptor-activated downstream effectors influenced the rate of self-renewal and should be further explored as regeneration-related targets. Considering malignant transformation, it is essential to control the level of self-renewal capacity. Proliferation of the progenitor cell should be synchronized with differentiation to provide healthy lifelong function of blood, immune systems, and replacement of damaged or dead cells. The differentiation-related role of SphK/S1P remains poorly assessed. A few pioneering investigations explored pharmacological tools that target sphingolipid signaling and can potentially confine and direct self-renewal towards normal differentiation. Further investigation is required to test the role of the SphK/S1P axis in regulation of self-renewal and differentiation.
Collapse
Affiliation(s)
- Mei Li Ng
- Centenary Institute of Cancer Medicine and Cell Biology, Sydney NSW 2050, Australia
| | - Nagendra S Yarla
- Department of Biochemistry and Bioinformatics, Institute of Science, GITAM University, Rushikonda, Visakhapatnam 530 045, Andhra Pradesh, India
| | - Mario Menschikowski
- Institute of Clinical Chemistry and Laboratory Medicine, Carl Gustav Carus University Hospital, Technical University of Dresden, Dresden D-01307, Germany
| | - Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park SA 5042, Australia.
| |
Collapse
|
10
|
Patmanathan SN, Wang W, Yap LF, Herr DR, Paterson IC. Mechanisms of sphingosine 1-phosphate receptor signalling in cancer. Cell Signal 2017; 34:66-75. [PMID: 28302566 DOI: 10.1016/j.cellsig.2017.03.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 03/11/2017] [Accepted: 03/11/2017] [Indexed: 12/12/2022]
Abstract
S1P is a small bioactive lipid which exerts its effects following binding to a family of five G protein-coupled receptors, known as S1P1-5. Following receptor activation, multiple signalling cascades are activated, allowing S1P to regulate a range of cellular processes, such as proliferation, apoptosis, migration and angiogenesis. There is strong evidence implicating the involvement of S1P receptors (S1PRs) in cancer progression and the oncogenic effects of S1P can result from alterations in the expression of one or more of the S1PRs and/or the enzymes that regulate the levels of S1P. However, cooperativity between the individual S1PRs, functional interactions with receptor tyrosine kinases and the sub-cellular localisation of the S1PRs within tumour cells also appear to play a role in mediating the effects of S1PR signalling during carcinogenesis. Here we review what is known regarding the role of individual S1PRs in cancer and discuss the recent evidence to suggest cross-talk between the S1PRs and other cellular signalling pathways in cancer. We will also discuss the therapeutic potential of targeting the S1PRs and their downstream signalling pathways for the treatment of cancer.
Collapse
Affiliation(s)
- Sathya Narayanan Patmanathan
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Wei Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Lee Fah Yap
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117456, Singapore
| | - Ian C Paterson
- Department of Oral and Craniofacial Sciences, Oral Cancer Research & Coordinating Centre, Faculty of Dentistry, University of Malaya, 50603 Kuala Lumpur, Malaysia.
| |
Collapse
|
11
|
Pyne NJ, Pyne S. Sphingosine 1-Phosphate Receptor 1 Signaling in Mammalian Cells. Molecules 2017; 22:molecules22030344. [PMID: 28241498 PMCID: PMC6155263 DOI: 10.3390/molecules22030344] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/15/2017] [Accepted: 02/18/2017] [Indexed: 12/16/2022] Open
Abstract
The bioactive lipid, sphingosine 1-phosphate (S1P) binds to a family of G protein-coupled receptors, termed S1P1-S1P5. These receptors function in, for example, the cardiovascular system to regulate vascular barrier integrity and tone, the nervous system to regulate neuronal differentiation, myelination and oligodendrocyte/glial cell survival and the immune system to regulate T- and B-cell subsets and trafficking. S1P receptors also participate in the pathophysiology of autoimmunity, inflammatory disease, cancer, neurodegeneration and others. In this review, we describe how S1P1 can form a complex with G-protein and β-arrestin, which function together to regulate effector pathways. We also discuss the role of the S1P1-Platelet derived growth factor receptor β functional complex (which deploys G-protein/β-arrestin and receptor tyrosine kinase signaling) in regulating cell migration. Possible mechanisms by which different S1P-chaperones, such as Apolipoprotein M-High-Density Lipoprotein induce biological programmes in cells are also described. Finally, the role of S1P1 in health and disease and as a target for clinical intervention is appraised.
Collapse
Affiliation(s)
- Nigel J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| | - Susan Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow G4 0RE, UK.
| |
Collapse
|
12
|
Beider K, Rosenberg E, Bitner H, Shimoni A, Leiba M, Koren-Michowitz M, Ribakovsky E, Klein S, Olam D, Weiss L, Wald H, Abraham M, Galun E, Peled A, Nagler A. The Sphingosine-1-Phosphate Modulator FTY720 Targets Multiple Myeloma via the CXCR4/CXCL12 Pathway. Clin Cancer Res 2016; 23:1733-1747. [PMID: 27697999 DOI: 10.1158/1078-0432.ccr-15-2618] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Revised: 07/20/2016] [Accepted: 09/01/2016] [Indexed: 11/16/2022]
Abstract
Purpose: To explore the functional consequences of possible cross-talk between the CXCR4/CXCL12 and the sphingosine-1-phosphate (S1P) pathways in multiple myeloma (MM) cells and to evaluate the effect of S1P targeting with the FTY720 modulator as a potential anti-MM therapeutic strategy.Experimental Design and Results: S1P targeting with FTY720 induces MM cell apoptosis. The combination of FTY720 with the SPHK1 inhibitor SKI-II results in synergistic inhibition of MM growth. CXCR4/CXCL12-enhanced expression correlates with reduced MM cell sensitivity to both FTY720 and SKI-II inhibitors, and with SPHK1 coexpression in both cell lines and primary MM bone marrow (BM) samples, suggesting regulative cross-talk between the CXCR4/CXCL12 and SPHK1 pathways in MM cells. FTY720 was found to directly target CXCR4. FTY720 profoundly reduces CXCR4 cell-surface levels and abrogates the CXCR4-mediated functions of migration toward CXCL12 and signaling pathway activation. Moreover, FTY720 cooperates with bortezomib, inducing its cytotoxic activity and abrogating the bortezomib-mediated increase in CXCR4 expression. FTY720 effectively targets bortezomib-resistant cells and increases their sensitivity to bortezomib, promoting DNA damage. Finally, in a recently developed novel xenograft model of CXCR4-dependent systemic MM with BM involvement, FTY720 treatment effectively reduces tumor burden in the BM of MM-bearing mice. FTY720 in combination with bortezomib demonstrates superior tumor growth inhibition and abrogates bortezomib-induced CXCR4 increase on MM cells.Conclusions: Altogether, our work identifies a cross-talk between the S1P and CXCR4 pathways in MM cells and provides a preclinical rationale for the therapeutic application of FTY720 in combination with bortezomib in patients with MM. Clin Cancer Res; 23(7); 1733-47. ©2016 AACR.
Collapse
Affiliation(s)
- Katia Beider
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Evgenia Rosenberg
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Hanna Bitner
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Avichai Shimoni
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Merav Leiba
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Maya Koren-Michowitz
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Elena Ribakovsky
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel
| | - Shiri Klein
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Devorah Olam
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Hanna Wald
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Michal Abraham
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Eithan Galun
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Amnon Peled
- Goldyne Savad Institute of Gene Therapy, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Arnon Nagler
- Hematology Division and CBB, Guy Weinshtock Multiple Myeloma Foundation, Chaim Sheba Medical Center, Tel-Hashomer, Israel.
| |
Collapse
|
13
|
Syrovatkina V, Alegre KO, Dey R, Huang XY. Regulation, Signaling, and Physiological Functions of G-Proteins. J Mol Biol 2016; 428:3850-68. [PMID: 27515397 DOI: 10.1016/j.jmb.2016.08.002] [Citation(s) in RCA: 306] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/31/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022]
Abstract
Heterotrimeric guanine-nucleotide-binding regulatory proteins (G-proteins) mainly relay the information from G-protein-coupled receptors (GPCRs) on the plasma membrane to the inside of cells to regulate various biochemical functions. Depending on the targeted cell types, tissues, and organs, these signals modulate diverse physiological functions. The basic schemes of heterotrimeric G-proteins have been outlined. In this review, we briefly summarize what is known about the regulation, signaling, and physiological functions of G-proteins. We then focus on a few less explored areas such as the regulation of G-proteins by non-GPCRs and the physiological functions of G-proteins that cannot be easily explained by the known G-protein signaling pathways. There are new signaling pathways and physiological functions for G-proteins to be discovered and further interrogated. With the advancements in structural and computational biological techniques, we are closer to having a better understanding of how G-proteins are regulated and of the specificity of G-protein interactions with their regulators.
Collapse
Affiliation(s)
- Viktoriya Syrovatkina
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Kamela O Alegre
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Raja Dey
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA
| | - Xin-Yun Huang
- Department of Physiology and Biophysics, Weill Cornell Medical College, of Cornell University, 1300 York Avenue, New York, NY 10065, USA.
| |
Collapse
|
14
|
Sysol JR, Natarajan V, Machado RF. PDGF induces SphK1 expression via Egr-1 to promote pulmonary artery smooth muscle cell proliferation. Am J Physiol Cell Physiol 2016; 310:C983-92. [PMID: 27099350 DOI: 10.1152/ajpcell.00059.2016] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/15/2016] [Indexed: 12/20/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive, life-threatening disease for which there is currently no curative treatment available. Pathologic changes in this disease involve remodeling of the pulmonary vasculature, including marked proliferation of pulmonary artery smooth muscle cells (PASMCs). Recently, the bioactive lipid sphingosine-1-phosphate (S1P) and its activating kinase, sphingosine kinase 1 (SphK1), have been shown to be upregulated in PAH and promote PASMC proliferation. The mechanisms regulating the transcriptional upregulation of SphK1 in PASMCs are unknown. In this study, we investigated the role of platelet-derived growth factor (PDGF), a PAH-relevant stimuli associated with enhanced PASMC proliferation, on SphK1 expression regulation. In human PASMCs (hPASMCs), PDGF significantly increased SphK1 mRNA and protein expression and induced cell proliferation. Selective inhibition of SphK1 attenuated PDGF-induced hPASMC proliferation. In silico promoter analysis for SphK1 identified several binding sites for early growth response protein 1 (Egr-1), a PDGF-associated transcription factor. Luciferase assays demonstrated that PDGF activates the SphK1 promoter in hPASMCs, and truncation of the 5'-promoter reduced PDGF-induced SphK1 expression. Stimulation of hPASMCs with PDGF induced Egr-1 protein expression, and direct binding of Egr-1 to the SphK1 promoter was confirmed by chromatin immunoprecipitation analysis. Inhibition of ERK signaling prevented induction of Egr-1 by PDGF. Silencing of Egr-1 attenuated PDGF-induced SphK1 expression and hPASMC proliferation. These studies demonstrate that SphK1 is regulated by PDGF in hPASMCs via the transcription factor Egr-1, promoting cell proliferation. This novel mechanism of SphK1 regulation may be a therapeutic target in pulmonary vascular remodeling in PAH.
Collapse
Affiliation(s)
- Justin R Sysol
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; and Medical Scientist Training Program, University of Illinois at Chicago, Chicago, Illinois
| | - Viswanathan Natarajan
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; and
| | - Roberto F Machado
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; Department of Pharmacology, Division of Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
15
|
Reeves PM, Kang YL, Kirchhausen T. Endocytosis of Ligand-Activated Sphingosine 1-Phosphate Receptor 1 Mediated by the Clathrin-Pathway. Traffic 2015; 17:40-52. [PMID: 26481905 DOI: 10.1111/tra.12343] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2013] [Revised: 10/16/2015] [Accepted: 10/16/2015] [Indexed: 12/31/2022]
Abstract
The sphingosine 1-phosphate receptor 1 (S1PR1) is one of five G protein-coupled receptors activated by the lipid sphingosine 1-phosphate (S1P). Stimulation of S1PR1 by binding S1P or the synthetic agonist FTY720P results in rapid desensitization, associated in part with depletion of receptor from the cell surface. We report here combining spinning disc confocal fluorescence microscopy and flow cytometry to show that rapid internalization of activated S1PR1 relies on a functional clathrin-mediated endocytic pathway. Uptake of activated S1PR1 was strongly inhibited in cells disrupted in their clathrin-mediated endocytosis by depleting clathrin or AP-2 or by treating cells with dynasore-OH. The uptake of activated S1P1R was strongly inhibited in cells lacking both β-arrestin 1 and β-arrestin 2, indicating that activated S1PR1 follows the canonical route of endocytosis for G-protein coupled receptor's (GPCR)'s.
Collapse
Affiliation(s)
- Patrick M Reeves
- Department of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, MA, USA
| | - Yuan-Lin Kang
- Department of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, MA, USA
| | - Tom Kirchhausen
- Department of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine at Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
16
|
Sukocheva O, Wadham C, Gamble J, Xia P. Sphingosine-1-phosphate receptor 1 transmits estrogens' effects in endothelial cells. Steroids 2015; 104:237-245. [PMID: 26476183 DOI: 10.1016/j.steroids.2015.10.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 09/29/2015] [Accepted: 10/11/2015] [Indexed: 02/08/2023]
Abstract
We have previously reported that the steroid hormone estrogens stimulate activation of sphingosine kinase 1 (SphK1) and sphingosine-1-phosphate (S1P) receptors in breast cancer cells. Both estrogens and S1P are potent biological modulators of endothelial function in vasculature able to activate multiple effectors, including endothelial nitric oxide synthase (eNOS). In this study we report that treatment of endothelial cells (ECs) with 17β-estradiol (E2) resulted in a rapid, transient, and dose-dependent increase in SphK activity and increased S1P production. The effect was not reproduced by the inactive E2 analogue 17α-E2. Expression of the dominant-negative mutant SphK1(G82D) or transfection of SphK1-targeted siRNA in ECs caused not only a defect in SphK activation by E2, but also a significant inhibition of E2-induced activation of Akt/eNOS. Furthermore, E2 treatment induced internalization of plasma membrane S1P1 receptor, accompanied with an increase in the amount of cytosolic S1P1. By down-regulating S1P1 receptor expression, the S1P1-specific antisense oligonucleotides significantly inhibited E2-induced activation of Akt/eNOS in ECs. E2-induced EC migration and tube formation were also inhibited by S1P1 down-regulation. Thus, the findings indicate an important role of the SphK1/S1P1 pathway in mediating estrogen signaling and its actions in vasculature.
Collapse
Affiliation(s)
- Olga Sukocheva
- School of Health Sciences, Flinders University, SA, Australia.
| | | | | | - Pu Xia
- Department of Endocrinology, Zhongsan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
17
|
Imeri F, Blanchard O, Jenni A, Schwalm S, Wünsche C, Zivkovic A, Stark H, Pfeilschifter J, Huwiler A. FTY720 and two novel butterfly derivatives exert a general anti-inflammatory potential by reducing immune cell adhesion to endothelial cells through activation of S1P(3) and phosphoinositide 3-kinase. Naunyn Schmiedebergs Arch Pharmacol 2015; 388:1283-92. [PMID: 26267293 DOI: 10.1007/s00210-015-1159-5] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 07/27/2015] [Indexed: 10/23/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a key lipid regulator of a variety of cellular responses including cell proliferation and survival, cell migration, and inflammatory reactions. Here, we investigated the effect of S1P receptor activation on immune cell adhesion to endothelial cells under inflammatory conditions. We show that S1P reduces both tumor necrosis factor (TNF)-α- and lipopolysaccharide (LPS)-stimulated adhesion of Jurkat and U937 cells to an endothelial monolayer. The reducing effect of S1P was reversed by the S1P1+3 antagonist VPC23019 but not by the S1P1 antagonist W146. Additionally, knockdown of S1P3, but not S1P1, by short hairpin RNA (shRNA) abolished the reducing effect of S1P, suggesting the involvement of S1P3. A suppression of immune cell adhesion was also seen with the immunomodulatory drug FTY720 and two novel butterfly derivatives ST-968 and ST-1071. On the molecular level, S1P and all FTY720 derivatives reduced the mRNA expression of LPS- and TNF-α-induced adhesion molecules including ICAM-1, VCAM-1, E-selectin, and CD44 which was reversed by the PI3K inhibitor LY294002, but not by the MEK inhibitor U0126.In summary, our data demonstrate a novel molecular mechanism by which S1P, FTY720, and two novel butterfly derivatives acted anti-inflammatory that is by suppressing gene transcription of various endothelial adhesion molecules and thereby preventing adhesion of immune cells to endothelial cells and subsequent extravasation.
Collapse
Affiliation(s)
- Faik Imeri
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Olivier Blanchard
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Aurelio Jenni
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland
| | - Stephanie Schwalm
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Christin Wünsche
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.,Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Aleksandra Zivkovic
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine-University Düsseldorf, Universitätsstr. 1, 40225, Düsseldorf, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Frankfurt am Main, Germany
| | - Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital, INO-F, CH-3010, Bern, Switzerland.
| |
Collapse
|
18
|
Kato H, Nishitoh H. Stress responses from the endoplasmic reticulum in cancer. Front Oncol 2015; 5:93. [PMID: 25941664 PMCID: PMC4403295 DOI: 10.3389/fonc.2015.00093] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/31/2015] [Indexed: 12/21/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic organelle that is essential for multiple cellular functions. During cellular stress conditions, including nutrient deprivation and dysregulation of protein synthesis, unfolded/misfolded proteins accumulate in the ER lumen, resulting in activation of the unfolded protein response (UPR). The UPR also contributes to the regulation of various intracellular signaling pathways such as calcium signaling and lipid signaling. More recently, the mitochondria-associated ER membrane (MAM), which is a site of close contact between the ER and mitochondria, has been shown to function as a platform for various intracellular stress responses including apoptotic signaling, inflammatory signaling, the autophagic response, and the UPR. Interestingly, in cancer, these signaling pathways from the ER are often dysregulated, contributing to cancer cell metabolism. Thus, the signaling pathway from the ER may be a novel therapeutic target for various cancers. In this review, we discuss recent research on the roles of stress responses from the ER, including the MAM.
Collapse
Affiliation(s)
- Hironori Kato
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| | - Hideki Nishitoh
- Laboratory of Biochemistry and Molecular Biology, Department of Medical Sciences, University of Miyazaki , Miyazaki , Japan
| |
Collapse
|
19
|
de Munnik SM, Smit MJ, Leurs R, Vischer HF. Modulation of cellular signaling by herpesvirus-encoded G protein-coupled receptors. Front Pharmacol 2015; 6:40. [PMID: 25805993 PMCID: PMC4353375 DOI: 10.3389/fphar.2015.00040] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 02/12/2015] [Indexed: 12/22/2022] Open
Abstract
Human herpesviruses (HHVs) are widespread infectious pathogens that have been associated with proliferative and inflammatory diseases. During viral evolution, HHVs have pirated genes encoding viral G protein-coupled receptors (vGPCRs), which are expressed on infected host cells. These vGPCRs show highest homology to human chemokine receptors, which play a key role in the immune system. Importantly, vGPCRs have acquired unique properties such as constitutive activity and the ability to bind a broad range of human chemokines. This allows vGPCRs to hijack human proteins and modulate cellular signaling for the benefit of the virus, ultimately resulting in immune evasion and viral dissemination to establish a widespread and lifelong infection. Knowledge on the mechanisms by which herpesviruses reprogram cellular signaling might provide insight in the contribution of vGPCRs to viral survival and herpesvirus-associated pathologies.
Collapse
Affiliation(s)
- Sabrina M de Munnik
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Martine J Smit
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Rob Leurs
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| | - Henry F Vischer
- Amsterdam Institute for Molecules Medicines and Systems - Division of Medicinal Chemistry, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, Amsterdam Netherlands
| |
Collapse
|
20
|
Noskovičová N, Petřek M, Eickelberg O, Heinzelmann K. Platelet-Derived Growth Factor Signaling in the Lung. From Lung Development and Disease to Clinical Studies. Am J Respir Cell Mol Biol 2015; 52:263-84. [DOI: 10.1165/rcmb.2014-0294tr] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
21
|
Chen J, Tang H, Sysol JR, Moreno-Vinasco L, Shioura KM, Chen T, Gorshkova I, Wang L, Huang LS, Usatyuk PV, Sammani S, Zhou G, Raj JU, Garcia JGN, Berdyshev E, Yuan JXJ, Natarajan V, Machado RF. The sphingosine kinase 1/sphingosine-1-phosphate pathway in pulmonary arterial hypertension. Am J Respir Crit Care Med 2014; 190:1032-43. [PMID: 25180446 DOI: 10.1164/rccm.201401-0121oc] [Citation(s) in RCA: 104] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
RATIONALE Sphingosine kinases (SphKs) 1 and 2 regulate the synthesis of the bioactive sphingolipid sphingosine-1-phosphate (S1P), an important lipid mediator that promotes cell proliferation, migration, and angiogenesis. OBJECTIVES We aimed to examine whether SphKs and their product, S1P, play a role in the development of pulmonary arterial hypertension (PAH). METHODS SphK1(-/-), SphK2(-/-), and S1P lyase heterozygous (Sgpl1(+/-)) mice, a pharmacologic SphK inhibitor (SKI2), and a S1P receptor 2 (S1PR2) antagonist (JTE013) were used in rodent models of hypoxia-mediated pulmonary hypertension (HPH). S1P levels in lung tissues from patients with PAH and pulmonary arteries (PAs) from rodent models of HPH were measured. MEASUREMENTS AND MAIN RESULTS mRNA and protein levels of SphK1, but not SphK2, were significantly increased in the lungs and isolated PA smooth muscle cells (PASMCs) from patients with PAH, and in lungs of experimental rodent models of HPH. S1P levels were increased in lungs of patients with PAH and PAs from rodent models of HPH. Unlike SphK2(-/-) mice, SphK1(-/-) mice were protected against HPH, whereas Sgpl1(+/-) mice were more susceptible to HPH. Pharmacologic SphK1 and S1PR2 inhibition prevented the development of HPH in rodent models of HPH. Overexpression of SphK1 and stimulation with S1P potentially via ligation of S1PR2 promoted PASMC proliferation in vitro, whereas SphK1 deficiency inhibited PASMC proliferation. CONCLUSIONS The SphK1/S1P axis is a novel pathway in PAH that promotes PASMC proliferation, a major contributor to pulmonary vascular remodeling. Our results suggest that this pathway is a potential therapeutic target in PAH.
Collapse
Affiliation(s)
- Jiwang Chen
- 1 Section of Pulmonary, Critical Care Medicine, Sleep and Allergy, Department of Medicine
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Cattaneo F, Guerra G, Parisi M, De Marinis M, Tafuri D, Cinelli M, Ammendola R. Cell-surface receptors transactivation mediated by g protein-coupled receptors. Int J Mol Sci 2014; 15:19700-28. [PMID: 25356505 PMCID: PMC4264134 DOI: 10.3390/ijms151119700] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Revised: 09/30/2014] [Accepted: 10/13/2014] [Indexed: 12/17/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are seven transmembrane-spanning proteins belonging to a large family of cell-surface receptors involved in many intracellular signaling cascades. Despite GPCRs lack intrinsic tyrosine kinase activity, tyrosine phosphorylation of a tyrosine kinase receptor (RTK) occurs in response to binding of specific agonists of several such receptors, triggering intracellular mitogenic cascades. This suggests that the notion that GPCRs are associated with the regulation of post-mitotic cell functions is no longer believable. Crosstalk between GPCR and RTK may occur by different molecular mechanism such as the activation of metalloproteases, which can induce the metalloprotease-dependent release of RTK ligands, or in a ligand-independent manner involving membrane associated non-receptor tyrosine kinases, such as c-Src. Reactive oxygen species (ROS) are also implicated as signaling intermediates in RTKs transactivation. Intracellular concentration of ROS increases transiently in cells stimulated with GPCR agonists and their deliberated and regulated generation is mainly catalyzed by enzymes that belong to nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family. Oxidation and/or reduction of cysteine sulfhydryl groups of phosphatases tightly controls the activity of RTKs and ROS-mediated inhibition of cellular phosphatases results in an equilibrium shift from the non-phosphorylated to the phosphorylated state of RTKs. Many GPCR agonists activate phospholipase C, which catalyze the hydrolysis of phosphatidylinositol 4,5-bis-phosphate to produce inositol 1,4,5-triphosphate and diacylglicerol. The consequent mobilization of Ca2+ from endoplasmic reticulum leads to the activation of protein kinase C (PKC) isoforms. PKCα mediates feedback inhibition of RTK transactivation during GPCR stimulation. Recent data have expanded the coverage of transactivation to include Serine/Threonine kinase receptors and Toll-like receptors. Herein, we discuss the main mechanisms of GPCR-mediated cell-surface receptors transactivation and the pathways involved in intracellular responses induced by GPCR agonists. These studies may suggest the design of novel strategies for therapeutic interventions.
Collapse
Affiliation(s)
- Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Germano Guerra
- Department of Medicine and Health Sciences, University of Molise, Campobasso 86100, Italy.
| | - Melania Parisi
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Marta De Marinis
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Domenico Tafuri
- Department of Sport Science and Wellness, University of Naples Parthenope, Naples 80133, Italy.
| | - Mariapia Cinelli
- Department of Public Health, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples 80131, Italy.
| |
Collapse
|
23
|
Sphingosine-1-phosphate-induced Flk-1 transactivation stimulates mouse embryonic stem cell proliferation through S1P1/S1P3-dependent β-arrestin/c-Src pathways. Stem Cell Res 2014; 12:69-85. [DOI: 10.1016/j.scr.2013.08.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 08/08/2013] [Accepted: 08/29/2013] [Indexed: 01/21/2023] Open
|
24
|
Rutherford C, Childs S, Ohotski J, McGlynn L, Riddick M, MacFarlane S, Tasker D, Pyne S, Pyne NJ, Edwards J, Palmer TM. Regulation of cell survival by sphingosine-1-phosphate receptor S1P1 via reciprocal ERK-dependent suppression of Bim and PI-3-kinase/protein kinase C-mediated upregulation of Mcl-1. Cell Death Dis 2013; 4:e927. [PMID: 24263101 PMCID: PMC3847331 DOI: 10.1038/cddis.2013.455] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2013] [Revised: 10/16/2013] [Accepted: 10/16/2013] [Indexed: 01/10/2023]
Abstract
Although the ability of bioactive lipid sphingosine-1-phosphate (S1P) to positively regulate anti-apoptotic/pro-survival responses by binding to S1P1 is well known, the molecular mechanisms remain unclear. Here we demonstrate that expression of S1P1 renders CCL39 lung fibroblasts resistant to apoptosis following growth factor withdrawal. Resistance to apoptosis was associated with attenuated accumulation of pro-apoptotic BH3-only protein Bim. However, although blockade of extracellular signal-regulated kinase (ERK) activation could reverse S1P1-mediated suppression of Bim accumulation, inhibition of caspase-3 cleavage was unaffected. Instead S1P1-mediated inhibition of caspase-3 cleavage was reversed by inhibition of phosphatidylinositol-3-kinase (PI3K) and protein kinase C (PKC), which had no effect on S1P1 regulation of Bim. However, S1P1 suppression of caspase-3 was associated with increased expression of anti-apoptotic protein Mcl-1, the expression of which was also reduced by inhibition of PI3K and PKC. A role for the induction of Mcl-1 in regulating endogenous S1P receptor-dependent pro-survival responses in human umbilical vein endothelial cells was confirmed using S1P receptor agonist FTY720-phosphate (FTY720P). FTY720P induced a transient accumulation of Mcl-1 that was associated with a delayed onset of caspase-3 cleavage following growth factor withdrawal, whereas Mcl-1 knockdown was sufficient to enhance caspase-3 cleavage even in the presence of FTY720P. Consistent with a pro-survival role of S1P1 in disease, analysis of tissue microarrays from ER(+) breast cancer patients revealed a significant correlation between S1P1 expression and tumour cell survival. In these tumours, S1P1 expression and cancer cell survival were correlated with increased activation of ERK, but not the PI3K/PKB pathway. In summary, pro-survival/anti-apoptotic signalling from S1P1 is intimately linked to its ability to promote the accumulation of pro-survival protein Mcl-1 and downregulation of pro-apoptotic BH3-only protein Bim via distinct signalling pathways. However, the functional importance of each pathway is dependent on the specific cellular context.
Collapse
Affiliation(s)
- C Rutherford
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - S Childs
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - J Ohotski
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - L McGlynn
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - M Riddick
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - S MacFarlane
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - D Tasker
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - S Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - N J Pyne
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, Scotland, UK
| | - J Edwards
- Institute of Cancer Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| | - T M Palmer
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Scotland, UK
| |
Collapse
|
25
|
Sciaccaluga M, D’Alessandro G, Pagani F, Ferrara G, Lopez N, Warr T, Gorello P, Porzia A, Mainiero F, Santoro A, Esposito V, Cantore G, Castigli E, Limatola C. Functional cross talk between CXCR4 and PDGFR on glioblastoma cells is essential for migration. PLoS One 2013; 8:e73426. [PMID: 24023874 PMCID: PMC3759384 DOI: 10.1371/journal.pone.0073426] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 07/19/2013] [Indexed: 11/19/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive form of brain tumor, characterized by high migratory behavior and infiltration in brain parenchyma which render classic therapeutic approach ineffective. The migratory behaviour of GBM cells could be conditioned by a number of tissue- and glioma-derived cytokines and growth factors. Although the pro-migratory action of CXCL12 on GBM cells in vitro and in vivo is recognized, the molecular mechanisms involved are not clearly identified. In fact the signaling pathways involved in the pro-migratory action of CXCL12 may differ in individual glioblastoma and integrate with those resulting from abnormal expression and activation of growth factor receptors. In this study we investigated whether some of the receptor tyrosine kinases commonly expressed in GBM cells could cooperate with CXCL12/CXCR4 in their migratory behavior. Our results show a functional cross-talk between CXCR4 and PDGFR which appears to be essential for GBM chemotaxis.
Collapse
Affiliation(s)
| | - Giuseppina D’Alessandro
- Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Fisiologia e Farmacologia Sapienza University of Rome, Rome, Italy
| | - Francesca Pagani
- Centre for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Giuseppina Ferrara
- Department of Cellular and Environmental Biology, University of Perugia, Perugia, Italy
| | - Nadia Lopez
- Department of Cellular and Environmental Biology, University of Perugia, Perugia, Italy
| | - Tracy Warr
- Department of Cellular and Environmental Biology, University of Perugia, Perugia, Italy
| | | | - Alessandra Porzia
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Fabrizio Mainiero
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Antonio Santoro
- Department of Neurology and Psychiatry, Sapienza University of Rome, Rome, Italy
| | | | | | - Emilia Castigli
- Department of Cellular and Environmental Biology, University of Perugia, Perugia, Italy
| | - Cristina Limatola
- IRCCS Neuromed, Venafro, Italy
- Istituto Pasteur Fondazione Cenci Bolognetti, Dipartimento di Fisiologia e Farmacologia Sapienza University of Rome, Rome, Italy
- * E-mail:
| |
Collapse
|
26
|
Zhao CG, Qin J, He XJ, Guan YC, Jia Y, Lei W. Sphingosine-1-phosphate is a possible fibrogenic factor in gluteal muscle fibrosis. Physiol Res 2013; 62:691-9. [PMID: 23869887 DOI: 10.33549/physiolres.932441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Gluteal muscle contracture (GMC) is a chronic fibrotic disease of gluteal muscles due to multiple etiologies. The main pathologic process is characterized by proliferation of fibroblasts and excessive accumulation of collagen in the extracellular matrix of the muscle. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid and has been reported to be associated with various fibrotic diseases. However, the role of S1P in GMC remains unknown. Here in this article, High-performance liquid chromatography and immunohistochemistry were applied to evaluate S1P localization and expression in clinical samples from patients with GMC, Quantitative real time PCR, Western blot, and enzyme-linked immunosorbent assay were used to explore the link between transforming growth factor-beta1 (TGF-beta1), plasminogen activator inhibitor-1 (PAI-1) and S1P. The results showed that S1P was enhanced in contraction band (CB) tissues. Studies using the cell proliferation and transformation assay indicated that exogenous S1P stimulated CB fibroblast proliferation in a time-dependent manner and in higher concentration also in a dose-dependent manner. Furthermore, we demonstrated that S1P not only promoted collagen type I production, but also up-regulated mRNA and protein expression of transforming growth factor-beta1 and plasminogen activator inhibitor-1. These findings suggest that S1P may regulate increased synthesis of collagen and other fibrogenic factors, and significantly contributes to the process of gluteal muscle scarring in patients with GMC.
Collapse
Affiliation(s)
- C G Zhao
- Orthopedic Center of Chinese PLA, Urumqi General Hospital of Lanzhou Military Region, Urumqi, Xinjiang, China.
| | | | | | | | | | | |
Collapse
|
27
|
Sphingosine 1-phosphate and cancer: lessons from thyroid cancer cells. Biomolecules 2013; 3:303-15. [PMID: 24970169 PMCID: PMC4030848 DOI: 10.3390/biom3020303] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 02/03/2023] Open
Abstract
Sphingomyelin is found in the cell membrane of all eukaryotic cells, and was for a long time considered merely as a structural component. However, during the last two decades, metabolites of sphingomyelin, especially sphingosine 1-phosphate (S1P), have proven to be physiologically significant regulators of cell function. Through its five different G protein-coupled receptors, S1P regulates a wide array of cellular processes, ranging from stimulating cellular proliferation and migration, to the inhibition of apoptosis and induction of angiogenesis and modulation of cellular calcium homeostasis. Many of the processes regulated by S1P are important for normal cell physiology, but may also induce severe pathological conditions, especially in malignancies like cancer. Thus, understanding S1P signaling mechanisms has been the aim of a multitude of investigations. Great interest has also been shown in understanding the action of sphingosine kinase (SphK), i.e., the kinase phosphorylating sphingosine to S1P, and the interactions between S1P and growth factor signaling. In the present review, we will discuss recent findings regarding the possible importance of S1P and SphK in the etiology of thyroid cancer. Although clinical data is still scarce, our in vitro findings suggest that S1P may function as a “double-edged sword”, as the receptor profile of thyroid cancer cells largely determines whether S1P stimulates or blocks cellular migration. We will also discuss the interactions between S1P- and VEGF-evoked signaling, and the importance of a S1P1-VEGF receptor 2 complex in thyroid cancer cells.
Collapse
|
28
|
Kruk JS, Vasefi MS, Liu H, Heikkila JJ, Beazely MA. 5-HT1A receptors transactivate the platelet-derived growth factor receptor type beta in neuronal cells. Cell Signal 2013; 25:133-43. [DOI: 10.1016/j.cellsig.2012.09.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Revised: 09/07/2012] [Accepted: 09/16/2012] [Indexed: 01/23/2023]
|
29
|
Abstract
Sphingosine-1-phosphate (S1P) regulates important functions in cardiac and vascular homeostasis. It has been implied to play causal roles in the pathogenesis of many cardiovascular disorders such as coronary artery disease, atherosclerosis, myocardial infarction, and heart failure. The majority of S1P in plasma is associated with high-density lipoproteins (HDL), and their S1P content has been shown to be responsible, at least in part, for several of the beneficial effects of HDL on cardiovascular risk. The attractiveness of S1P-based drugs for potential cardiovascular applications is increasing in the wake of the clinical approval of FTY720, but answers to important questions on the effects of S1P in cardiovascular biology and medicine must still be found. This chapter focuses on the current understanding of the role of S1P and its receptors in cardiovascular physiology, pathology, and disease.
Collapse
Affiliation(s)
- Bodo Levkau
- University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
30
|
Berce V, Kozmus CEP, Potočnik U. Association among ORMDL3 gene expression, 17q21 polymorphism and response to treatment with inhaled corticosteroids in children with asthma. THE PHARMACOGENOMICS JOURNAL 2012; 13:523-9. [PMID: 22986918 DOI: 10.1038/tpj.2012.36] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 07/20/2012] [Accepted: 08/13/2012] [Indexed: 02/04/2023]
Abstract
Recent genome-wide association studies linked childhood asthma with single-nucleotide polymorphisms (SNPs) in ORM1-like protein 3 (ORMDL3) gene region on chromosome 17q21. We analyzed the effect of functional SNP rs2872507 in ORMDL3 gene region on the response to antiasthmatic treatment with inhaled corticosteroids (ICSs) and ORMDL3 gene expression. Forced expiratory volume in 1 s increased significantly by 13.3% of predicted value after therapy in atopic asthmatics with AA genotype, compared with 7.0% in heterozygotes and 4.9% increase in GG homozygotes (P=0.0176). Median relative expression of ORMDL3 gene in asthmatics with AA, AG and GG genotypes was 0.75, 1.05 and 1.21, respectively (P<0.0001). Treatment with ICSs was significantly associated with the increase of median relative expression of ORMDL3 gene, from 0.88 to 1.21 (P=0.0032) in atopic asthmatics. Our results suggest that rs2872507 is associated with ORMDL3 gene expression and with ICS treatment response in children with atopic asthma.
Collapse
Affiliation(s)
- V Berce
- Department of Pediatrics, University Medical Centre Maribor, Maribor, Slovenia
| | | | | |
Collapse
|
31
|
Schwalm S, Pfeilschifter J, Huwiler A. Sphingosine-1-phosphate: a Janus-faced mediator of fibrotic diseases. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:239-50. [PMID: 22889995 DOI: 10.1016/j.bbalip.2012.07.022] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/27/2012] [Accepted: 07/28/2012] [Indexed: 12/12/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a pleiotropic lipid mediator that acts either on G protein-coupled S1P receptors on the cell surface or via intracellular target sites. In addition to the well established effects of S1P in angiogenesis, carcinogenesis and immunity, evidence is now continuously accumulating which demonstrates that S1P is an important regulator of fibrosis. The contribution of S1P to fibrosis is of a Janus-faced nature as S1P exhibits both pro- and anti-fibrotic effects depending on its site of action. Extracellular S1P promotes fibrotic processes in a S1P receptor-dependent manner, whereas intracellular S1P has an opposite effect and dampens a fibrotic reaction by yet unidentified mechanisms. Fibrosis is a result of chronic irritation by various factors and is defined by an excess production of extracellular matrix leading to tissue scarring and organ dysfunction. In this review, we highlight the general effects of extracellular and intracellular S1P on the multistep cascade of pathological fibrogenesis including tissue injury, inflammation and the action of pro-fibrotic cytokines that stimulate ECM production and deposition. In a second part we summarize the current knowledge about the involvement of S1P signaling in the development of organ fibrosis of the lung, kidney, liver, heart and skin. Altogether, it is becoming clear that targeting the sphingosine kinase-1/S1P signaling pathway offers therapeutic potential in the treatment of various fibrotic processes. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- Stephanie Schwalm
- Pharmazentrum Frankfurt/ZAFES, Klinikum der Goethe-Universität Frankfurt am Main, Germany
| | | | | |
Collapse
|
32
|
Liu X, Xiong SL, Yi GH. ABCA1, ABCG1, and SR-BI: Transit of HDL-associated sphingosine-1-phosphate. Clin Chim Acta 2012; 413:384-90. [DOI: 10.1016/j.cca.2011.11.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2011] [Revised: 10/28/2011] [Accepted: 11/03/2011] [Indexed: 01/07/2023]
|
33
|
Abstract
There is an increasing body of evidence demonstrating a critical role for the bioactive lipid S1P (sphingosine 1-phosphate) in cancer. S1P is synthesized and metabolized by a number of enzymes, including sphingosine kinase, S1P lyase and S1P phosphatases. S1P binds to cell-surface G-protein-coupled receptors (S1P1–S1P5) to elicit cell responses and can also regulate, by direct binding, a number of intracellular targets such as HDAC (histone deacetylase) 1/2 to induce epigenetic regulation. S1P is involved in cancer progression including cell transformation/oncogenesis, cell survival/apoptosis, cell migration/metastasis and tumour microenvironment neovascularization. In the present paper, we describe our research findings regarding the correlation of sphingosine kinase 1 and S1P receptor expression in tumours with clinical outcome and we define some of the molecular mechanisms underlying the involvement of sphingosine kinase 1 and S1P receptors in the formation of a cancer cell migratory phenotype. The role of sphingosine kinase 1 in the acquisition of chemotherapeutic resistance and the interaction of S1P receptors with oncogenes such as HER2 is also reviewed. We also discuss novel aspects of the use of small-molecule inhibitors of sphingosine kinase 1 in terms of allosterism, ubiquitin–proteasomal degradation of sphingosine kinase 1 and anticancer activity. Finally, we describe how S1P receptor-modulating agents abrogate S1P receptor–receptor tyrosine kinase interactions, with potential to inhibit growth-factor-dependent cancer progression.
Collapse
|
34
|
Liu X, Zhang QH, Yi GH. Regulation of metabolism and transport of sphingosine-1-phosphate in mammalian cells. Mol Cell Biochem 2011; 363:21-33. [DOI: 10.1007/s11010-011-1154-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 11/11/2011] [Indexed: 02/04/2023]
|
35
|
Schuchardt M, Tölle M, Prüfer J, van der Giet M. Pharmacological relevance and potential of sphingosine 1-phosphate in the vascular system. Br J Pharmacol 2011; 163:1140-62. [PMID: 21309759 DOI: 10.1111/j.1476-5381.2011.01260.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) was identified as a crucial molecule for regulating immune responses, inflammatory processes as well as influencing the cardiovascular system. S1P mediates differentiation, proliferation and migration during vascular development and homoeostasis. S1P is a naturally occurring lipid metabolite and is present in human blood in nanomolar concentrations. S1P is not only involved in physiological but also in pathophysiological processes. Therefore, this complex signalling system is potentially interesting for pharmacological intervention. Modulation of the system might influence inflammatory, angiogenic or vasoregulatory processes. S1P activates G-protein coupled receptors, namely S1P(1-5) , whereas only S1P(1-3) is present in vascular cells. S1P can also act as an intracellular signalling molecule. This review highlights the pharmacological potential of S1P signalling in the vascular system by giving an overview of S1P-mediated processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). After a short summary of S1P metabolism and signalling pathways, the role of S1P in EC and VSMC proliferation and migration, the cause of relaxation and constriction of arterial blood vessels, the protective functions on endothelial apoptosis, as well as the regulatory function in leukocyte adhesion and inflammatory responses are summarized. This is followed by a detailed description of currently known pharmacological agonists and antagonists as new tools for mediating S1P signalling in the vasculature. The variety of effects influenced by S1P provides plenty of therapeutic targets currently under investigation for potential pharmacological intervention.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité- Universitätsmedizin Berlin, CharitéCentrum 10, Department of Nephrology, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
36
|
Wang L, Guo D, Xing B, Zhang JJ, Shu HB, Guo L, Huang XY. Resistance to inhibitors of cholinesterase-8A (Ric-8A) is critical for growth factor receptor-induced actin cytoskeletal reorganization. J Biol Chem 2011; 286:31055-31061. [PMID: 21771786 DOI: 10.1074/jbc.m111.253427] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Heterotrimeric G proteins are critical transducers of cellular signaling. In addition to their classic roles in relaying signals from G protein-coupled receptors (GPCRs), heterotrimeric G proteins also mediate physiological functions from non-GPCRs. Previously, we have shown that Gα(13), a member of the heterotrimeric G proteins, is essential for growth factor receptor-induced actin cytoskeletal reorganization such as dynamic dorsal ruffle turnover and cell migration. These Gα(13)-mediated dorsal ruffle turnover and cell migration by growth factors acting on their receptor tyrosine kinases (RTKs) are independent of GPCRs. However, the mechanism by which RTKs signal to Gα(13) is not known. Here, we show that cholinesterase-8A (Ric-8A), a nonreceptor guanine nucleotide exchange factor for some heterotrimeric G proteins, is critical for coupling RTKs to Gα(13). Down-regulation of Ric-8A protein levels in cells by RNA interference slowed down platelet-derived growth factor (PDGF)-induced dorsal ruffle turnover and inhibited PDGF-initiated cell migration. PDGF was able to increase the activity of Ric-8A in cells. Furthermore, purified Ric-8A proteins interact directly with purified Gα(13) protein in a nucleotide-dependent manner. Deficiency of Ric-8A prevented the translocation of Gα(13) to the cell cortex. Hence, Ric-8A is critical for growth factor receptor-induced actin cytoskeletal reorganization.
Collapse
Affiliation(s)
- Limin Wang
- College of Life Sciences, Wuhan University, Wuhan, China 430072
| | - Dagang Guo
- Department of Physiology, Cornell University Weill Medical College, New York, New York 10065
| | - Bowen Xing
- College of Life Sciences, Wuhan University, Wuhan, China 430072
| | - J Jillian Zhang
- Department of Physiology, Cornell University Weill Medical College, New York, New York 10065
| | - Hong-Bing Shu
- College of Life Sciences, Wuhan University, Wuhan, China 430072
| | - Lin Guo
- College of Life Sciences, Wuhan University, Wuhan, China 430072
| | - Xin-Yun Huang
- College of Life Sciences, Wuhan University, Wuhan, China 430072.
| |
Collapse
|
37
|
Pyne NJ, Pyne S. Selectivity and specificity of sphingosine 1-phosphate receptor ligands: "off-targets" or complex pharmacology? Front Pharmacol 2011; 2:26. [PMID: 21687518 PMCID: PMC3108476 DOI: 10.3389/fphar.2011.00026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Accepted: 05/20/2011] [Indexed: 01/19/2023] Open
Abstract
A recent perspective published in Frontiers of Pharmacology by Salomone and Waeber (2011) discussed the selectivity and specificity of sphingosine 1-phosphate (S1P) receptor ligands. This perspective surveyed the use of various S1P receptor ligands and attempted to reconcile a number of inconsistencies in the predicted biological outcomes: these were interpreted as “off-target” effects. Therefore the perspective cautioned against the use of these S1P receptor ligands. Here we highlight the complex pharmacology of S1P receptors, which along with “inside-out” signaling might provide an alternative explanation for “off-target” effects.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, UK
| | | |
Collapse
|
38
|
Pyne NJ, Pyne S. Receptor tyrosine kinase-G-protein-coupled receptor signalling platforms: out of the shadow? Trends Pharmacol Sci 2011; 32:443-50. [PMID: 21612832 DOI: 10.1016/j.tips.2011.04.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2011] [Revised: 04/20/2011] [Accepted: 04/26/2011] [Indexed: 12/29/2022]
Abstract
Receptor tyrosine kinases (RTKs) and G-protein-coupled receptors (GPCRs) can form platforms in which protein signalling components specific for each receptor are shared (owing to close proximity) to produce an integrated response upon engagement of ligands. RTK-GPCR signalling platforms respond to growth factors and GPCR agonists to increase gain over and above that which is normally produced by separate receptors. They can also function to change the spatial context of signalling in response to growth factor activation. The function of RTK-GPCR signalling platforms can be modulated with conformational-specific inhibitors that stabilise defined GPCR states to abrogate both GPCR agonist- and growth factor-stimulated cell responses. In this paper, we provide an opinion of the biology and unusual pharmacology of RTK-GPCR signalling platforms and make comparisons with a more traditional model of crosstalk between RTKs and GPCRs.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow, G4 0RE, UK.
| | | |
Collapse
|
39
|
Schröder M, Richter C, Juan MHS, Maltusch K, Giegold O, Quintini G, Pfeilschifter JM, Huwiler A, Radeke HH. The sphingosine kinase 1 and S1P1 axis specifically counteracts LPS-induced IL-12p70 production in immune cells of the spleen. Mol Immunol 2011; 48:1139-48. [DOI: 10.1016/j.molimm.2011.02.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2010] [Revised: 02/14/2011] [Accepted: 02/17/2011] [Indexed: 01/29/2023]
|
40
|
Pitson SM. Regulation of sphingosine kinase and sphingolipid signaling. Trends Biochem Sci 2011; 36:97-107. [DOI: 10.1016/j.tibs.2010.08.001] [Citation(s) in RCA: 217] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Revised: 08/16/2010] [Accepted: 08/16/2010] [Indexed: 01/09/2023]
|
41
|
Catarzi S, Romagnoli C, Marcucci G, Favilli F, Iantomasi T, Vincenzini MT. Redox regulation of ERK1/2 activation induced by sphingosine 1-phosphate in fibroblasts: involvement of NADPH oxidase and platelet-derived growth factor receptor. Biochim Biophys Acta Gen Subj 2011; 1810:446-56. [PMID: 21256191 DOI: 10.1016/j.bbagen.2011.01.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2010] [Revised: 12/20/2010] [Accepted: 01/13/2011] [Indexed: 01/05/2023]
Abstract
BACKGROUND Sphingosine 1-phosphate (S1P) is a sphingolipid metabolite synthesized after stimulation with growth factors or cytokines. S1P extracellular effects are mediated through specific Gi-protein coupled receptors (GPCRs). Recently, we demonstrated in NIH3T3 fibroblasts stimulated by platelet-derived growth factor (PDGF) or S1P the NADPH oxidase activation and the H(2)O(2) intracellular level increase trough the Gi protein involvement. METHODS NIH3T3 fibroblast cell cultures were used. Western blot and quantitative analyses by Chemidoc-Quantity-One software were performed. H(2)O(2) level was assayed by fluorescence spectrophotometric analysis, and cell proliferation by counted manually or ELISA kit. RESULTS This study demonstrates, in NIH 3T3 fibroblasts, a novel redox regulated mechanism of S1P-induced activation of ERK 1/2 related to NADPH oxidase activity and intracellular H(2)O(2) level increase with PDGF receptor tyrosine kinase involvement through a transactivation mechanism. This event is mediated by S1P(1) and S1P(3) receptors by Gi proteins and can contribute to S1P mitogenic signaling. CONCLUSION These results can be related to mechanisms of cross-talk previously identified between receptor tyrosine kinase, including PDGFreceptor, and several GPCR ligands. GENERAL SIGNIFICANCE The redox-sensitive ERK1/2 and PDGFr tyrosine kinase activity could be targets for therapies in diseases in which deregulation of intracellular oxidative status and the consequent alteration of S1P and/or PDGF signaling pathway are involved.
Collapse
Affiliation(s)
- Serena Catarzi
- Department of Biochemical Science, University of Florence, Viale Morgagni 50, 50134, Florence, Italy
| | | | | | | | | | | |
Collapse
|
42
|
Nincheri P, Bernacchioni C, Cencetti F, Donati C, Bruni P. Sphingosine kinase-1/S1P1 signalling axis negatively regulates mitogenic response elicited by PDGF in mouse myoblasts. Cell Signal 2010; 22:1688-99. [PMID: 20600848 DOI: 10.1016/j.cellsig.2010.06.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2010] [Revised: 06/17/2010] [Accepted: 06/17/2010] [Indexed: 10/19/2022]
Abstract
PDGF is known to be critically implicated in skeletal muscle repair; however its molecular mechanism of action has been only marginally investigated. In this study we show that in mouse myoblasts PDGF transactivates S1P(1) receptor via sphingosine kinase (SK)-1 activation and that this molecular event exerts a negative regulation of the mitogenic effect elicited by this growth factor. Indeed, pharmacological inhibition of S1P(1), or its specific silencing increased PDGF-dependent cell proliferation, whereas S1P(1) overexpression diminished the biological effect. Moreover, the mitogenic response to PDGF was enhanced by pharmacological inhibition of SK activity as well as specific silencing of SK1 but not SK2. Furthermore, ERK1/2 signalling pathway was found to be upstream of the observed attenuation of PDGF-induced cell proliferation. Interestingly, PDGF-directed engagement of S1P(1) exerted also a positive modulatory action of the growth factor-dependent cell motility. The here highlighted dual role of S1P(1)-mediated signalling in response to myoblast challenge with PDGF is likely important to guarantee the fine control of the biological response to this growth factor, finalized to efficient repopulation of skeletal muscle after damage, where a tight balance between proliferation and migration of tissue progenitor cells is required.
Collapse
Affiliation(s)
- Paola Nincheri
- Dipartimento di Scienze Biochimiche, Universita; di Firenze, 50134 Firenze, Italy
| | | | | | | | | |
Collapse
|
43
|
Siow DL, Anderson CD, Berdyshev EV, Skobeleva A, Pitson SM, Wattenberg BW. Intracellular localization of sphingosine kinase 1 alters access to substrate pools but does not affect the degradative fate of sphingosine-1-phosphate. J Lipid Res 2010; 51:2546-59. [PMID: 20386061 PMCID: PMC2918438 DOI: 10.1194/jlr.m004374] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2009] [Revised: 04/11/2010] [Indexed: 02/05/2023] Open
Abstract
Sphingosine kinase 1 (SK1) produces sphingosine-1-phosphate (S1P), a potent signaling lipid. The subcellular localization of SK1 can dictate its signaling function. Here, we use artificial targeting of SK1 to either the plasma membrane (PM) or the endoplasmic reticulum (ER) to test the effects of compartmentalization of SK1 on substrate utilization and downstream metabolism of S1P. Expression of untargeted or ER-targeted SK1, but surprisingly not PM-targeted SK1, results in a dramatic increase in the phosphorylation of dihydrosphingosine, a metabolic precursor in de novo ceramide synthesis. Conversely, knockdown of endogenous SK1 diminishes both dihydrosphingosine-1-phosphate and S1P levels. We tested the effects of SK1 localization on degradation of S1P by depletion of the ER-localized S1P phosphatases and lyase. Remarkably, S1P produced at the PM was degraded to the same extent as that produced in the ER. This indicates that there is an efficient mechanism for the transport of S1P from the PM to the ER. In acute labeling experiments, we find that S1P degradation is primarily driven by lyase cleavage of S1P. Counterintuitively, when S1P-specific phosphatases are depleted, acute labeling of S1P is significantly reduced, indicative of a phosphatase-dependent recycling process. We conclude that the localization of SK1 influences the substrate pools that it has access to and that S1P can rapidly translocate from the site where it is synthesized to other intracellular sites.
Collapse
Affiliation(s)
- Deanna L. Siow
- Departments of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY
- Departments of Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY
| | - Charles D. Anderson
- Departments of Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY
| | - Evgeny V. Berdyshev
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL
| | - Anastasia Skobeleva
- Biological Sciences Division, Department of Medicine, University of Chicago, Chicago, IL
| | - Stuart M. Pitson
- Centre for Cancer Biology, SA Pathology, Frome Road, Adelaide, Australia
- School of Molecular and Biomedical Science, University of Adelaide, Adelaide, Australia
| | - Binks W. Wattenberg
- Departments of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, KY
- Departments of Brown Cancer Center, School of Medicine, University of Louisville, Louisville, KY
- Departments of Pharmacology and Toxicology, School of Medicine, University of Louisville, Louisville, KY
| |
Collapse
|
44
|
Bergelin N, Löf C, Balthasar S, Kalhori V, Törnquist K. S1P1 and VEGFR-2 form a signaling complex with extracellularly regulated kinase 1/2 and protein kinase C-alpha regulating ML-1 thyroid carcinoma cell migration. Endocrinology 2010; 151:2994-3005. [PMID: 20501673 DOI: 10.1210/en.2009-1387] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Sphingosine 1-phosphate (S1P) and vascular endothelial growth factor receptor 2 (VEGFR-2) signaling have been shown to integrate in many biological processes. The follicular thyroid carcinoma cell line ML-1 expresses VEGFR-2 and secretes substantial amounts of both vascular endothelial growth factor (VEGF)-A and VEGF-C. ML-1 cells also express S1P-receptors (S1P(1-3,5)). S1P is able to phosphorylate VEGFR-2, and inhibiting VEGFR-2 attenuates S1P-induced migration and down-regulates S1P(1) expression in ML-1 cells. In the present study, we focused on the interactions between S1P(1) and VEGFR-2. We show that S1P receptors form complexes with VEGFR-2 and that the S1P(1)/VEGFR-2 complex associates with protein kinase C (PKC)-alpha and ERK1/2. Furthermore, the complex evokes bidirectional signaling since the S1P-induced ERK1/2 phosphorylation is sensitive to VEGFR-2 kinase inhibition and VEGF-A-induced ERK1/2 phosphorylation is sensitive to pertussis toxin treatment as well as S1P(1) small interfering RNA (siRNA) treatment. Both S1P- and VEGF-A-induced haptotaxis is sensitive to pertussis toxin treatment and S1P(1) siRNA treatment. Phosphorylation of ERK1/2 evoked by both VEGF-A and the S1P(1) agonist SEW-2871 is inhibited by PKC-alpha and PKC-betaI siRNA. We hypothesize that VEGFR-2 forms a signaling complex with S1P(1), evoking bidirectional signaling regulating both ERK1/2 phosphorylation and haptotaxis of ML-1 cells.
Collapse
Affiliation(s)
- Nina Bergelin
- Department of Biosciences, Abo Akademi University, BioCity, Tykistökatu 6, 20520 Turku, Finland
| | | | | | | | | |
Collapse
|
45
|
Abstract
There is substantial evidence that sphingosine 1-phosphate (S1P) is involved in cancer. S1P regulates processes such as inflammation, which can drive tumorigenesis; neovascularization, which provides cancer cells with nutrients and oxygen; and cell growth and survival. This occurs at multiple levels and involves S1P receptors, sphingosine kinases, S1P phosphatases and S1P lyase. This Review summarizes current research findings and examines the potential for new therapeutics designed to alter S1P signalling and function in cancer.
Collapse
Affiliation(s)
- Nigel J Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 27 Taylor Street, Glasgow G4 0NR, UK.
| | | |
Collapse
|
46
|
Rodgers A, Mormeneo D, Long JS, Delgado A, Pyne NJ, Pyne S. Sphingosine 1-phosphate regulation of extracellular signal-regulated kinase-1/2 in embryonic stem cells. Stem Cells Dev 2010; 18:1319-30. [PMID: 19228106 DOI: 10.1089/scd.2009.0023] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Recent evidence suggests that sphingosine 1-phosphate (S1P) regulates self-renewal of human embryonic stem (ES) cells and differentiation of mouse embryoid bodies (derived from mouse ES cells) to cardiomyocytes. We have investigated the role of S1P in regulating ERK-1/2 signaling in mouse ES cells. In this regard, we found that both mouse ES-D3 and CGR8 cells express S1P(1), S1P(2), S1P(3), and S1P(5) but lack S1P(4). The treatment of ES cells with S1P induced the activation of ERK-1/2 via a mechanism that was not mediated by S1P(1), S1P(2), or S1P(3). This was based on: (i) the failure of S1P(1), S1P(2), or S1P(3) antagonists to inhibit S1P-stimulated ERK-1/2 activation and (ii) the failure of SEW 2871 (S1P(1) receptor agonist) to stimulate ERK-1/2 activation. The treatment of ES cells with phytosphingosine 1-phosphate (phyto-S1P), which we show here is an agonist of the S1P(5) receptor, stimulated ERK-1/2 activation. These findings therefore suggest that S1P(5) may mediate the effects of S1P in terms of regulating ERK-1/2 signaling in ES cells. The S1P-dependent activation of ERK-1/2 was sensitive to inhibition by pertussis toxin (uncouples the G-protein, G(i) from GPCR), bisindolylmaleimide I (PKC inhibitor), and PP2 (c-Src inhibitor), but was not reduced by LY29004 (PI3K inhibitor) suggesting that S1P uses G(i)-, PKC-, and c-Src-dependent mechanisms to activate the ERK-1/2 pathway in ES cells.
Collapse
Affiliation(s)
- Alayna Rodgers
- Cell Biology Group, SIPBS, University of Strathclyde, Glasgow G4 0NR, United Kingdom
| | | | | | | | | | | |
Collapse
|
47
|
Sengupta N, Afzal A, Caballero S, Chang KH, Shaw LC, Pang JJ, Bond VC, Bhutto I, Baba T, Lutty GA, Grant MB. Paracrine modulation of CXCR4 by IGF-1 and VEGF: implications for choroidal neovascularization. Invest Ophthalmol Vis Sci 2009; 51:2697-704. [PMID: 20007826 DOI: 10.1167/iovs.09-4137] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Modulators of angiogenesis typically work in an orchestrated manner. The authors examined the interaction between insulinlike growth factor (IGF)-1, vascular endothelial growth factor (VEGF), and stromal derived factor (SDF)-1 in vivo and in vitro using angiogenesis models. METHODS The angiogenic effect of SDF-1, alone or in combination with IGF-1 and VEGF, was assessed in human lung microvascular endothelial cells using capillary tube formation and thymidine incorporation. Immunohistochemical analysis for CD31, SDF-1, and CXCR4 was performed on mouse eyes 2 weeks after the initiation of laser rupture of Bruch's membrane, a choroidal neovascularization (CNV) model. CXCR4 antagonist and CXCR4 blocking antibody were tested on inhibition of CNV lesion size in this model. Real-time PCR was used to determine mRNA levels for SDF-1, VEGF, IGF-1, and their cognate receptors in the retinal pigment epithelium/choroid complex of mice that underwent this CNV model. RESULTS IGF-1 and VEGF demonstrated an additive effect on SDF-1-induced in vitro angiogenesis. CXCR4 immunoreactivity was present in both normal and laser-injured mice at the laser burn site and at the ganglion cell layer, the anterior portion of the inner nuclear layer, photoreceptors, and choroidal stroma. SDF-1 was observed in identical locations but was not seen in photoreceptors. mRNA levels for SDF-1, VEGF, and IGF-1 and their receptors were increased after laser injury. CXCR4-neutralizing antibody reduced neovascularization when injected subretinally but not intraperitoneally or intravitreally. CONCLUSIONS The potent proangiogenic factors IGF-1 and VEGF both stimulate SDF-1-induced angiogenesis. Local inhibition of CXCR4 is required for an antiangiogenic effect in CNV lesions.
Collapse
Affiliation(s)
- Nilanjana Sengupta
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610-0267, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rapizzi E, Taddei ML, Fiaschi T, Donati C, Bruni P, Chiarugi P. Sphingosine 1-phosphate increases glucose uptake through trans-activation of insulin receptor. Cell Mol Life Sci 2009; 66:3207-18. [PMID: 19662499 PMCID: PMC11115622 DOI: 10.1007/s00018-009-0106-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2009] [Revised: 06/22/2009] [Accepted: 07/14/2009] [Indexed: 01/12/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid that acts through a family of G-protein-coupled receptors. Herein, we report evidence of a novel redox-based cross-talk between S1P and insulin signaling pathways. In skeletal muscle cells S1P, through engagement of its S1P(2) receptor, is found to produce a transient burst of reactive oxygen species through a calcium-dependent activation of the small GTPase Rac1. S1P-induced redox-signaling is sensed by protein tyrosine phosphatase-1B, the main negative regulator of insulin receptor phosphorylation, which undergoes oxidation and enzymatic inhibition. This redox-based inhibition of the phosphatase provokes a ligand-independent trans-phosphorylation of insulin receptor and a strong increase in glucose uptake. Our results propose a new role of S1P, recognizing the lipid as an insulin-mimetic cue and pointing at reactive oxygen species as critical regulators of the cross-talk between S1P and insulin pathways. Any possible implication of S1P-directed insulin signaling in diabetes and insulin resistance remains to be established.
Collapse
Affiliation(s)
- Elena Rapizzi
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
- Interuniversity Institute of Myology, Florence, Italy
| | - Maria Letizia Taddei
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
| | - Tania Fiaschi
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
- Interuniversity Institute of Myology, Florence, Italy
| | - Chiara Donati
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
- Interuniversity Institute of Myology, Florence, Italy
| | - Paola Bruni
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
- Interuniversity Institute of Myology, Florence, Italy
| | - Paola Chiarugi
- Department of Biochemical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134 Florence, Italy
- Interuniversity Institute of Myology, Florence, Italy
| |
Collapse
|
49
|
Pyne NJ, Long JS, Lee SC, Loveridge C, Gillies L, Pyne S. New aspects of sphingosine 1-phosphate signaling in mammalian cells. ACTA ACUST UNITED AC 2009; 49:214-21. [PMID: 19534035 DOI: 10.1016/j.advenzreg.2009.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Nigel J Pyne
- Cell Biology Group, SIPBS, University of Strathclyde, 27 Taylor St, Glasgow G40NR, UK.
| | | | | | | | | | | |
Collapse
|
50
|
Rosen H, Gonzalez-Cabrera PJ, Sanna MG, Brown S. Sphingosine 1-phosphate receptor signaling. Annu Rev Biochem 2009; 78:743-68. [PMID: 19231986 DOI: 10.1146/annurev.biochem.78.072407.103733] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The sphingosine 1-phosphate (S1P) receptor signaling system is a productive model system. A hydrophobic zwitterionic lysophospholipid ligand with difficult physical properties interacts with five high-affinity G protein-coupled receptors to generate multiple downstream signals. These signals modulate homeostasis and pathology on a steep agonist concentration-response curve. Ligand presence is essential for vascular development and endothelial integrity, while acute increases in ligand concentrations result in cardiac death. Understanding this integrated biochemical system has exemplified the impact of both genetics and chemistry. Developing specific tools with defined biochemical properties for the reversible modulation of signals in real time has been essential to complement insights gained from genetic approaches that may be irreversible and compensated. Despite its knife-edge between life and death, this system, based in part on receptor subtype-selectivity and in part on differential attenuation of deleterious signals, now appears to be on the cusp of meaningful therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Hugh Rosen
- Departments of Chemical Physiology and Immunology and The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|