1
|
Kajita K, Ishii I, Mori I, Asano M, Fuwa M, Morita H. Sphingosine 1-Phosphate Regulates Obesity and Glucose Homeostasis. Int J Mol Sci 2024; 25:932. [PMID: 38256005 PMCID: PMC10816022 DOI: 10.3390/ijms25020932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
One of the major global health and welfare issues is the treatment of obesity and associated metabolic disorders, such as type 2 diabetes mellitus and nonalcoholic fatty liver disease. Obesity, caused by the excessive accumulation of triglycerides in adipose tissues, induces adipocyte dysfunction, followed by inflammation, in adipose tissues and lipotoxicity in nonadipose tissues. Several studies have shown that obesity and glucose homeostasis are influenced by sphingolipid mediators, including ceramide and sphingosine 1-phosphate (S1P). Cellular accumulation of ceramide impairs pancreatic β-cell survival, confers insulin resistance in the liver and the skeletal muscle, and deteriorates adipose tissue inflammation via unknown molecular mechanisms. The roles of S1P are more complicated, because there are five cell-surface S1P receptors (S1PRs: S1P1-5) which have altered functions, different cellular expression patterns, and inapparent intracellular targets. Recent findings, including those by our group, support the notable concept that the pharmacological activation of S1P1 or S1P3 improves obesity and associated metabolic disorders, whereas that of S1P2 has the opposite effect. In addition, the regulation of S1P production by sphingosine kinase (SphK) is an essential factor affecting glucose homeostasis. This review summarizes the current knowledge on SphK/S1P/S1PR signaling in and against obesity, insulin resistance, and associated disorders.
Collapse
Affiliation(s)
- Kazuo Kajita
- Department of Health and Nutrition, Faculty of Home Economics, Gifu Women’s University, 80 Taromaru, Gifu 501-2592, Japan
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, 3-3165 Higashitamagawagakuen, Machida 194-8543, Japan
| | - Ichiro Mori
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Motochika Asano
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Masayuki Fuwa
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| | - Hiroyuki Morita
- Department of General Medicine and General Internal Medicine, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu 501-1194, Japan; (I.M.); (M.A.); (M.F.); (H.M.)
| |
Collapse
|
2
|
Zhang Y, Wang H, Lu J, Lv Q, Yun B, Ge Z, Yan L. Down-regulation of S1PR2 is correlated with poor prognosis and immune infiltrates in cervical squamous cell carcinoma and endocervical adenocarcinoma. Int J Immunopathol Pharmacol 2023; 37:3946320231178131. [PMID: 37232164 PMCID: PMC10226337 DOI: 10.1177/03946320231178131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 05/09/2023] [Indexed: 05/27/2023] Open
Abstract
Objectives: Cervical squamous cell carcinoma and cervical adenocarcinoma (CESC) are the second leading cause of deaths from malignant tumors in women, while their therapeutic and diagnostic aims are still finited. A growing body of evidence indicated that sphingosine-1-phosphate receptor 2 (S1PR2) plays essential roles in the occurrence and development about several human cancers. Nevertheless, the key mechanism and role mechanism of S1PR2 in CESC are still unclear.Methods: We first used Tissue Expression (GTEx) and Genotypic Cancer Genome Atlas (TCGA) data to perform pan-cancer analysis on the expression and prognosis of S1PR2, and found that S1PR2 may have a potential impact on CESC. To generate a protein-protein interaction (PPI) network using the STRING database. The clusterProfiler package is used for feature-rich analysis. The Tumor IMmune Estimation Resource was used to determine the connection between S1PR2 mRNA expression and immune infiltrates. Results: S1PR2 expression in CESC tissues was down-regulated compared with adjacent normal tissues. Kaplan-Meier analysis indicated that compared with patients with high expression of S1PR2, CESC patients with low S1PR2 expression had a worse prognosis. Reduced S1PR2 expression is associated with patients with high clinical stage, more histological types of squamous cell carcinoma, and poor primary treatment outcomes. The receiver operating characteristic curve of S1PR2 was 0.870. Correlation analysis showed that the mRNA expression of S1PR2 was related to immune infiltrates and tumor purity.Conclusion: Down-regulated S1PR2 expression is related to poor survival and immune infiltration in CESC. S1PR2 is a potential biomarker for poor prognosis and as a potential target for CESC immune therapy.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Emergency Medicine, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Haichuan Wang
- Department of General Surgery, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Jie Lu
- Department of General Surgery, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Qiang Lv
- Department of General Surgery, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Bei Yun
- Department of General Surgery, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Zhiru Ge
- Department of Cardiology, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| | - Li Yan
- Department of Cardiology, Pudong New District Gongli Hospital of
Shanghai, Shanghai, China
| |
Collapse
|
3
|
Stamatellos VP, Siafis S, Papazisis G. Disease-modifying agents for multiple sclerosis and the risk for reporting cancer: A disproportionality analysis using the US Food and Drug Administration Adverse Event Reporting System database. Br J Clin Pharmacol 2021; 87:4769-4779. [PMID: 33998034 DOI: 10.1111/bcp.14916] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/25/2021] [Accepted: 05/04/2021] [Indexed: 11/27/2022] Open
Abstract
AIM While the efficacy of disease-modifying therapies (DMTs) for patients with multiple sclerosis (MS) is established, little is known about their long-term safety. Cancer-risk after DMT use remains unclear. This study aimed to investigate whether the prescription of DMTs for patients with MS increases the risk of reporting cancer. METHODS Data from the Food and Drug Administration Adverse Event Reporting System were extracted from 2004 to 2020. After data cleaning, the crude and adjusted reported odds ratios (cROR and aROR) for cancer were calculated for DMTs with Interferon beta-1a as the reference drug. Sensitivity analyses investigated the group of reports with multiple registered DMTs, the effect of indication restriction and the results when using the rest of the DMTs as reference. RESULTS For malignant tumours, aROR (95% confidence interval [CI]) values were Cladribine 0.46 (0.18-0.95), Dimethyl fumarate 0.30 (0.27-0.34), Fingolimod 0.61 (0.53-0.70), Glatiramer 0.50 (0.43-0.58), Alemtuzumab 0.84 (0.64-1.08), Interferon beta-1b 0.49 (0.42-0.56), Natalizumab 0.36 (0.34-0.39), Ocrelizumab 0.48 (0.29-0.74), Peginterferon beta-1a 0.35 (0.26-0.48), Siponimod 0.89 (0.47-1.54) and Teriflunomide 0.25(0.21-0.30) adjusted to age, gender and concomitant medications. In the sensitivity analysis, when the rest of the drugs were used as a reference, Interferon beta-1a and Peginterferon beta-1a had aROR (95% CI) 2.60 (2.47-2.74, P < .001) and Alemtuzumab had aROR 1.47 (1.13-1.88, I = .003). CONCLUSIONS No safety signal for increased cancer risk was detected among the approved DMTs. A potential safety signal detected in the sensitivity analysis concerning Interferon beta-1a and Alemtuzumab requires further evaluation with more robust evidence.
Collapse
Affiliation(s)
| | - Spyridon Siafis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Psychiatry and Psychotherapy, School of Medicine, Technical University of Munich, Munich, Germany
| | - Georgios Papazisis
- Department of Clinical Pharmacology, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
4
|
Qu SL, Chen L, Wen XS, Zuo JP, Wang XY, Lu ZJ, Yang YF. Suppression of Th17 cell differentiation via sphingosine-1-phosphate receptor 2 by cinnamaldehyde can ameliorate ulcerative colitis. Biomed Pharmacother 2021; 134:111116. [PMID: 33341041 DOI: 10.1016/j.biopha.2020.111116] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 12/03/2020] [Accepted: 12/04/2020] [Indexed: 12/12/2022] Open
Abstract
Ulcerative colitis (UC) is chronic disease characterized by diffuse inflammation of the mucosa of the colon and rectum. Although the etiology is unknown, dysregulation of the intestinal mucosal immune system is closely related to UC. Cinnamaldehyde (CA) is a major active compound from cinnamon, is known as its anti-inflammatory and antibacterial. However, little research focused on its regulatory function on immune cells in UC. Therefore, we set out to explore the modulating effects of CA on immune cells in UC. We found that CA reduced the progression of colitis through controlling the production of proinflammatory cytokines and inhibiting the proportion of Th17 cells. Furthermore, the liquid chromatography-mass spectrometry (LC-MS) method was employed for analyzing and differentiating metabolites, data showed that sphingolipid pathway has a great influence on the effect of CA on UC. Meanwhile, sphingosine-1-phosphate receptor 2 (S1P2) and Rho-GTP protein levels were downregulated in colonic tissues after CA treatment. Moreover, in vitro assays showed that CA inhibited Th17 cell differentiation and downregulated of S1P2 and Rho-GTP signaling. Notably, we found that treatment with S1P2 antagonist (JTE-013) weakened the inhibitory effect of CA on Th17 cells. Furthermore, S1P2 deficiency (S1P2-/-) blocked the effect of CA on Th17 cell differentiation. In addition, CA can also improve inflammation via lncRNA H19 and MIAT. To sum up, this study provides clear evidence that CA can ameliorate ulcerative colitis through suppressing Th17 cells via S1P2 pathway and regulating lncRNA H19 and MIAT, which further supports S1P2 as a potential drug target for immunity-mediated UC.
Collapse
Affiliation(s)
- Shu-Lan Qu
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Long Chen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xue-Shan Wen
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Jian-Ping Zuo
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Laboratory of Anti-inflammation and Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xiao-Yu Wang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; Laboratory of Anti-inflammation and Immunopharmacology, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Zhi-Jie Lu
- Department of Anesthesiology and Intensive Care Unit, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, 200438, China.
| | - Yi-Fu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
5
|
Petti L, Rizzo G, Rubbino F, Elangovan S, Colombo P, Restelli S, Piontini A, Arena V, Carvello M, Romano B, Cavalleri T, Anselmo A, Ungaro F, D'Alessio S, Spinelli A, Stifter S, Grizzi F, Sgambato A, Danese S, Laghi L, Malesci A, Vetrano S. Unveiling role of sphingosine-1-phosphate receptor 2 as a brake of epithelial stem cell proliferation and a tumor suppressor in colorectal cancer. J Exp Clin Cancer Res 2020; 39:253. [PMID: 33225975 PMCID: PMC7682101 DOI: 10.1186/s13046-020-01740-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Sphingosine-1-phosphate receptor 2 (S1PR2) mediates pleiotropic functions encompassing cell proliferation, survival, and migration, which become collectively de-regulated in cancer. Information on whether S1PR2 participates in colorectal carcinogenesis/cancer is scanty, and we set out to fill the gap. METHODS We screened expression changes of S1PR2 in human CRC and matched normal mucosa specimens [N = 76]. We compared CRC arising in inflammation-driven and genetically engineered models in wild-type (S1PR2+/+) and S1PR2 deficient (S1PR2-/-) mice. We reconstituted S1PR2 expression in RKO cells and assessed their growth in xenografts. Functionally, we mimicked the ablation of S1PR2 in normal mucosa by treating S1PR2+/+ organoids with JTE013 and characterized intestinal epithelial stem cells isolated from S1PR2-/-Lgr5-EGFP- mice. RESULTS S1PR2 expression was lost in 33% of CRC; in 55%, it was significantly decreased, only 12% retaining expression comparable to normal mucosa. Both colitis-induced and genetic Apc+/min mouse models of CRC showed a higher incidence in size and number of carcinomas and/or high-grade adenomas, with increased cell proliferation in S1PR2-/- mice compared to S1PR2+/+ controls. Loss of S1PR2 impaired mucosal regeneration, ultimately promoting the expansion of intestinal stem cells. Whereas its overexpression attenuated cell cycle progression, it reduced the phosphorylation of AKT and augmented the levels of PTEN. CONCLUSIONS In normal colonic crypts, S1PR2 gains expression along with intestinal epithelial cells differentiation, but not in intestinal stem cells, and contrasts intestinal tumorigenesis by promoting epithelial differentiation, preventing the expansion of stem cells and braking their malignant transformation. Targeting of S1PR2 may be of therapeutic benefit for CRC expressing high Lgr5.
Collapse
Affiliation(s)
- Luciana Petti
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Giulia Rizzo
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Federica Rubbino
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Sudharshan Elangovan
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Piergiuseppe Colombo
- Department of Pathology, Humanitas Clinical, and Research Center-IRCCS, Milan, Italy
| | - Silvia Restelli
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Andrea Piontini
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Vincenzo Arena
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
| | - Michele Carvello
- Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Barbara Romano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Tommaso Cavalleri
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Achille Anselmo
- Flow Cytometry Core, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Federica Ungaro
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Silvia D'Alessio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Antonino Spinelli
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
- Colon and Rectal Surgery Unit, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
| | - Sanja Stifter
- Department of Pathology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | - Fabio Grizzi
- Department of Immunology and Inflammation, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Alessandro Sgambato
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Rome, Italy
- Centro di Riferimento Oncologico della Basilicata (IRCCS-CROB), Rionero in Vulture, Italy
| | - Silvio Danese
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
| | - Luigi Laghi
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Alberto Malesci
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy
- Laboratory of Molecular Gastroenterology, Department of Gastroenterology, Humanitas Clinical and Research Center IRCCS, Rozzano, Italy
| | - Stefania Vetrano
- IBD Center, Department of Gastroenterology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Italy.
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini, Pieve Emanuele, Italy.
| |
Collapse
|
6
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
7
|
Carrié L, Virazels M, Dufau C, Montfort A, Levade T, Ségui B, Andrieu-Abadie N. New Insights into the Role of Sphingolipid Metabolism in Melanoma. Cells 2020; 9:E1967. [PMID: 32858889 PMCID: PMC7565650 DOI: 10.3390/cells9091967] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/22/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Cutaneous melanoma is a deadly skin cancer whose aggressiveness is directly linked to its metastatic potency. Despite remarkable breakthroughs in term of treatments with the emergence of targeted therapy and immunotherapy, the prognosis for metastatic patients remains uncertain mainly because of resistances. Better understanding the mechanisms responsible for melanoma progression is therefore essential to uncover new therapeutic targets. Interestingly, the sphingolipid metabolism is dysregulated in melanoma and is associated with melanoma progression and resistance to treatment. This review summarises the impact of the sphingolipid metabolism on melanoma from the initiation to metastatic dissemination with emphasis on melanoma plasticity, immune responses and resistance to treatments.
Collapse
Affiliation(s)
- Lorry Carrié
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Mathieu Virazels
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Carine Dufau
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Anne Montfort
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Thierry Levade
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
- Laboratoire de Biochimie Métabolique, CHU, 31059 Toulouse, France
| | - Bruno Ségui
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| | - Nathalie Andrieu-Abadie
- Centre de Recherches en Cancérologie de Toulouse, Equipe Labellisée Fondation ARC, Université Fédérale de Toulouse Midi-Pyrénées, Université Toulouse III Paul-Sabatier, Inserm 1037, 2 avenue Hubert Curien, CS 53717, 31037 Toulouse CEDEX 1, France; (L.C.); (M.V.); (C.D.); (A.M.); (T.L.); (B.S.)
| |
Collapse
|
8
|
Pang M, Li C, Zheng D, Wang Y, Wang J, Zhang W, Li F, Jing H. S1PR2 Knockdown Promotes Migration and Invasion in Multiple Myeloma Cells via NF-κB Activation. Cancer Manag Res 2020; 12:7857-7865. [PMID: 32922084 PMCID: PMC7457837 DOI: 10.2147/cmar.s237330] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 08/04/2020] [Indexed: 01/10/2023] Open
Abstract
Background The presence of circulating plasma cells (cPCs) was associated with a worse prognosis in multiple myeloma patients. However, the underlying mechanisms involved in the migration and invasion of bone marrow myeloma cells (BMMCs) to cPCs remains unclear. Here, we investigate the possible factors related to hematogenous myeloma cell dissemination and potential regulatory mechanisms. Methods BMMCs and cPCs of five extramedullary plasmacytoma (EMP) patients were selected for single cell RNA sequencing, We found that the expression level of sphingosine-1-phosphate receptor 2 (S1RP2) was lower in cPCs compared with that in BMMCs. Then, we investigated the effect of S1PR2 in cell migration and invasion through pharmacologic inhibition with a S1PR2-selective antagonist JTE-013 or knockdown of S1PR2 expression in MM cell line U266. Results The results showed that S1PR2 inhibition with JTE-013 or S1PR2-shRNA significantly promoted cell migration and invasion in U266 cells. We measured the expression of invasion-related proteins by Western blot and found that knockdown of S1PR2 could reduce MMP-9 expression in U266 cells. Furthermore, we found NF-κB pathway may mediate the inhibition effects of S1PR2 on cell migration and invasion in MM cells. Conclusion Our findings demonstrated that S1PR2 downregulation may contribute to the initial extramedullary translocation by promoting cell migration and invasion through NF-κB pathway activation.
Collapse
Affiliation(s)
- Meng Pang
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Chunyuan Li
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Dong Zheng
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Ying Wang
- Department of Immunology, Key Laboratory of Medical Immunology of Ministry of Health, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, People's Republic of China
| | - Jing Wang
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Weilong Zhang
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Fang Li
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| | - Hongmei Jing
- Department of Hematology, Third Hospital of Peking University, Beijing, People's Republic of China
| |
Collapse
|
9
|
Abstract
As basic research into GPCR signaling and its association with disease has come into fruition, greater clarity has emerged with regards to how these receptors may be amenable to therapeutic intervention. As a diverse group of receptor proteins, which regulate a variety of intracellular signaling pathways, research in this area has been slow to yield tangible therapeutic agents for the treatment of a number of diseases including cancer. However, recently such research has gained momentum based on a series of studies that have sought to define GPCR proteins dynamics through the elucidation of their crystal structures. In this chapter, we define the approaches that have been adopted in developing better therapeutics directed against the specific parts of the receptor proteins, such as the extracellular and the intracellular domains, including the ligands and auxiliary proteins that bind them. Finally, we also briefly outline how GPCR-derived signaling transduction pathways hold great potential as additional targets.
Collapse
Affiliation(s)
- Surinder M Soond
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.
| | - Andrey A Zamyatnin
- Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation.
| |
Collapse
|
10
|
Park KM, Wang JW, Yoo YM, Choi MJ, Hwang KC, Jeung EB, Jeong YW, Hwang WS. Sphingosine-1-phosphate (S1P) analog phytosphingosine-1-phosphate (P1P) improves the in vitro maturation efficiency of porcine oocytes via regulation of oxidative stress and apoptosis. Mol Reprod Dev 2019; 86:1705-1719. [PMID: 31490595 DOI: 10.1002/mrd.23264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/03/2019] [Indexed: 12/11/2022]
Abstract
Phytosphingosine-1-phosphate (P1P) is a signaling sphingolipid that regulates various physiological activities. However, little is known about the effect of P1P in the context of reproduction. Thus, we aimed to investigate the influence of P1P on oocyte maturation during porcine in vitro maturation (IVM). Here, we report the expression of S1PR1-3 among P1P receptors (S1PR1-4) in cumulus cells and oocytes. When P1P was administered at concentrations of 10, 50, 100, and 1,000 nM during IVM, the metaphase II rate was significantly increased in the 1,000 nM (1 μM) P1P treatment group. Maturation rate improvement by P1P supplementation was observed only in the presence of epidermal growth factor (EGF). Oocytes under the influence of P1P showed decreased intracellular reactive oxygen species levels but no significant differences in glutathione levels. In our molecular studies, P1P treatment upregulated gene expression involved in cumulus expansion (Has2 and EGF), antioxidant enzymes (SOD3 and Cat), and developmental competence (Oct4) while activating extracellular signal-regulated kinase1/2 and Akt signaling. P1P treatment also influenced oocyte survival by shifting the ratio of Bcl-2 to Bax while inactivating JNK signaling. We further demonstrated that oocytes matured with P1P displayed significantly higher developmental competence (cleavage and blastocyst [BL] formation rate) and greater BL quality (total cell number and the ratio of apoptotic cells) when activated via parthenogenetic activation (PA) and in vitro fertilization. Despite the low levels of endogenous P1P found in animals, exogenous P1P influenced animal reproduction, as shown by increased porcine oocyte maturation as well as preimplantation embryo development. This study and its findings are potentially relevant for both human and animal-assisted reproduction.
Collapse
Affiliation(s)
- Kyu-Mi Park
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Jae Woong Wang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Yeong-Min Yoo
- Laboratory of Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | | | - Kyu Chan Hwang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Eui-Bae Jeung
- Laboratory of Biochemistry and Molecular Biology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Yeon Woo Jeong
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| | - Woo Suk Hwang
- Sooam Biotech Research Foundation, Guro-gu, Seoul, Republic of Korea
| |
Collapse
|
11
|
Diring J, Mouilleron S, McDonald NQ, Treisman R. RPEL-family rhoGAPs link Rac/Cdc42 GTP loading to G-actin availability. Nat Cell Biol 2019; 21:845-855. [PMID: 31209295 PMCID: PMC6960015 DOI: 10.1038/s41556-019-0337-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 04/29/2019] [Indexed: 12/29/2022]
Abstract
RPEL proteins, which contain the G-actin-binding RPEL motif, coordinate cytoskeletal processes with actin dynamics. We show that the ArhGAP12- and ArhGAP32-family GTPase-activating proteins (GAPs) are RPEL proteins. We determine the structure of the ArhGAP12/G-actin complex, and show that G-actin contacts the RPEL motif and GAP domain sequences. G-actin inhibits ArhGAP12 GAP activity, and this requires the G-actin contacts identified in the structure. In B16 melanoma cells, ArhGAP12 suppresses basal Rac and Cdc42 activity, F-actin assembly, invadopodia formation and experimental metastasis. In this setting, ArhGAP12 mutants defective for G-actin binding exhibit more effective downregulation of Rac GTP loading following HGF stimulation and enhanced inhibition of Rac-dependent processes, including invadopodia formation. Potentiation or disruption of the G-actin/ArhGAP12 interaction, by treatment with the actin-binding drugs latrunculin B or cytochalasin D, has corresponding effects on Rac GTP loading. The interaction of G-actin with RPEL-family rhoGAPs thus provides a negative feedback loop that couples Rac activity to actin dynamics.
Collapse
Affiliation(s)
- Jessica Diring
- Signalling and Transcription Group, The Francis Crick Institute, London, UK
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Neil Q McDonald
- Signalling and Structural Biology Group, The Francis Crick Institute, London, UK
- Institute of Structural and Molecular Biology, Department of Biological Sciences, Birkbeck College, London, UK
| | - Richard Treisman
- Signalling and Transcription Group, The Francis Crick Institute, London, UK.
| |
Collapse
|
12
|
Palangi A, Shakhssalim N, Parvin M, Bayat S, Allameh A. Differential expression of S1P receptor subtypes in human bladder transitional cell carcinoma. Clin Transl Oncol 2019; 21:1240-1249. [PMID: 30712233 DOI: 10.1007/s12094-019-02049-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Accepted: 01/16/2019] [Indexed: 12/13/2022]
Abstract
PURPOSE Sphingosine 1 phosphate (S1P), S1P receptors (S1PRs) and their signaling pathways play an important role in the fate of cancer cells. The expression pattern of S1PR subtypes (S1PR1-S1PR5) may alter in cancer development stages, depending on the origin and the pathologic features of tumors. The present study aimed to examine the relationship between plasma S1P levels and the expression of S1PR subtypes in bladder tumors. METHODS/PATIENTS These changes were evaluated in terms of the pathologic grades and stages of human bladder cancer samples. For this, tumor biopsies from 41 new bladder cancer patients as well as 26 normal-looking bladder tissues were collected and processed for immunohistochemistry (IHC) and quantitative real-time RT-PCR of S1PR subtypes. Plasma S1P level was measured using liquid chromatography-tandem mass spectrometry (LC-MS/MS). RESULTS The results show that tissue S1PR1, S1PR2 and S1PR3 are over-expressed in all tumors regardless of their pathological grade (~ 3, ~ 6 and ~ 104 folds, respectively). These results were corroborated by IHC data showing accumulation of S1PR subtypes 1 and 2 in the tissues. Plasma S1P in the plasma samples from patients was in the range of control samples (Controls; 256 ± 47; patients, 270 ± 41). CONCLUSIONS Overexpression of S1PR1, S1PR2 and S1PR3 in bladder tumor biopsies which were corroborated with the pathological grades and stages may suggest that S1PR profile in tumor biopsies is a promising marker in the diagnosis of bladder carcinoma.
Collapse
Affiliation(s)
- A Palangi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - N Shakhssalim
- Urology and Nephrology Research Center, Shahid Labbafinejad Medical Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - M Parvin
- Department of Pathology, Shahid Labbafinejad Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - S Bayat
- Department of Biochemistry, Faculty of Science, Payam Noor University Tehran Unit, Tehran, Iran
| | - A Allameh
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
13
|
Abstract
While normal angiogenesis is critical for development and tissue growth, pathological angiogenesis is important for the growth and spread of cancers by supplying nutrients and oxygen as well as providing a conduit for distant metastasis. The interaction among extracellular matrix molecules, tumor cells, endothelial cells, fibroblasts, and immune cells is critical in pathological angiogenesis, in which various angiogenic growth factors, chemokines, and lipid mediators produced from these cells as well as hypoxic microenvironment promote angiogenesis by regulating expression and/or activity of various related genes. Sphingosine 1-phosphate and lysophosphatidic acid, bioactive lipid mediators which act via specific G protein-coupled receptors, play critical roles in angiogenesis. In addition, other lipid mediators including prostaglandin E2, lipoxin, and resolvins are produced in a stimulus-dependent manner and have pro- or anti-angiogenic effects, presumably through their specific GPCRs. Dysregulated lipid mediator signaling pathways are observed in the contxt of some tumors. This review will focus on LPA and S1P, two bioactive lipid mediators in their regulation of angiogenesis and cell migration that are critical for tumor growth and spread.
Collapse
Affiliation(s)
- Yu Hisano
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Timothy Hla
- Vascular Biology Program, Boston Children's Hospital, Department of Surgery, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, United States.
| |
Collapse
|
14
|
Selenium unmasks protective iron armor: A possible defense against cutaneous inflammation and cancer. Biochim Biophys Acta Gen Subj 2018; 1862:2518-2527. [PMID: 29852199 DOI: 10.1016/j.bbagen.2018.05.018] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 05/09/2018] [Accepted: 05/23/2018] [Indexed: 01/06/2023]
Abstract
BACKGROUND A link between selenium deficiency and inflammatory skin diseases have been noted by many, but this link is still not well understood. We have previously studied the efficacy of ceramide analogs, based on the fire ant venom Solenopsin A, against our psoriasis animal model. Treatment of animals with solenopsin analogs resulted in significantly improved skin as well as in a coordinate downregulation of selenoproteins, namely Glutathione Peroxidase 4 (GPX4). We thus hypothesize that ferroptosis may be a physiologic process that may protect the skin from both inflammatory and neoplastic processes. METHODS We analyze and compare gene expression profiles in the GEO database from clinical skin samples taken from healthy patients and psoriasis patients (both involved and noninvolved skin lesions). We validated the gene expression results against a second, independent, cohort from the GEO database. RESULTS Significant reduction in gene expression of GPX4, elevated expression of Nrf2 downstream targets, and expression profiles mirroring erastin-inhibition of Cystine/Glutamate Antiporter-System XC activity in psoriatic skin lesions, compared to both noninvolved skin and healthy patient samples, suggest an innately inducible mechanism of ferroptosis. CONCLUSIONS We present data that may indicate selenoproteins, particularly GPX4, in resolving inflammation and skin cancer, including the novel hypothesis that the human organism may downregulate GPX4 and reactive oxygen (REDOX) regulating proteins in the skin as a way of resolving psoriasis and nonmelanoma skin cancer through increased reactive oxygen species. Further studies are needed to investigate ferroptosis as a possible physiologic mechanism for eliminating inflammatory and malignant tissues. GENERAL SIGNIFICANCE This study provides a fresh framework for understanding the seemingly contradictory effects of selenium supplementation. In addition, it offers a novel explanation of how physiologic upregulation of ferroptosis and downregulation of selenoprotein synthesis may mediate resolution of inflammation and carcinogenesis. This is of therapeutic significance.
Collapse
|
15
|
Affiliation(s)
- Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta 30322, GA, USA; Veterans Affairs Medical Center, Decatur 30322, GA, USA.
| |
Collapse
|
16
|
Sukocheva OA. Expansion of Sphingosine Kinase and Sphingosine-1-Phosphate Receptor Function in Normal and Cancer Cells: From Membrane Restructuring to Mediation of Estrogen Signaling and Stem Cell Programming. Int J Mol Sci 2018; 19:420. [PMID: 29385066 PMCID: PMC5855642 DOI: 10.3390/ijms19020420] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 01/21/2018] [Accepted: 01/24/2018] [Indexed: 02/05/2023] Open
Abstract
Sphingolipids, sphingolipid metabolizing enzymes, and their receptors network are being recognized as part of the signaling mechanisms, which govern breast cancer cell growth, migration, and survival during chemotherapy treatment. Approximately 70% of breast cancers are estrogen receptor (ER) positive and, thus, rely on estrogen signaling. Estrogen activates an intracellular network composed of many cytoplasmic and nuclear mediators. Some estrogen effects can be mediated by sphingolipids. Estrogen activates sphingosine kinase 1 (SphK1) and amplifies the intracellular concentration of sphingosine-1-phosphate (S1P) in breast cancer cells during stimulation of proliferation and survival. Specifically, Estrogen activates S1P receptors (S1PR) and induces growth factor receptor transactivation. SphK, S1P, and S1PR expression are causally associated with endocrine resistance and progression to advanced tumor stages in ER-positive breast cancers in vivo. Recently, the network of SphK/S1PR was shown to promote the development of ER-negative cancers and breast cancer stem cells, as well as stimulating angiogenesis. Novel findings confirm and broaden our knowledge about the cross-talk between sphingolipids and estrogen network in normal and malignant cells. Current S1PRs therapeutic inhibition was indicated as a promising chemotherapy approach in non-responsive and advanced malignancies. Considering that sphingolipid signaling has a prominent role in terminally differentiated cells, the impact should be considered when designing specific SphK/S1PR inhibitors. This study analyzes the dynamic of the transformation of sphingolipid axis during a transition from normal to pathological condition on the level of the whole organism. The sphingolipid-based mediation and facilitation of global effects of estrogen were critically accented as a bridging mechanism that should be explored in cancer prevention.
Collapse
Affiliation(s)
- Olga A Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, SA 5042, Australia.
| |
Collapse
|
17
|
Solid lipid nanoparticles improve octyl gallate antimetastatic activity and ameliorate its renal and hepatic toxic effects. Anticancer Drugs 2017; 28:977-988. [PMID: 28746057 DOI: 10.1097/cad.0000000000000539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Metastasis is the main cause of cancer-related death and requires the development of effective treatments with reduced toxicity and effective anticancer activity. Gallic acid derivatives have shown significant biological properties including antitumoral activity as shown in a previous study with octyl gallate (G8) in vitro. Thus, the aim of this work was to evaluate the antimetastatic effect of free and solid lipid nanoparticle-loaded G8 in mice in a lung metastasis model. Animals inoculated with melanoma cells presented metastasis in lungs, which was significantly inhibited by treatment with G8 and solid lipid nanoparticle-loaded G8, named G8-NVM. However, G8-treated mice showed an increase in several toxicological parameters, which were almost completely circumvented by G8-NVM treatment. This study supports the need for pharmacological studies on new potential medicinal plants to treat cancer and can provide new perspectives on using nanotechnology to improve biological activities while decreasing the chemotherapy toxicological effects of anticancer drugs.
Collapse
|
18
|
Hatoum D, Haddadi N, Lin Y, Nassif NT, McGowan EM. Mammalian sphingosine kinase (SphK) isoenzymes and isoform expression: challenges for SphK as an oncotarget. Oncotarget 2017; 8:36898-36929. [PMID: 28415564 PMCID: PMC5482707 DOI: 10.18632/oncotarget.16370] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 03/02/2017] [Indexed: 12/16/2022] Open
Abstract
The various sphingosine kinase (SphK) isoenzymes (isozymes) and isoforms, key players in normal cellular physiology, are strongly implicated in cancer and other diseases. Mutations in SphKs, that may justify abnormal physiological function, have not been recorded. Nonetheless, there is a large and growing body of evidence demonstrating the contribution of gain or loss of function and the imbalance in the SphK/S1P rheostat to a plethora of pathological conditions including cancer, diabetes and inflammatory diseases. SphK is expressed as two isozymes SphK1 and SphK2, transcribed from genes located on different chromosomes and both isozymes catalyze the phosphorylation of sphingosine to S1P. Expression of each SphK isozyme produces alternately spliced isoforms. In recent years the importance of the contribution of SpK1 expression to treatment resistance in cancer has been highlighted and, additionally, differences in treatment outcome appear to also be dependent upon SphK isoform expression. This review focuses on an exciting emerging area of research involving SphKs functions, expression and subcellular localization, highlighting the complexity of targeting SphK in cancer and also comorbid diseases. This review also covers the SphK isoenzymes and isoforms from a historical perspective, from their first discovery in murine species and then in humans, their role(s) in normal cellular function and in disease processes, to advancement of SphK as an oncotarget.
Collapse
Affiliation(s)
- Diana Hatoum
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Nahal Haddadi
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Yiguang Lin
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Najah T. Nassif
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| | - Eileen M. McGowan
- School of Life Sciences, University of Technology Sydney, Ultimo, Sydney, NSW 2007, Australia
| |
Collapse
|
19
|
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
|
20
|
Blankenbach KV, Schwalm S, Pfeilschifter J, Meyer Zu Heringdorf D. Sphingosine-1-Phosphate Receptor-2 Antagonists: Therapeutic Potential and Potential Risks. Front Pharmacol 2016; 7:167. [PMID: 27445808 PMCID: PMC4914510 DOI: 10.3389/fphar.2016.00167] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 06/03/2016] [Indexed: 12/26/2022] Open
Abstract
The sphingosine-1-phosphate (S1P) signaling system with its specific G-protein-coupled S1P receptors, the enzymes of S1P metabolism and the S1P transporters, offers a multitude of promising targets for drug development. Until today, drug development in this area has nearly exclusively focused on (functional) antagonists at the S1P1 receptor, which cause a unique phenotype of immunomodulation. Accordingly, the first-in class S1P1 receptor modulator, fingolimod, has been approved for the treatment of relapsing-remitting multiple sclerosis, and novel S1P1 receptor (functional) antagonists are being developed for autoimmune and inflammatory diseases such as psoriasis, inflammatory bowel disease, lupus erythematodes, or polymyositis. Besides the S1P1 receptor, also S1P2 and S1P3 are widely expressed and regulate many diverse functions throughout the body. The S1P2 receptor, in particular, often exerts cellular functions which are opposed to the functions of the S1P1 receptor. As a consequence, antagonists at the S1P2 receptor have the potential to be useful in a contrasting context and different areas of indication compared to S1P1 antagonists. The present review will focus on the therapeutic potential of S1P2 receptor antagonists and discuss their opportunities as well as their potential risks. Open questions and areas which require further investigations will be emphasized in particular.
Collapse
Affiliation(s)
- Kira V Blankenbach
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| | - Dagmar Meyer Zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Klinikum der Johann Wolfgang Goethe-Universität Frankfurt am Main, Germany
| |
Collapse
|
21
|
Kitada Y, Kajita K, Taguchi K, Mori I, Yamauchi M, Ikeda T, Kawashima M, Asano M, Kajita T, Ishizuka T, Banno Y, Kojima I, Chun J, Kamata S, Ishii I, Morita H. Blockade of Sphingosine 1-Phosphate Receptor 2 Signaling Attenuates High-Fat Diet-Induced Adipocyte Hypertrophy and Systemic Glucose Intolerance in Mice. Endocrinology 2016; 157:1839-51. [PMID: 26943364 PMCID: PMC4870879 DOI: 10.1210/en.2015-1768] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Sphingosine 1-phosphate (S1P) is known to regulate insulin resistance in hepatocytes, skeletal muscle cells, and pancreatic β-cells. Among its 5 cognate receptors (S1pr1-S1pr5), S1P seems to counteract insulin signaling and confer insulin resistance via S1pr2 in these cells. S1P may also regulate insulin resistance in adipocytes, but the S1pr subtype(s) involved remains unknown. Here, we investigated systemic glucose/insulin tolerance and phenotypes of epididymal adipocytes in high-fat diet (HFD)-fed wild-type and S1pr2-deficient (S1pr2(-/-)) mice. Adult S1pr2(-/-) mice displayed smaller body/epididymal fat tissue weights, but the differences became negligible after 4 weeks with HFD. However, HFD-fed S1pr2(-/-) mice displayed better scores in glucose/insulin tolerance tests and had smaller epididymal adipocytes that expressed higher levels of proliferating cell nuclear antigen than wild-type mice. Next, proliferation/differentiation of 3T3-L1 and 3T3-F442A preadipocytes were examined in the presence of various S1pr antagonists: JTE-013 (S1pr2 antagonist), VPC-23019 (S1pr1/S1pr3 antagonist), and CYM-50358 (S1pr4 antagonist). S1P or JTE-013 treatment of 3T3-L1 preadipocytes potently activated their proliferation and Erk phosphorylation, whereas VPC-23019 inhibited both of these processes, and CYM-50358 had no effects. In contrast, S1P or JTE-013 treatment inhibited adipogenic differentiation of 3T3-F442A preadipocytes, whereas VPC-23019 activated it. The small interfering RNA knockdown of S1pr2 promoted proliferation and inhibited differentiation of 3T3-F442A preadipocytes, whereas that of S1pr1 acted oppositely. Moreover, oral JTE-013 administration improved glucose tolerance/insulin sensitivity in ob/ob mice. Taken together, S1pr2 blockade induced proliferation but suppressed differentiation of (pre)adipocytes both in vivo and in vitro, highlighting a novel therapeutic approach for obesity/type 2 diabetes.
Collapse
Affiliation(s)
- Yoshihiko Kitada
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Kazuo Kajita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Koichiro Taguchi
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Ichiro Mori
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Masahiro Yamauchi
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Takahide Ikeda
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Mikako Kawashima
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Motochika Asano
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Toshiko Kajita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Tatsuo Ishizuka
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Yoshiko Banno
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Itaru Kojima
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Jerold Chun
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Shotaro Kamata
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Isao Ishii
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| | - Hiroyuki Morita
- Department of General Internal Medicine (Y.K., K.K., K.T., I.M., M.Y., T.Ik., M.K., M.A., T.K., H.M.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of General Internal Medicine and Rheumatology (T.Is.), Gifu Municipal Hospital, Gifu 500-8513, Japan; Department of Dermatology (Y.B.), Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Laboratory of Cell Physiology (I.K.), Institute for Molecular and Cellular Regulation, Gunma University, Gunma 371-8512, Japan; Molecular and Cellular Neuroscience Department (J.C.), Dorris Neuroscience Center, The Scripps Research Institute, La Jolla, California 92037; and Department of Biochemistry (S.K., I.I.), Keio University Graduate School of Pharmaceutical Sciences, Tokyo 105-8512, Japan
| |
Collapse
|
22
|
Ren S, Shao Y, Zhao X, Hong CS, Wang F, Lu X, Li J, Ye G, Yan M, Zhuang Z, Xu C, Xu G, Sun Y. Integration of Metabolomics and Transcriptomics Reveals Major Metabolic Pathways and Potential Biomarker Involved in Prostate Cancer. Mol Cell Proteomics 2015; 15:154-63. [PMID: 26545398 DOI: 10.1074/mcp.m115.052381] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Indexed: 12/16/2022] Open
Abstract
Prostate cancer is a highly prevalent tumor affecting millions of men worldwide, but poor understanding of its pathogenesis has limited effective clinical management of patients. In addition to transcriptional profiling or transcriptomics, metabolomics is being increasingly utilized to discover key molecular changes underlying tumorigenesis. In this study, we integrated transcriptomics and metabolomics to analyze 25 paired human prostate cancer tissues and adjacent noncancerous tissues, followed by further validation of our findings in an additional cohort of 51 prostate cancer patients and 16 benign prostatic hyperplasia patients. We found several altered pathways aberrantly expressed at both metabolic and transcriptional levels, including cysteine and methionine metabolism, nicotinamide adenine dinucleotide metabolism, and hexosamine biosynthesis. Additionally, the metabolite sphingosine demonstrated high specificity and sensitivity for distinguishing prostate cancer from benign prostatic hyperplasia, particularly for patients with low prostate specific antigen level (0-10 ng/ml). We also found impaired sphingosine-1-phosphate receptor 2 signaling, downstream of sphingosine, representing a loss of tumor suppressor gene and a potential key oncogenic pathway for therapeutic targeting. By integrating metabolomics and transcriptomics, we have provided both a broad picture of the molecular perturbations underlying prostate cancer and a preliminary study of a novel metabolic signature, which may help to discriminate prostate cancer from normal tissue and benign prostatic hyperplasia.
Collapse
Affiliation(s)
- Shancheng Ren
- From the ‡Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yaping Shao
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Xinjie Zhao
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Christopher S Hong
- ¶Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Fubo Wang
- From the ‡Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xin Lu
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Jia Li
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Guozhu Ye
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Min Yan
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China
| | - Zhengping Zhuang
- ¶Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD
| | - Chuanliang Xu
- From the ‡Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Guowang Xu
- §Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023 Dalian, China,
| | - Yinghao Sun
- From the ‡Department of Urology, Shanghai Changhai Hospital, Second Military Medical University, Shanghai, China,
| |
Collapse
|
23
|
Binder BYK, Williams PA, Silva EA, Leach JK. Lysophosphatidic Acid and Sphingosine-1-Phosphate: A Concise Review of Biological Function and Applications for Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:531-42. [PMID: 26035484 DOI: 10.1089/ten.teb.2015.0107] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The presentation and controlled release of bioactive signals to direct cellular growth and differentiation represents a widely used strategy in tissue engineering. Historically, work in this field has primarily focused on the delivery of large cytokines and growth factors, which can be costly to manufacture and difficult to deliver in a sustained manner. There has been a marked increase over the past decade in the pursuit of lipid mediators due to their wide range of effects over multiple cell types, low cost, and ease of scale-up. Lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) are two bioactive lysophospholipids (LPLs) that have gained attention for use as pharmacological agents in tissue engineering applications. While these lipids can have similar effects on cellular response, they possess distinct chemical backbones, mechanisms of synthesis and degradation, and signaling pathways using a discrete set of G-protein-coupled receptors (GPCRs). LPA and S1P predominantly act extracellularly on their GPCRs and can directly regulate cell survival, differentiation, cytokine secretion, proliferation, and migration--each of the important functions that must be considered in regenerative medicine. In addition to these potent physiological functions, these LPLs play pivotal roles in a number of pathophysiological processes. To capitalize on the promise of these molecules in tissue engineering, these lipids have been incorporated into biomaterials for in vivo delivery. Here, we survey the effects of LPA and S1P on both cellular- and tissue-level phenotypes, with an eye toward regulating stem/progenitor cell growth and differentiation. In particular, we examine work that has translational applications for cell-based tissue engineering strategies in promoting cell survival, bone and cartilage engineering, and therapeutic angiogenesis.
Collapse
Affiliation(s)
- Bernard Y K Binder
- 1 Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Priscilla A Williams
- 1 Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - Eduardo A Silva
- 1 Department of Biomedical Engineering, University of California , Davis, Davis, California
| | - J Kent Leach
- 1 Department of Biomedical Engineering, University of California , Davis, Davis, California.,2 Department of Orthopaedic Surgery, School of Medicine, University of California , Davis, Sacramento, California
| |
Collapse
|
24
|
Dória ML, Ribeiro AS, Wang J, Cotrim CZ, Domingues P, Williams C, Domingues MR, Helguero LA. Fatty acid and phospholipid biosynthetic pathways are regulated throughout mammary epithelial cell differentiation and correlate to breast cancer survival. FASEB J 2014; 28:4247-64. [PMID: 24970396 DOI: 10.1096/fj.14-249672] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This work combined gene and protein expression, gas chromatography-flame ionization detector, and hydrophilic interaction liquid chromatography-tandem mass spectrometry to compare lipid metabolism changes in undifferentiated/proliferating vs. functionally differentiated mammary epithelial cells (MECs) and to study their correlation to breast cancer survival. Sixty-eight genes involved in lipid metabolism were changed in MEC differentiation. Differentiated cells showed induction of Elovl6 (2-fold), Scd1 (4-fold), and Fads2 (2-fold), which correlated with increased levels of C16:1 n-7 and C18:1 n-9 (1.5-fold), C20:3 n-6 (2.5-fold), and C20:4 n-6 (6-fold) fatty acids (FAs) and more phospholipids (PLs) containing these species. Further, increased expression (2- to 3-fold) of genes in phosphatidylethanolamine (PE) de novo biosynthesis resulted in a 20% PE increase. Proliferating/undifferentiated cells showed higher C16:0 (1.7-fold) and C18:2 n-6 (4.2-fold) levels and more PLs containing C16:0 FAs [PC(16:0/16:1), PG(16:0/18:2), PG(16:0/18:1), and SM(16:0/18:0)]. Kaplan-Meier analysis of data from 3455 patients with breast cancer disclosed a positive correlation for 59% of genes expressed in differentiated MECs with better survival. PE biosynthesis and FA oxidation correlated with better prognosis in patients with breast cancer, including the basal-like subtype. Therefore, genes involved in mammary gland FA and PL metabolism and their resulting molecular species reflect the cellular proliferative ability and differentiation state and deserve further studies as potential markers of breast cancer progression
Collapse
Affiliation(s)
- M Luisa Dória
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| | - Ana S Ribeiro
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| | - Jun Wang
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - Cândida Z Cotrim
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| | - Pedro Domingues
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| | - Cecilia Williams
- Department of Biology and Biochemistry, Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, Texas, USA
| | - M Rosário Domingues
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| | - Luisa A Helguero
- Mass Spectrometry Centre, Organic Chemistry and Natural Products Research Unit, Department of Chemistry, Universidade de Aveiro, Campus de Santiago, Aveiro, Portugal; and
| |
Collapse
|
25
|
Adada M, Canals D, Hannun YA, Obeid LM. Sphingosine-1-phosphate receptor 2. FEBS J 2013; 280:6354-66. [PMID: 23879641 DOI: 10.1111/febs.12446] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/15/2013] [Accepted: 07/22/2013] [Indexed: 12/15/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a potent bioactive sphingolipid involved in cell proliferation, angiogenesis, inflammation and malignant transformation among other functions. S1P acts either directly on intracellular targets or activates G protein-coupled receptors, specifically five S1P receptors (S1PRs). The identified S1PRs differ in cellular and tissue distribution, and each is coupled to specific G proteins, which mediate unique functions. Here, we describe functional characteristics of all five receptors, emphasizing S1PR2, which is critical in the immune, nervous, metabolic, cardiovascular, musculoskeletal, and renal systems. This review also describes the role of this receptor in tumor growth and metastasis and suggests potential therapeutic avenues that exploit S1PR2.
Collapse
Affiliation(s)
- Mohamad Adada
- Department of Medicine, Stony Brook University, NY, USA
| | | | | | | |
Collapse
|
26
|
Stradner MH, Gruber G, Angerer H, Huber V, Setznagl D, Kremser ML, Moazedi-Fürst FC, Windhager R, Graninger WB. Sphingosine 1-phosphate counteracts the effects of interleukin-1β in human chondrocytes. ARTHRITIS AND RHEUMATISM 2013; 65:2113-22. [PMID: 23666803 PMCID: PMC3763206 DOI: 10.1002/art.37989] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 04/18/2013] [Accepted: 04/18/2013] [Indexed: 01/26/2023]
Abstract
OBJECTIVE The lipid mediator sphingosine 1-phosphate (S1P) is found in the synovial fluid of osteoarthritis (OA) patients. S1P protects bovine cartilage by counteracting the effects of interleukin-1β (IL-1β). This study was undertaken to examine the interaction of S1P and IL-1β in human OA chondrocytes. METHODS Human cartilage was obtained from patients undergoing total knee joint replacement. Chondrocytes were cultured in monolayer and treated with IL-1β and S1P. Expression of S1P receptor subtypes and genes involved in cartilage degradation was evaluated using real-time polymerase chain reaction, immunohistochemistry, and Western blotting. S1P signaling was evaluated using inhibitors of S1P receptors and small interfering RNA (siRNA) knockdown of the S1P2 receptor. Phosphorylation of MAP kinases and NF-κB in response to IL-1β and S1P was detected by Western blotting. RESULTS S1P2 was identified as the most prevalent S1P receptor subtype in human OA cartilage and chondrocytes in vitro. S1P reduced expression of inducible nitric oxide synthase (iNOS) in IL-1β-treated chondrocytes. Reduction of ADAMTS-4 and matrix metalloproteinase 13 expression by S1P correlated with S1P2 expression. Pharmacologic inhibition of the S1P2 receptor, but not the S1P1 and S1P3 receptors, abrogated the inhibition of iNOS expression. Similar results were observed using siRNA knockdown. S1P signaling inhibited IL-1β-induced phosphorylation of p38 MAPK. CONCLUSION In human chondrocytes, S1P reduces the induction of catabolic genes in the presence of IL-1β. Activation of the S1P2 receptor counteracts the detrimental phosphorylation of p38 MAPK by IL-1β.
Collapse
Affiliation(s)
- Martin H Stradner
- University of California at San Diego, La Jolla, California; Medical University of Graz, Graz, Austria.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Dual role of sphingosine kinase-1 in promoting the differentiation of dermal fibroblasts and the dissemination of melanoma cells. Oncogene 2013; 33:3364-73. [PMID: 23893239 DOI: 10.1038/onc.2013.303] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/14/2013] [Accepted: 06/18/2013] [Indexed: 12/14/2022]
Abstract
Despite progress in the understanding of the biology and genetics of melanoma, no effective treatment against this cancer is available. The adjacent microenvironment has an important role in melanoma progression. Defining the molecular signals that control the bidirectional dialog between malignant cells and the surrounding stroma is crucial for efficient targeted therapy. Our study aimed at defining the role of sphingosine-1-phosphate (S1P) in melanoma-stroma interactions. Transcriptomic analysis of human melanoma cell lines showed increased expression of sphingosine kinase-1 (SPHK1), the enzyme that produces S1P, as compared with normal melanocytes. Such an increase was also observed by immunohistochemistry in melanoma specimens as compared with nevi, and occurred downstream of ERK activation because of BRAF or NRAS mutations. Importantly, migration of melanoma cells was not affected by changes in SPHK1 activity in tumor cells, but was stimulated by comparable modifications of S1P-metabolizing enzymes in cocultured dermal fibroblasts. Reciprocally, incubation of fibroblasts with the conditioned medium from SPHK1-expressing melanoma cells resulted in their differentiation to myofibroblasts, increased production of matrix metalloproteinases and enhanced SPHK1 expression and activity. In vivo tumorigenesis experiments showed that the lack of S1P in the microenvironment prevented the development of orthotopically injected melanoma cells. Finally, local tumor growth and dissemination were enhanced more efficiently by coinjection of wild-type skin fibroblasts than by fibroblasts from Sphk1(-/-) mice. This report is the first to document that SPHK1/S1P modulates the communication between melanoma cells and dermal fibroblasts. Altogether, our findings highlight SPHK1 as a potential therapeutic target in melanoma progression.
Collapse
|
28
|
Orr Gandy KA, Adada M, Canals D, Carroll B, Roddy P, Hannun YA, Obeid LM. Epidermal growth factor-induced cellular invasion requires sphingosine-1-phosphate/sphingosine-1-phosphate 2 receptor-mediated ezrin activation. FASEB J 2013; 27:3155-66. [PMID: 23629860 DOI: 10.1096/fj.13-228460] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Ezrin, radixin, and moesin (ERM) proteins link cortical actin to the plasma membrane and coordinate cellular events that require cytoskeletal rearrangement, including cell division, migration, and invasion. While ERM proteins are involved in many important cellular events, the mechanisms regulating their function are not completely understood. Our laboratory previously identified reciprocal roles for the sphingolipids ceramide and sphingosine-1-phosphate (S1P) in the regulation of ERM proteins. We recently showed that ceramide-induced activation of PP1α leads to dephosphorylation and inactivation of ERM proteins, while S1P results in phosphorylation and activation of ERM proteins. Following these findings, we aimed to examine known inducers of the SK/S1P pathway and evaluate their ability to regulate ERM proteins. We examined EGF, a known inducer of the SK/S1P pathway, for its ability to regulate the ERM family of proteins. We found that EGF induces ERM c-terminal threonine phosphorylation via activation of the SK/S1P pathway, as this was prevented by siRNA knockdown or pharmacological inhibition of SK. Using pharmacological, as well as genetic, knockdown approaches, we determined that EGF induces ERM phosphorylation via activation of S1PR2. In addition, EGF led to cell polarization in the form of lamellipodia, and this occurred through a mechanism involving S1PR2-mediated phosphorylation of ezrin T567. EGF-induced cellular invasion was also found to be dependent on S1PR2-induced T567 ezrin phosphorylation, such that S1PR2 antagonist, JTE-013, and expression of a dominant-negative ezrin mutant prevented cellular invasion toward EGF. In this work, a novel mechanism of EGF-stimulated invasion is unveiled, whereby S1P-mediated activation of S1PR2 and phosphorylation of ezrin T567 is required.
Collapse
Affiliation(s)
- K Alexa Orr Gandy
- Department of Molecular and Cellular Biology and Pathobiology, Medical University of South Carolina, Charleston, South Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Takuwa N, Okamoto Y, Yoshioka K, Takuwa Y. Sphingosine-1-phosphate signaling and cardiac fibrosis. Inflamm Regen 2013. [DOI: 10.2492/inflammregen.33.096] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
30
|
Abstract
Cancer is frequently associated with activation of blood coagulation, which in turn has been suggested to promote tumor growth and metastasis. Indeed, low molecular weight heparin treatment significantly prolongs the survival of a wide variety of patients with cancer. Based on this notion that anticoagulant treatment seems to benefit cancer patients, recent experiments aimed to elucidate the importance of the natural anticoagulant protein C pathways in cancer progression. Interestingly, these experiments showed that the repeated administration of exogenous activated protein C limits cancer cell extravasation in experimental animal models. In line, reducing endogenous activated protein C activity dramatically increased the number of experimental metastasis. These data thus strongly suggest that exogenous activated protein C administration may be a novel therapeutic avenue to limit cancer metastasis thereby prolonging overall survival of cancer patients. The current review provides an overview of recent data on the role of the protein C pathway in cancer metastasis. It discusses the potential of activated protein C as a novel target to reduce cancer progression, it points to several limitations of activated protein C administration in the setting of cancer cell metastasis and it suggest zymogen protein C as an attractive alternative.
Collapse
Affiliation(s)
- C Arnold Spek
- Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, The Netherlands.
| | | |
Collapse
|
31
|
Takuwa Y, Okamoto Y, Yoshioka K, Takuwa N. Sphingosine-1-phosphate signaling in physiology and diseases. Biofactors 2012; 38:329-37. [PMID: 22674845 DOI: 10.1002/biof.1030] [Citation(s) in RCA: 281] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2012] [Accepted: 05/11/2012] [Indexed: 11/10/2022]
Abstract
Sphingosine-1-phosphate (S1P), which acts as both the extracellular and intracellular messenger, exerts pleiotropic biological activities including regulation of formation of the vasculature, vascular barrier integrity, and lymphocyte trafficking. Many of these S1P actions are mediated by five members of the G protein-coupled S1P receptors (S1P(1) -S1P(5) ) with overlapping but distinct coupling to heterotrimeric G proteins. The biological activities of S1P are based largely on the cellular actions of S1P on migration, adhesion, and proliferation. Notably, S1P often exhibits receptor subtype-specific, bimodal effects in these cellular actions. For example, S1P(1) mediates cell migration toward S1P, that is, chemotaxis, via G(i) /Rac pathway whereas S1P(2) mediates inhibition of migration toward a chemoattractant, that is, chemorepulsion, via G(12/13) /Rho pathway, which induces Rac inhibition. In addition, S1P(1) mediates stimulation of cell proliferation through the G(i) -mediated signaling pathways including phosphatidylinositol 3-kinase (PI3K)/Akt and ERK whereas S1P(2) mediates inhibition of cell proliferation through mechanisms involving G(12/13) /Rho/Rho kinase/PTEN-dependent Akt inhibition. These differential effects of S1P receptor subtypes on migration and proliferation lead to bimodal regulation of various biological responses. An observed biological response is likely determined by an integrated outcome of the counteracting signals input by S1P receptor subtypes. More recent studies identified the new intracellular targets of S1P including the inflammatory signaling molecule TRAF2 and histone deacetylases HDAC1 and HDAC2. These interactions of S1P regulate NF-κB activity and gene expression, respectively. Development of S1P receptor agonists and antagonists with improved receptor subtype-selectivity, inhibitors, or modulators of sphingolipid-metabolizing enzymes, and their optimal drug delivery system provide novel therapeutic tactics.
Collapse
Affiliation(s)
- Yoh Takuwa
- Department of Physiology, Kanazawa University School of Medicine, Kanazawa, Ishikawa, Japan.
| | | | | | | |
Collapse
|
32
|
Nakano T, Shimizu K, Kawashima O, Kamiyoshihara M, Kakegawa S, Sugano M, Ibe T, Nagashima T, Kaira K, Sunaga N, Ohtaki Y, Atsumi J, Takeyoshi I. Establishment of a human lung cancer cell line with high metastatic potential to multiple organs: gene expression associated with metastatic potential in human lung cancer. Oncol Rep 2012; 28:1727-35. [PMID: 22922681 DOI: 10.3892/or.2012.1972] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Accepted: 03/07/2012] [Indexed: 11/06/2022] Open
Abstract
Convenient and reliable multiple organ metastasis model systems might contribute to understanding the mechanism(s) of metastasis of lung cancer, which may lead to overcoming metastasis and improvement in the treatment outcome of lung cancer. We isolated a highly metastatic subline, PC14HM, from the human pulmonary adenocarcinoma cell line, PC14, using an in vivo selection method. The expression of 34,580 genes was compared between PC14HM and parental PC14 by cDNA microarray analysis. Among the differentially expressed genes, expression of four genes in human lung cancer tissues and adjacent normal lung tissues were compared using real-time reverse transcription polymerase chain reaction. Although BALB/c nude mice inoculated with parental PC14 cells had few metastases, almost all mice inoculated with PC14HM cells developed metastases in multiple organs, including the lung, bone and adrenal gland, the same progression seen in human lung cancer. cDNA microarray analysis revealed that 981 genes were differentially (more than 3-fold) expressed between the two cell lines. Functional classification revealed that many of those genes were associated with cell growth, cell communication, development and transcription. Expression of three upregulated genes (HRB-2, HS3ST3A1 and RAB7) was higher in human cancer tissue compared to normal lung tissue, while expression of EDG1, which was downregulated, was lower in the cancer tissue compared to the normal lung. These results suggest that the newly established PC14HM cell line may provide a mouse model of widespread metastasis of lung cancer. This model system may provide insights into the key genetic determinants of widespread metastasis of lung cancer.
Collapse
Affiliation(s)
- Tetsuhiro Nakano
- Department of Thoracic and Visceral Organ Surgery, Gunma University Graduate School of Medicine, Gunma 371-8511, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
A comparison of epithelial-to-mesenchymal transition and re-epithelialization. Semin Cancer Biol 2012; 22:471-83. [PMID: 22863788 DOI: 10.1016/j.semcancer.2012.07.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2012] [Accepted: 07/20/2012] [Indexed: 12/21/2022]
Abstract
Wound healing and cancer metastasis share a common starting point, namely, a change in the phenotype of some cells from stationary to motile. The term, epithelial-to-mesenchymal transition (EMT) describes the changes in molecular biology and cellular physiology that allow a cell to transition from a sedentary cell to a motile cell, a process that is relevant not only for cancer and regeneration, but also for normal development of multicellular organisms. The present review compares the similarities and differences in cellular response at the molecular level as tumor cells enter EMT or as keratinocytes begin the process of re-epithelialization of a wound. Looking toward clinical interventions that might modulate these processes, the mechanisms and outcomes of current and potential therapies are reviewed for both anti-cancer and pro-wound healing treatments related to the pathways that are central to EMT. Taken together, the comparison of re-epithelialization and tumor EMT serves as a starting point for the development of therapies that can selectively modulate different forms of EMT.
Collapse
|
34
|
Härmä V, Knuuttila M, Virtanen J, Mirtti T, Kohonen P, Kovanen P, Happonen A, Kaewphan S, Ahonen I, Kallioniemi O, Grafström R, Lötjönen J, Nees M. Lysophosphatidic acid and sphingosine-1-phosphate promote morphogenesis and block invasion of prostate cancer cells in three-dimensional organotypic models. Oncogene 2012; 31:2075-89. [PMID: 21996742 PMCID: PMC3330266 DOI: 10.1038/onc.2011.396] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2011] [Revised: 06/26/2011] [Accepted: 07/28/2011] [Indexed: 12/12/2022]
Abstract
Normal prostate and some malignant prostate cancer (PrCa) cell lines undergo acinar differentiation and form spheroids in three-dimensional (3-D) organotypic culture. Acini formed by PC-3 and PC-3M, less pronounced also in other PrCa cell lines, spontaneously undergo an invasive switch, leading to the disintegration of epithelial structures and the basal lamina, and formation of invadopodia. This demonstrates the highly dynamic nature of epithelial plasticity, balancing epithelial-to-mesenchymal transition against metastable acinar differentiation. This study assessed the role of lipid metabolites on epithelial maturation. PC-3 cells completely failed to form acinar structures in delipidated serum. Adding back lysophosphatidic acid (LPA) and sphingosine-1-phosphate (S1P) rescued acinar morphogenesis and repressed invasion effectively. Blocking LPA receptor 1 (LPAR1) functions by siRNA (small interference RNA) or the specific LPAR1 inhibitor Ki16425 promoted invasion, while silencing of other G-protein-coupled receptors responsive to LPA or S1P mainly caused growth arrest or had no effects. The G-proteins Gα(12/13) and Gα(i) were identified as key mediators of LPA signalling via stimulation of RhoA and Rho kinases ROCK1 and 2, activating Rac1, while inhibition of adenylate cyclase and accumulation of cAMP may be secondary. Interfering with these pathways specifically impeded epithelial polarization in transformed cells. In contrast, blocking the same pathways in non-transformed, normal cells promoted differentiation. We conclude that LPA and LPAR1 effectively promote epithelial maturation and block invasion of PrCa cells in 3-D culture. The analysis of clinical transcriptome data confirmed reduced expression of LPAR1 in a subset of PrCa's. Our study demonstrates a metastasis-suppressor function for LPAR1 and Gα(12/13) signalling, regulating cell motility and invasion versus epithelial maturation.
Collapse
Affiliation(s)
- V Härmä
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| | - M Knuuttila
- Biotechnology Centre, University of Turku, Turku, Finland
| | - J Virtanen
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Biotechnology Centre, University of Turku, Turku, Finland
| | - T Mirtti
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - P Kohonen
- Biotechnology Centre, University of Turku, Turku, Finland
| | - P Kovanen
- Department of Pathology, Haartman Institute, University of Helsinki and HUSLAB, Helsinki University Central Hospital, Helsinki, Finland
| | - A Happonen
- Department of Signal Processing, Tampere University of Technology, Tampere, Finland
| | - S Kaewphan
- Department of Information Technology, University of Turku, Turku, Finland
| | - I Ahonen
- Biotechnology Centre, University of Turku, Turku, Finland
| | - O Kallioniemi
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - R Grafström
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
- Laboratory for Toxicology, Karolinska Institute, Stockholm, Sweden
| | - J Lötjönen
- Knowledge Intensive Services, VTT Technical Research Centre of Finland, Tampere, Finland
| | - M Nees
- Medical Biotechnology Knowledge Centre, VTT Technical Research Centre of Finland, Turku, Finland
| |
Collapse
|
35
|
Rolin J, Maghazachi AA. Effects of lysophospholipids on tumor microenvironment. CANCER MICROENVIRONMENT 2011; 4:393-403. [PMID: 21904916 DOI: 10.1007/s12307-011-0088-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 08/26/2011] [Indexed: 12/20/2022]
Abstract
The effects of lysophospholipids (LPLs) on cancer microenvironment is a vast and growing field. These lipids are secreted physiologically by various cell types. They play highly important roles in the development, activation and regulation of the immune system. They are also secreted by cancerous cells and there is a strong association between LPLs and cancer. It is clear that these lipids and in particular sphingosine 1-phosphate (S1P) and lysophosphatidic acid (LPA) play major roles in regulating the growth of tumor cells, and in manipulating the immune system. These activities can be divided into two parts; the first involves the ability of S1P and LPA to either directly or through some of the enzymes that generate them such as sphingosine kinases or phospholipases, induce the motility and invasiveness of tumor cells. The second mechanism involves the recently discovered effects of these lipids on the anti-tumor effector natural killer (NK) cells. Whereas S1P and LPA induce the recruitment of these effector cells, they also inhibit their cytolysis of tumor cells. This may support the environment of cancer and the ability of cancer cells to grow, spread and metastasize. Consequently, LPLs or their receptors may be attractive targets for developing drugs in the treatment of cancer where LPLs or their receptors are up-regulated.
Collapse
Affiliation(s)
- Johannes Rolin
- Department of Physiology, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, POB 1103 Blindern, 0317, Oslo, Norway,
| | | |
Collapse
|
36
|
Kim ES, Kim JS, Kim SG, Hwang S, Lee CH, Moon A. Sphingosine 1-phosphate regulates matrix metalloproteinase-9 expression and breast cell invasion through S1P3-Gαq coupling. J Cell Sci 2011; 124:2220-30. [PMID: 21652634 DOI: 10.1242/jcs.076794] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent evidence suggests that inflammation is involved in malignant progression of breast cancer. Sphingosine 1-phosphate (S1P), acting on the G-protein-coupled receptors, is known as a potent inflammatory mediator. In this study, the effect of the inflammatory lipid S1P on the regulation of invasive/migratory phenotypes of MCF10A human breast epithelial cells was investigated to elucidate a causal relationship between inflammation and the control of invasiveness of breast cells. We show that S1P causes induction of matrix metalloproteinase-9 (MMP-9) in vitro and in vivo, and thus enhances invasion and migration. We also show that fos plays a crucial role in the transcriptional activation of MMP-9 by S1P. In addition, activation of extracellular-signal-regulated kinases 1 and 2 (ERK1/2), p38 and alpha serine/threonine-protein kinase (Akt) are involved in the process of S1P-mediated induction of MMP-9 expression and invasion. Activation of the S1P receptor S1P₃ and G(αq) are required for S1P-induced invasive/migratory responses, suggesting that the enhancement of S1P-mediated invasiveness is triggered by the specific coupling of S1P₃ to the heterotrimeric G(αq) subunit. Activation of phospholipase C-β₄ and intracellular Ca²⁺ release are required for S1P-induced MMP-9 upregulation. Taken together, this study demonstrated that S1P regulates MMP-9 induction and invasiveness through coupling of S1P₃ and G(αq) in MCF10A cells, thus providing a molecular basis for the crucial role of S1P in promoting breast cell invasion.
Collapse
Affiliation(s)
- Eun-Sook Kim
- College of Pharmacy, Duksung Women's University, Seoul 132-714, Korea
| | | | | | | | | | | |
Collapse
|
37
|
Takuwa N, Du W, Kaneko E, Okamoto Y, Yoshioka K, Takuwa Y. Tumor-suppressive sphingosine-1-phosphate receptor-2 counteracting tumor-promoting sphingosine-1-phosphate receptor-1 and sphingosine kinase 1 - Jekyll Hidden behind Hyde. Am J Cancer Res 2011; 1:460-481. [PMID: 21984966 PMCID: PMC3186046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Accepted: 02/19/2011] [Indexed: 05/31/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a plasma lipid mediator with multiple roles in mammalian development, physiology and pathophysiology. It is constitutively produced mostly by erythrocytes by the action of sphingosine kinase 1 (SphK1), resulting in high (∼0.5 micromolar) steady-state plasma S1P content and steep S1P concentration gradient imposed between plasma/lymph/tissue interstitial fluid. S1P is also locally produced by activated platelets and tumor cells, in the latter case SphK1 is a downstream target of activated Ras mutant and hypoxia, and is frequently upregulated especially in advanced stages of tumors. Most if not all of the S1P actions in vertebrates are mediated through evolutionarily conserved G protein-coupled S1P receptor family. Ubiquitously expressed mammalian subtypes S1PR1, S1PR2 and S1PR3 mediate pleiotropic actions of S1P in diverse cell types, through coupling to distinctive repertoire of heterotrimeric G proteins. S1PR1 and S1PR3 mediate directed cell migration toward S1P through coupling to G(i) and activating Rac, a Rho family small G protein essential for cell migration. Indeed, S1PR1 expressed in lymphocytes directs their egress from lymph nodes into lymph and recirculation, serving as the target for downregulation by the immunosuppressant FTY720 (fingolimod). S1PR1 in endothelial cells plays an essential role in vascular maturation in embryonic stage, and mediates angiogenic and vascular protective roles of S1P which include eNOS activation and maintenance of barrier integrity. It is likely that S1PR1 and SphK1 expressed in host endothelial cells and tumor cells act in concert in a paracrine loop to contribute to tumor angiogenesis, tumor invasion and progression. In sharp contrast, S1PR2 mediates S1P inhibition of Rac at the site downstream of G(12/13)-mediated Rho activation, thus identified as the first G protein-coupled receptor that negatively regulates Rac and cell migration. S1PR2 could also mediate inhibition of Akt and cell proliferation/survival signaling via Rho-ROCK-PTEN pathway. S1PR2 expressed in tumor cells mediates inhibition of cell migration and invasion in vitro and metastasis in vivo. Moreover, S1PR2 expressed in host endothelial cells and tumor-infiltrating myeloid cells in concert mediates potent inhibition of tumor angiogenesis and tumor growth in vivo, with inhibition of VEGF expression and MMP9 activity. These recent findings provide further basis for S1P receptor subtype-specific, novel therapeutic tactics for individualized treatment of patients with cancer.
Collapse
Affiliation(s)
- Noriko Takuwa
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
- Department of Health and Medical Sciences, Ishikawa Prefectural Nursing University7-1 Nakanuma-tu, Kahoku, Ishikawa 929-1212, Japan
| | - Wa Du
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Erika Kaneko
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Yasuo Okamoto
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Yoh Takuwa
- Department of Physiology, Kanazawa University Graduate School of Medicine13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| |
Collapse
|
38
|
Antimetastatic activity and low systemic toxicity of tetradecyl gallate in a preclinical melanoma mouse model. Invest New Drugs 2011; 30:870-9. [DOI: 10.1007/s10637-010-9628-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 12/27/2010] [Indexed: 01/08/2023]
|
39
|
Becker S, von Otte S, Robenek H, Diedrich K, Nofer JR. Follicular fluid high-density lipoprotein-associated sphingosine 1-phosphate (S1P) promotes human granulosa lutein cell migration via S1P receptor type 3 and small G-protein RAC1. Biol Reprod 2010; 84:604-12. [PMID: 20980685 DOI: 10.1095/biolreprod.110.084152] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Coordinated migration and progesterone production by granulosa cells is critical to the development of the corpus luteum, but the underlying mechanisms remain obscure. Sphingosine 1-phosphate (S1P), which is associated with follicular fluid high-density lipoprotein (FF-HDL), was previously shown to regulate ovarian angiogenesis. We herein examined the effects of S1P and FF-HDL on the function of granulosa lutein cells. Both FF-HDL and S1P induced migration of primary human granulosa lutein cells (hGCs) and the granulosa lutein cell line HGL5. In addition, FF-HDL but not S1P promoted progesterone synthesis, and neither of the two compounds stimulated proliferation of granulosa lutein cells. Polymerase chain reaction and Western blot experiments demonstrated the expression of S1P receptor type 1 (S1PR1), S1PR2, S1PR3, and S1PR5 but not S1PR4 in hGCs and HGL5 cells. The FF-HDL- and S1P-induced granulosa lutein cell migration was emulated by FTY720, an agonist of S1PR1, S1PR3, S1PR4, and S1PR5, and by VPC24191, an agonist of S1PR1 and S1PR3, but not by SEW2871 and phytosphingosine 1-phosphate, agonists of S1PR1 and S1PR4, respectively. In addition, blockade of S1PR3 with CAY1044, suramine, or pertussis toxin inhibited hGC and HGL5 cell migration toward FF-HDL or S1P, while blockade of S1PR1 and S1PR2 with W146 and JTE013, respectively, had no effect. Both FF-HDL and S1P triggered activation of small G-protein RAC1 and actin polymerization in granulosa cells, and RAC1 inhibition with Clostridium difficile toxin B or NSC23766 abolished FF-HDL- and S1P-induced migration. The FF-HDL-associated S1P promotes granulosa lutein cell migration via S1PR3 and RAC1 activation. This may represent a novel mechanism contributing to the development of the corpus luteum.
Collapse
Affiliation(s)
- Steffi Becker
- Department of Obstetrics and Gynecology, University of Schleswig-Holstein, Campus Lübeck, Lübeck, Germany
| | | | | | | | | |
Collapse
|
40
|
Brocklyn JRV. Regulation of cancer cell migration and invasion by sphingosine-1-phosphate. World J Biol Chem 2010; 1:307-12. [PMID: 21537464 PMCID: PMC3083934 DOI: 10.4331/wjbc.v1.i10.307] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 07/28/2010] [Accepted: 08/04/2010] [Indexed: 02/05/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid that has been implicated in regulation of a number of cancer cell malignant behaviors, including cell proliferation, survival, chemotherapeutic resistance and angiogenesis. However, the effects of S1P on cancer cell migration, invasion and metastasis, are perhaps its most complex, due to the fact that, depending upon the S1P receptors that mediate its responses and the crosstalk with other signaling pathways, S1P can either positively or negatively regulate invasion. This review summarizes the effects of S1P on cancer cell invasion and the mechanisms by which it affects this important aspect of cancer cell behavior.
Collapse
Affiliation(s)
- James R Van Brocklyn
- James R Van Brocklyn, Department of Pathology, The Ohio State University, Columbus, OH 43210, United States
| |
Collapse
|
41
|
Takuwa Y, Du W, Qi X, Okamoto Y, Takuwa N, Yoshioka K. Roles of sphingosine-1-phosphate signaling in angiogenesis. World J Biol Chem 2010; 1:298-306. [PMID: 21537463 PMCID: PMC3083935 DOI: 10.4331/wjbc.v1.i10.298] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2010] [Revised: 09/15/2010] [Accepted: 09/22/2010] [Indexed: 02/05/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a blood-borne lipid mediator with pleiotropic biological activities. S1P acts via the specific cell surface G-protein-coupled receptors, S1P1-5. S1P1 and S1P2 were originally identified from vascular endothelial cells (ECs) and smooth muscle cells, respectively. Emerging evidence shows that S1P plays crucial roles in the regulation of vascular functions, including vascular formation, barrier protection and vascular tone via S1P1, S1P2 and S1P3. In particular, S1P regulates vascular formation through multiple mechanisms; S1P exerts both positive and negative effects on angiogenesis and vascular maturation. The positive and negative effects of S1P are mediated by S1P1 and S1P2, respectively. These effects of S1P1 and S1P2 are probably mediated by the S1P receptors expressed in multiple cell types including ECs and bone-marrow-derived cells. The receptor-subtype-specific, distinct effects of S1P favor the development of novel therapeutic tactics for antitumor angiogenesis in cancer and therapeutic angiogenesis in ischemic diseases.
Collapse
Affiliation(s)
- Yoh Takuwa
- Yoh Takuwa, Wa Du, Xun Qi, Yasuo Okamoto, Noriko Takuwa, Kazuaki Yoshioka, Department of Physiology, Kanazawa University Graduate School of Medicine, 13-1 Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | | | | | | | | | | |
Collapse
|
42
|
Ponnusamy S, Meyers-Needham M, Senkal CE, Saddoughi SA, Sentelle D, Selvam SP, Salas A, Ogretmen B. Sphingolipids and cancer: ceramide and sphingosine-1-phosphate in the regulation of cell death and drug resistance. Future Oncol 2010; 6:1603-24. [PMID: 21062159 PMCID: PMC3071292 DOI: 10.2217/fon.10.116] [Citation(s) in RCA: 238] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sphingolipids have emerged as bioeffector molecules, controlling various aspects of cell growth and proliferation in cancer, which is becoming the deadliest disease in the world. These lipid molecules have also been implicated in the mechanism of action of cancer chemotherapeutics. Ceramide, the central molecule of sphingolipid metabolism, generally mediates antiproliferative responses, such as cell growth inhibition, apoptosis induction, senescence modulation, endoplasmic reticulum stress responses and/or autophagy. Interestingly, recent studies suggest de novo-generated ceramides may have distinct and opposing roles in the promotion/suppression of tumors, and that these activities are based on their fatty acid chain lengths, subcellular localization and/or direct downstream targets. For example, in head and neck cancer cells, ceramide synthase 6/C(16)-ceramide addiction was revealed, and this was associated with increased tumor growth, whereas downregulation of its synthesis resulted in ER stress-induced apoptosis. By contrast, ceramide synthase 1-generated C(18)-ceramide has been shown to suppress tumor growth in various cancer models, both in situ and in vivo. In addition, ceramide metabolism to generate sphingosine-1-phosphate (S1P) by sphingosine kinases 1 and 2 mediates, with or without the involvement of G-protein-coupled S1P receptor signaling, prosurvival, angiogenesis, metastasis and/or resistance to drug-induced apoptosis. Importantly, recent findings regarding the mechanisms by which sphingolipid metabolism and signaling regulate tumor growth and progression, such as identifying direct intracellular protein targets of sphingolipids, have been key for the development of new chemotherapeutic strategies. Thus, in this article, we will present conclusions of recent studies that describe opposing roles of de novo-generated ceramides by ceramide synthases and/or S1P in the regulation of cancer pathogenesis, as well as the development of sphingolipid-based cancer therapeutics and drug resistance.
Collapse
Affiliation(s)
- Suriyan Ponnusamy
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Marisa Meyers-Needham
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Can E Senkal
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Sahar A Saddoughi
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - David Sentelle
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Shanmugam Panneer Selvam
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Arelis Salas
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| | - Besim Ogretmen
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
- Hollings Cancer Center, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
43
|
van Sluis GL, Büller HR, Spek CA. The role of activated protein C in cancer progression. Thromb Res 2010; 125 Suppl 2:S138-42. [PMID: 20433994 DOI: 10.1016/s0049-3848(10)70032-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Activated protein C (APC) is best known as a natural anticoagulant that also has direct cell signaling properties which (among others) enhance vascular barrier function. We recently established the relevance of APC-induced barrier enhancement by showing that endogenous APC limits cancer cell extravasation. In line with this concept, repeated administration of exogenous APC reduced the number of experimental metastasis. It is thus tempting to speculate that exogenous APC administration would be a novel therapeutic avenue to fight cancer metastasis. The current review summarizes recent data on the role of the protein C pathway in cancer metastasis. It discusses the APC pathway as a potential novel target to influence cancer progression, but it also points to several limitations of APC administration in the setting of cancer cell metastasis.
Collapse
Affiliation(s)
- Geerte L van Sluis
- Department of Vascular Medicine, Academic Medical Center, Amsterdam, the Netherlands.
| | | | | |
Collapse
|
44
|
König K, Diehl L, Rommerscheidt-Fuss U, Golletz C, Quast T, Kahl P, Kolanus W, Knolle P, Buettner R, Heukamp LC. Four-and-a-Half LIM Domain Protein 2 Is a Novel Regulator of Sphingosine 1-Phosphate Receptor 1 in CCL19-Induced Dendritic Cell Migration. THE JOURNAL OF IMMUNOLOGY 2010; 185:1466-75. [DOI: 10.4049/jimmunol.0903449] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
45
|
Leong WI, Saba JD. S1P metabolism in cancer and other pathological conditions. Biochimie 2010; 92:716-23. [PMID: 20167244 PMCID: PMC2878883 DOI: 10.1016/j.biochi.2010.02.014] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 02/12/2010] [Indexed: 12/28/2022]
Abstract
Nearly two decades ago, the sphingolipid metabolite sphingosine 1-phosphate was discovered to function as a lipid mediator and regulator of cell proliferation. Since that time, sphingosine 1-phosphate has been shown to mediate a diverse array of fundamental biological processes including cell proliferation, migration, invasion, angiogenesis, vascular maturation and lymphocyte trafficking. Sphingosine 1-phosphate acts primarily via signaling through five ubiquitously expressed G protein-coupled receptors. Intracellular sphingosine 1-phosphate molecules are transported extracellularly and gain access to cognate receptors for autocrine and paracrine signaling and for signaling at distant sites reached through blood and lymphatic circulation systems. Intracellular pools of sphingosine 1-phosphate available for signaling are tightly regulated primarily by three enzymes: sphinosine kinase, S1P lyase and S1P phosphatase. Alterations in sphingosine 1-phosphate as well as the enzymes involved in its synthesis and catabolism have been observed in many types of malignancy. These enzymes are being evaluated for their role in mediating cancer formation and progression, as well as their potential to serve as targets of anti-cancer therapeutics. In this review, the impact of sphingosine 1-phosphate, its cognate receptors, and the enzymes of sphingosine 1-phosphate metabolism on cell survival, apoptosis, autophagy, cellular transformation, invasion, angiogenesis and hypoxia in relation to cancer biology and treatment are discussed.
Collapse
Affiliation(s)
- Weng In Leong
- Children’s Hospital Oakland Research Institute, 5700 Martin Luther King, Jr. Way, Oakland, CA 94609, USA, Tel: +1 510 450 7690, Fax: +1 510 450 7910,
| | - Julie D. Saba
- Children’s Hospital Oakland Research Institute, 5700 Martin Luther King, Jr. Way, Oakland, CA 94609, USA, Tel: +1 510 450 7690, Fax: +1 510 450 7910,
| |
Collapse
|
46
|
Müller R, Berliner C, Leptin J, Pörtner D, Bialecki W, Kleuser B, Schumacher U, Milićević NM. Expression of sphingosine-1-phosphate receptors and lysophosphatidic acid receptors on cultured and xenografted human colon, breast, melanoma, and lung tumor cells. Tumour Biol 2010; 31:341-9. [PMID: 20480410 DOI: 10.1007/s13277-010-0043-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 04/21/2010] [Indexed: 12/25/2022] Open
Abstract
The lysophospholipids sphingosine-1-phosphate (S1P) and lysophosphatidic acid (LPA) are small lipid molecules with a variety of physiological roles. Additionally, their involvement in the initiation and progression of malignant tumors has been increasingly recognized in recent years. However, the data on the expression of S1P and LPA receptors on different cancer cells are very few. Real-time polymerase chain reaction was used for the analysis of mRNA expression of five S1P((1-5)) and three LPA((1-3)) receptors on a large panel of 13 colon, breast, melanoma, and lung cancer cell lines. Furthermore, the modulation of S1P and LPA receptor mRNA expression was studied upon xenotransplantation of tumor cells into severe combined immunodeficient (SCID) mice. The S1P and LPA receptors were expressed to a variable degree on all tumor cell lines tested (with exception of colon cancer SW480). Most notably, tumor cell lines in vitro expressed S1P(2) mRNA that was down-regulated upon xenotransplantation, whereas LPA(2) receptor mRNA was strongly expressed both in vitro and in vivo (except by breast cancer cells). The latter was especially distinctive for small cell lung tumor cells. The S1P and LPA receptors are differentially expressed on tumor cell lines in vitro. Their expression is modulated upon xenografting into SCID mice in vivo.
Collapse
Affiliation(s)
- Reinhard Müller
- Center for Experimental Medicine, Institute of Anatomy II: Experimental Morphology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Yoshida Y, Nakada M, Sugimoto N, Harada T, Hayashi Y, Kita D, Uchiyama N, Hayashi Y, Yachie A, Takuwa Y, Hamada JI. Sphingosine-1-phosphate receptor type 1 regulates glioma cell proliferation and correlates with patient survival. Int J Cancer 2010; 126:2341-52. [PMID: 19810093 DOI: 10.1002/ijc.24933] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lipid that signals through a family of G protein-coupled receptors consisting of 5 members termed S1P(1-5), and it regulates cellular proliferation, migration and survival. We investigated the expression and role of S1P receptors in glioma. Human glioma expressed S1P(1), S1P(2), S1P(3), and S1P(5) by quantitative real-time PCR analysis. Expression of the S1P(1) was significantly lower in glioblastoma than in the normal brain (p < 0.01) and diffuse astrocytoma (p < 0.05). Immunoblotting showed that normal brain expressed more S1P(1) protein than did glioblastoma. Immunohistochemistry showed that S1P(1) was localized predominantly in the astrocytes in the normal brain, but no staining was observed in glioblastoma. Downregulation of S1P(1) expression correlated with poor survival of patients with glioblastoma (p < 0.05). S1P(1) small interfering RNA promoted cell proliferation in high-expressor glioma cell lines (T98G, G112). Cell proliferation was promoted by the pertussis toxin, which deactivates G(i/o) type of G proteins; the S1P(1) is exclusively coupled to these proteins. Forced expression of the S1P(1) in low-expressor cell lines (U87, U251) resulted in decreased cell growth and led to suppressed tumor growth in transplanted gliomas in vivo. Furthermore, we found a significant association between the S1P(1) expression and early growth response-1, a transcriptional factor that exhibits tumor suppression in glioblastoma cells (p < 0.05). These data indicate that the downregulation of S1P(1) expression enhances the malignancy of glioblastoma by increasing cell proliferation and correlates with the shorter survival of patients with glioblastoma.
Collapse
Affiliation(s)
- Yuya Yoshida
- Department of Neurosurgery, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Du W, Takuwa N, Yoshioka K, Okamoto Y, Gonda K, Sugihara K, Fukamizu A, Asano M, Takuwa Y. S1P(2), the G protein-coupled receptor for sphingosine-1-phosphate, negatively regulates tumor angiogenesis and tumor growth in vivo in mice. Cancer Res 2010; 70:772-81. [PMID: 20068174 DOI: 10.1158/0008-5472.can-09-2722] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sphingosine-1-phosphate (S1P) has been implicated in tumor angiogenesis by acting through the G(i)-coupled chemotactic receptor S1P(1). Here, we report that the distinct receptor S1P(2) is responsible for mediating the G(12/13)/Rho-dependent inhibitory effects of S1P on Akt, Rac, and cell migration, thereby negatively regulating tumor angiogenesis and tumor growth. By using S1P(2)(LacZ/+) mice, we found that S1P(2) was expressed in both tumor and normal blood vessels in many organs, in both endothelial cells (EC) and vascular smooth muscle cells, as well as in tumor-associated, CD11b-positive bone marrow-derived cells (BMDC). Lewis lung carcinoma or B16 melanoma cells implanted in S1P(2)-deficient (S1P(2)(-/-)) mice displayed accelerated tumor growth and angiogenesis with enhanced association of vascular smooth muscle cells and pericytes. S1P(2)(-/-) ECs exhibited enhanced Rac activity, Akt phosphorylation, cell migration, proliferation, and tube formation in vitro. Coinjection of S1P(2)(-/-) ECs and tumor cells into wild-type mice also produced a relative enhancement of tumor growth and angiogenesis in vivo. S1P(2)(-/-) mice were also more efficient at recruiting CD11b-positive BMDCs into tumors compared with wild-type siblings. Bone marrow chimera experiments revealed that S1P(2) acted in BMDCs to promote tumor growth and angiogenesis. Our results indicate that, in contrast to endothelial S1P(1), which stimulates tumor angiogenesis, S1P(2) on ECs and BMDCs mediates a potent inhibition of tumor angiogenesis, suggesting a novel therapeutic tactic for anticancer treatment.
Collapse
Affiliation(s)
- Wa Du
- Department of Physiology, Kanazawa University Graduate School of Medicine, Kanazawa 920-8640, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Suppression of hepatocellular carcinoma recurrence after rat liver transplantation by FTY720, a sphingosine-1-phosphate analog. Transplantation 2010; 88:980-6. [PMID: 19855243 DOI: 10.1097/tp.0b013e3181b9ca69] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
BACKGROUND.: Although the outcome of liver transplant patients with hepatocellular carcinoma (HCC) has improved with the introduction of strict criteria, tumor recurrence still remains a significant problem. Sphingosine-1-phosphate (S1P) is a phospholipid mediator that can induce diverse cellular responses, such as proliferation, migration, adhesion, and cell-rounding, in cancer cells. We investigated whether FTY720, a S1P analog, suppresses tumor recurrence after experimental liver transplantation in a rat HCC model. METHODS.: HCC-bearing rats were subjected to orthotropic liver transplantation. HCC cells were analyzed for cell migration, proliferation, and S1P receptors. RESULTS.: FTY720 induced the down-regulation of the S1P-1 receptor of HCC cells and suppressed both cancer cell migration and proliferation. FTY720 also suppressed mitogen-activated protein kinase phosphorylation. The suppression of tumor recurrence after liver transplantation and a significant prolongation of survival were observed in the FTY720-treated rats, in comparison with FTY720-untreated rats. CONCLUSION.: FTY720 suppresses the invasiveness and proliferation of HCC through a down-regulating S1P-1 receptor to suppress the recurrence of HCC after liver transplantation; FTY720 may be used as a new antimetastatic agent for the prevention of tumor recurrence after liver transplantation.
Collapse
|
50
|
Cattoretti G, Mandelbaum J, Lee N, Chaves AH, Mahler AM, Chadburn A, Dalla-Favera R, Pasqualucci L, MacLennan AJ. Targeted disruption of the S1P2 sphingosine 1-phosphate receptor gene leads to diffuse large B-cell lymphoma formation. Cancer Res 2009; 69:8686-92. [PMID: 19903857 DOI: 10.1158/0008-5472.can-09-1110] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
S1P(2) sphingosine 1-phosphate receptor signaling can regulate proliferation, survival, morphology, and migration in many cell types in vitro. Here, we report that S1P(2)(-/-) mice develop clonal B-cell lymphomas with age, such that approximately half of the animals display this neoplasm by 1.5 to 2 years of age. Histologic, immunophenotypic, and molecular analyses revealed a uniform tumor phenotype with features of germinal center (GC)-derived diffuse large B-cell lymphoma (DLBCL). Tumor formation was preceded by increases in GC B cells and CD69(+) T cells, as well as an increased formation of spontaneous GCs, suggesting that S1P(2) loss may promote lymphomagenesis in part by disrupting GC B-cells homeostasis. With the sole exception of rare lung tumors, the effect of S1P(2) gene disruption is remarkably restricted to DLBCL. In humans, 28 of 106 (26%) DLBCL samples were found to harbor multiple somatic mutations in the 5' sequences of the S1P(2) gene. Mutations displayed features resembling those generated by the IgV-associated somatic hypermutation mechanism, but were not detected at significant levels in normal GC B cells, indicating a tumor-associated aberrant function. Collectively, our data suggest that S1P(2) signaling may play a critical role in suppressing DLBCL formation in vivo. The high incidence of DLBCL in S1P(2)(-/-) mice, its onset at old age, and the relative lack of other neoplasms identify these mice as a novel, and potentially valuable, model for this highly prevalent and aggressive human malignancy.
Collapse
Affiliation(s)
- Giorgio Cattoretti
- Institute for Cancer Genetics and the Department of Pathology, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, New York, USA
| | | | | | | | | | | | | | | | | |
Collapse
|