1
|
Navone SE, Guarnaccia L, Rizzaro MD, Begani L, Barilla E, Alotta G, Garzia E, Caroli M, Ampollini A, Violetti A, Gervasi N, Campanella R, Riboni L, Locatelli M, Marfia G. Role of Luteolin as Potential New Therapeutic Option for Patients with Glioblastoma through Regulation of Sphingolipid Rheostat. Int J Mol Sci 2023; 25:130. [PMID: 38203299 PMCID: PMC10779390 DOI: 10.3390/ijms25010130] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 01/12/2024] Open
Abstract
Glioblastoma (GBM) is the most aggressive brain tumor, still considered incurable. In this study, conducted on primary GBM stem cells (GSCs), specifically selected as the most therapy-resistant, we examined the efficacy of luteolin, a natural flavonoid, as an anti-tumoral compound. Luteolin is known to impact the sphingolipid rheostat, a pathway regulated by the proliferative sphingosine-1-phosphate (S1P) and the proapoptotic ceramide (Cer), and implicated in numerous oncopromoter biological processes. Here, we report that luteolin is able to inhibit the expression of SphK1/2, the two kinases implicated in S1P formation, and to increase the expression of both SGPL1, the lyase responsible for S1P degradation, and CERS1, the ceramide synthase 1, thus shifting the balance toward the production of ceramide. In addition, luteolin proved to decrease the expression of protumoral signaling as MAPK, RAS/MEK/ERK and PI3K/AKT/mTOR and cyclins involved in cell cycle progression. In parallel, luteolin succeeded in upregulation of proapoptotic mediators as caspases and Bcl-2 family and cell cycle controllers as p53 and p27. Furthermore, luteolin determined the shutdown of autophagy contributing to cell survival. Overall, our data support the use of luteolin as add-on therapy, having demonstrated a good ability in impairing GSC viability and survival and increasing cell sensitivity to TMZ.
Collapse
Affiliation(s)
- Stefania Elena Navone
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Guarnaccia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Massimiliano D. Rizzaro
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Laura Begani
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Emanuela Barilla
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Giovanni Alotta
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Emanuele Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital Medical School, ASST Santi Paolo e Carlo, 20142 Milan, Italy;
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| | - Manuela Caroli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Antonella Ampollini
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
| | - Aniello Violetti
- Space Attache’, Embassy of Italy in Washington DC, Washington, DC 20008, USA
| | - Noreen Gervasi
- Alcamena Stem Cell Therapeutics, 1450 South Rolling Road, Suite 4.069, Halethorpe, MD 21227, USA
| | - Rolando Campanella
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Laura Riboni
- Andremacon Biotech Srl, Viale Ortles, 22/4, 20141 Milan, Italy; (E.B.); (G.A.); (R.C.); (L.R.)
| | - Marco Locatelli
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Department of Medical-Surgical Physiopathology and Transplantation, University of Milan, 20122 Milan, Italy
| | - Giovanni Marfia
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Foundation IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (S.E.N.); (L.G.); (M.D.R.); (L.B.); (M.C.); (A.A.); (M.L.)
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, 20138 Milan, Italy
| |
Collapse
|
2
|
Mebarek S, Skafi N, Brizuela L. Targeting Sphingosine 1-Phosphate Metabolism as a Therapeutic Avenue for Prostate Cancer. Cancers (Basel) 2023; 15:2732. [PMID: 37345069 DOI: 10.3390/cancers15102732] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Prostate cancer (PC) is the second most common cancer in men worldwide. More than 65% of men diagnosed with PC are above 65. Patients with localized PC show high long-term survival, however with the disease progression into a metastatic form, it becomes incurable, even after strong radio- and/or chemotherapy. Sphingosine 1-phosphate (S1P) is a bioactive lipid that participates in all the steps of oncogenesis including tumor cell proliferation, survival, migration, invasion, and metastatic spread. The S1P-producing enzymes sphingosine kinases 1 and 2 (SK1 and SK2), and the S1P degrading enzyme S1P lyase (SPL), have been shown to be highly implicated in the onset, development, and therapy resistance of PC during the last 20 years. In this review, the most important studies demonstrating the role of S1P and S1P metabolic partners in PC are discussed. The different in vitro, ex vivo, and in vivo models of PC that were used to demonstrate the implication of S1P metabolism are especially highlighted. Furthermore, the most efficient molecules targeting S1P metabolism that are under preclinical and clinical development for curing PC are summarized. Finally, the possibility of targeting S1P metabolism alone or combined with other therapies in the foreseeable future as an alternative option for PC patients is discussed. Research Strategy: PubMed from INSB was used for article research. First, key words "prostate & sphingosine" were used and 144 articles were found. We also realized other combinations of key words as "prostate cancer bone metastasis" and "prostate cancer treatment". We used the most recent reviews to illustrate prostate cancer topic and sphingolipid metabolism overview topic.
Collapse
Affiliation(s)
- Saida Mebarek
- CNRS UMR 5246, INSA Lyon, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), 69622 Lyon, France
| | - Najwa Skafi
- CNRS, LAGEPP UMR 5007, University of Lyon, Université Claude Bernard Lyon 1, 43 Bd 11 Novembre 1918, 69622 Villeurbanne, France
| | - Leyre Brizuela
- CNRS UMR 5246, INSA Lyon, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), 69622 Lyon, France
| |
Collapse
|
3
|
Scheinberg T, Mak B, Butler L, Selth L, Horvath LG. Targeting lipid metabolism in metastatic prostate cancer. Ther Adv Med Oncol 2023; 15:17588359231152839. [PMID: 36743527 PMCID: PMC9893394 DOI: 10.1177/17588359231152839] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
Despite key advances in the treatment of prostate cancer (PCa), a proportion of men have de novo resistance, and all will develop resistance to current therapeutics over time. Aberrant lipid metabolism has long been associated with prostate carcinogenesis and progression, but more recently there has been an explosion of preclinical and clinical data which is informing new clinical trials. This review explores the epidemiological links between obesity and metabolic syndrome and PCa, the evidence for altered circulating lipids in PCa and their potential role as biomarkers, as well as novel therapeutic strategies for targeting lipids in men with PCa, including therapies widely used in cardiovascular disease such as statins, metformin and lifestyle modification, as well as novel targeted agents such as sphingosine kinase inhibitors, DES1 inhibitors and agents targeting FASN and beta oxidation.
Collapse
Affiliation(s)
- Tahlia Scheinberg
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Blossom Mak
- Medical Oncology, Chris O’Brien Lifehouse, Camperdown NSW, Australia,Advanced Prostate Cancer Group, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia,University of Sydney, Camperdown, NSW, Australia
| | - Lisa Butler
- Prostate Cancer Research Group, South Australian Health and Medical Research Institute, Adelaide, South Australia, Australia,South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia
| | - Luke Selth
- South Australian Immunogenomics Cancer Institute and Freemason’s Centre for Male Health and Wellbeing, University of Adelaide, South Australia, Australia,Dame Roma Mitchell Cancer Research Labs, Adelaide Medical School, University of Adelaide, Adelaide, South Australia, Australia,Flinders Health and Medical Research Institute, Flinders University, College of Medicine and Public Health, Bedford Park, Australia
| | | |
Collapse
|
4
|
Yang C, Zhou S, Zhu J, Sheng H, Mao W, Fu Z, Chen Z. Plasma Lipid-based Machine Learning Models Provides a Potential Diagnostic Tool for Colorectal Cancer Patients. Clin Chim Acta 2022; 536:191-199. [PMID: 36191612 DOI: 10.1016/j.cca.2022.09.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/23/2022] [Accepted: 09/01/2022] [Indexed: 11/29/2022]
Abstract
Colorectal cancer is the second leading cause of cancer-related death across the world. So far, screening methods for colorectal cancer are limited to blood test, imaging test, and digital rectal examination, that are either invasive or ineffective. So, this study aims to explore novel, more convenient and effective diagnostic methods for colorectal cancer. First, the experiment cohort was randomly split to train set and test set, and LC-MS-based plasma lipidomics was applied to identify lipid features in colorectal cancer. Second, univariate and multivariate analyses were performed to screen for significantly differentially expressed lipids. Third, single-lipid-based ROC analysis and multiple-lipid-based machine learning modelling were conducted to assess differential lipids' diagnostic performance. Lastly, survival analyses were used to evaluate lipids' prognostic values. In total, 41 differential lipids were screened out, 10 were upregulated and 31 were downregulated in CRC. Only CerP(d15:0_22:0+O) showed fine predictive accuracy in single-lipid-base ROC analysis. Among the four machine learning models, SVM showed best predictive performance with accuracy (in predicting test set) of 1.0000 (95%CI: 0.8806, 1.0000), that can be reached by modelling with only 14 lipids. Four lipids had significant prognostic values, that were TG(11:0_18:0_18:0) (HR: 0.34), TG(18:0_18:0_18:1) (HR: 0.34), PC(22:1_12:3) (HR: 2.22), LPC(17:0) (HR: 3.16). In conclusion, this study discovered novel lipid features that has potential diagnostic and prognostic values, and showed combination of plasma lipidomics and machine learning modelling could have outstanding diagnostic performance and may serve as a convenient and more accessible way to aid clinical diagnosis of colorectal cancer.
Collapse
Affiliation(s)
- Chenxi Yang
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Sicheng Zhou
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Jing Zhu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Huaying Sheng
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Weimin Mao
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, Zhejiang, China
| | - Zhixuan Fu
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
| | - Zhongjian Chen
- The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China; Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou, Zhejiang, China; Zhejiang Key Laboratory of Diagnosis & Treatment Technology on Thoracic Oncology (Lung and Esophagus), Hangzhou, Zhejiang, China.
| |
Collapse
|
5
|
Li RZ, Wang XR, Wang J, Xie C, Wang XX, Pan HD, Meng WY, Liang TL, Li JX, Yan PY, Wu QB, Liu L, Yao XJ, Leung ELH. The key role of sphingolipid metabolism in cancer: New therapeutic targets, diagnostic and prognostic values, and anti-tumor immunotherapy resistance. Front Oncol 2022; 12:941643. [PMID: 35965565 PMCID: PMC9364366 DOI: 10.3389/fonc.2022.941643] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/24/2022] [Indexed: 12/13/2022] Open
Abstract
Biologically active sphingolipids are closely related to the growth, differentiation, aging, and apoptosis of cancer cells. Some sphingolipids, such as ceramides, are favorable metabolites in the sphingolipid metabolic pathway, usually mediating antiproliferative responses, through inhibiting cancer cell growth and migration, as well as inducing autophagy and apoptosis. However, other sphingolipids, such as S1P, play the opposite role, which induces cancer cell transformation, migration and growth and promotes drug resistance. There are also other sphingolipids, as well as enzymes, played potentially critical roles in cancer physiology and therapeutics. This review aimed to explore the important roles of sphingolipid metabolism in cancer. In this article, we summarized the role and value of sphingolipid metabolism in cancer, including the distribution of sphingolipids, the functions, and their relevance to cancer diagnosis and prognosis. We also summarized the known and potential antitumor targets present in sphingolipid metabolism, analyzed the correlation between sphingolipid metabolism and tumor immunity, and summarize the antitumor effects of natural compounds based on sphingolipids. Through the analysis and summary of sphingolipid antitumor therapeutic targets and immune correlation, we aim to provide ideas for the development of new antitumor drugs, exploration of new therapeutic means for tumors, and study of immunotherapy resistance mechanisms.
Collapse
Affiliation(s)
- Run-Ze Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
| | - Xuan-Run Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jian Wang
- Department of Oncology, Luzhou People’s Hospital, Luzhou, Sichuan, China
| | - Chun Xie
- Cancer Center, Faculty of Health Science, University of Macau, Macao, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao, Macao SAR, China
| | - Xing-Xia Wang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Hu-Dan Pan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
| | - Wei-Yu Meng
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Tu-Liang Liang
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Jia-Xin Li
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Pei-Yu Yan
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Qi-Biao Wu
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Liang Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine (Guangdong Provincial Hospital of Chinese Medicine), Guangzhou, China
- Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Macao, Macao SAR, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| | - Xiao-Jun Yao
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery/State Key Laboratory of Quality Research in Chinese Medicine/Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macao, Macao SAR, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macao, Macao SAR, China
- MOE Frontiers Science Center for Precision Oncology, University of Macau, Macao, Macao SAR, China
- Breast Surgery, Zhuhai Hospital of Traditional Chinese and Western Medicine, Zhuhai, China
- *Correspondence: Xiao-Jun Yao, ; Liang Liu, ; Elaine Lai-Han Leung,
| |
Collapse
|
6
|
Pherez-Farah A, López-Sánchez RDC, Villela-Martínez LM, Ortiz-López R, Beltrán BE, Hernández-Hernández JA. Sphingolipids and Lymphomas: A Double-Edged Sword. Cancers (Basel) 2022; 14:2051. [PMID: 35565181 PMCID: PMC9104519 DOI: 10.3390/cancers14092051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 04/14/2022] [Indexed: 11/24/2022] Open
Abstract
Lymphomas are a highly heterogeneous group of hematological neoplasms. Given their ethiopathogenic complexity, their classification and management can become difficult tasks; therefore, new approaches are continuously being sought. Metabolic reprogramming at the lipid level is a hot topic in cancer research, and sphingolipidomics has gained particular focus in this area due to the bioactive nature of molecules such as sphingoid bases, sphingosine-1-phosphate, ceramides, sphingomyelin, cerebrosides, globosides, and gangliosides. Sphingolipid metabolism has become especially exciting because they are involved in virtually every cellular process through an extremely intricate metabolic web; in fact, no two sphingolipids share the same fate. Unsurprisingly, a disruption at this level is a recurrent mechanism in lymphomagenesis, dissemination, and chemoresistance, which means potential biomarkers and therapeutical targets might be hiding within these pathways. Many comprehensive reviews describing their role in cancer exist, but because most research has been conducted in solid malignancies, evidence in lymphomagenesis is somewhat limited. In this review, we summarize key aspects of sphingolipid biochemistry and discuss their known impact in cancer biology, with a particular focus on lymphomas and possible therapeutical strategies against them.
Collapse
Affiliation(s)
- Alfredo Pherez-Farah
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico
| | | | - Luis Mario Villela-Martínez
- Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán Rosales 80030, Sinaloa, Mexico
- Hospital Fernando Ocaranza, ISSSTE, Hermosillo 83190, Sonora, Mexico
- Centro Médico Dr. Ignacio Chávez, ISSSTESON, Hermosillo 83000, Sonora, Mexico
| | - Rocío Ortiz-López
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Monterrey 64710, Nuevo Leon, Mexico
| | - Brady E Beltrán
- Hospital Edgardo Rebagliati Martins, Lima 15072, Peru
- Instituto de Investigaciones en Ciencias Biomédicas, Universidad Ricardo Palma, Lima 1801, Peru
| | | |
Collapse
|
7
|
GNE-493 inhibits prostate cancer cell growth via Akt-mTOR-dependent and -independent mechanisms. Cell Death Dis 2022; 8:120. [PMID: 35296639 PMCID: PMC8927604 DOI: 10.1038/s41420-022-00911-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
GNE-493 is a novel PI3K/mTOR dual inhibitor with improved metabolic stability, oral bioavailability, and excellent pharmacokinetic parameters. Here GNE-493 potently inhibited viability, proliferation, and migration in different primary and established (LNCaP and PC-3 lines) prostate cancer cells, and provoking apoptosis. GNE-493 blocked Akt-mTOR activation in primary human prostate cancer cells. A constitutively-active mutant Akt1 restored Akt-mTOR activation but only partially ameliorated GNE-493-induced prostate cancer cell death. Moreover, GNE-493 was still cytotoxic in Akt1/2-silenced primary prostate cancer cells. Significant oxidative stress and programmed necrosis cascade activation were detected in GNE-493-treated prostate cancer cells. Moreover, GNE-493 downregulated Sphingosine Kinase 1 (SphK1), causing ceramide accumulation in primary prostate cancer cells. Daily single dose GNE-493 oral administration robustly inhibited the growth of the prostate cancer xenograft in the nude mice. Akt-mTOR inactivation, SphK1 downregulation, ceramide level increase, and oxidative injury were detected in GNE-493-treated prostate cancer xenograft tissues. Together, GNE-493 inhibited prostate cancer cell growth possibly through the Akt-mTOR-dependent and -independent mechanisms.
Collapse
|
8
|
George DJ, Halabi S, Heath EI, Sartor AO, Sonpavde GP, Das D, Bitting RL, Berry W, Healy P, Anand M, Winters C, Riggan C, Kephart J, Wilder R, Shobe K, Rasmussen J, Milowsky MI, Fleming MT, Bearden J, Goodman M, Zhang T, Harrison MR, McNamara M, Zhang D, LaCroix BL, Kittles RA, Patierno BM, Sibley AB, Patierno SR, Owzar K, Hyslop T, Freedman JA, Armstrong AJ. A prospective trial of abiraterone acetate plus prednisone in Black and White men with metastatic castrate-resistant prostate cancer. Cancer 2021; 127:2954-2965. [PMID: 33951180 PMCID: PMC9527760 DOI: 10.1002/cncr.33589] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 01/12/2023]
Abstract
BACKGROUND Retrospective analyses of randomized trials suggest that Black men with metastatic castration-resistant prostate cancer (mCRPC) have longer survival than White men. The authors conducted a prospective study of abiraterone acetate plus prednisone to explore outcomes by race. METHODS This race-stratified, multicenter study estimated radiographic progression-free survival (rPFS) in Black and White men with mCRPC. Secondary end points included prostate-specific antigen (PSA) kinetics, overall survival (OS), and safety. Exploratory analysis included genome-wide genotyping to identify single nucleotide polymorphisms associated with progression in a model incorporating genetic ancestry. One hundred patients self-identified as White (n = 50) or Black (n = 50) were enrolled. Eligibility criteria were modified to facilitate the enrollment of individual Black patients. RESULTS The median rPFS for Black and White patients was 16.6 and 16.8 months, respectively; their times to PSA progression (TTP) were 16.6 and 11.5 months, respectively; and their OS was 35.9 and 35.7 months, respectively. Estimated rates of PSA decline by ≥50% in Black and White patients were 74% and 66%, respectively; and PSA declines to <0.2 ng/mL were 26% and 10%, respectively. Rates of grade 3 and 4 hypertension, hypokalemia, and hyperglycemia were higher in Black men. CONCLUSIONS Multicenter prospective studies by race are feasible in men with mCRPC but require less restrictive eligibility. Despite higher comorbidity rates, Black patients demonstrated rPFS and OS similar to those of White patients and trended toward greater TTP and PSA declines, consistent with retrospective reports. Importantly, Black men may have higher side-effect rates than White men. This exploratory genome-wide analysis of TTP identified a possible candidate marker of ancestry-dependent treatment outcomes.
Collapse
Affiliation(s)
- Daniel J. George
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Susan Halabi
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | | | - A. Oliver Sartor
- Tulane Cancer Center, Tulane Health Sciences Center, New Orleans, Louisiana
| | - Guru P. Sonpavde
- Hematology and Oncology Division, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Devika Das
- Hematology and Oncology Division, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama
| | - Rhonda L. Bitting
- Comprehensive Cancer Center, Wake Forest University, Winston Salem, North Carolina
| | - William Berry
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Patrick Healy
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Monika Anand
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Carol Winters
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Colleen Riggan
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Julie Kephart
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Rhonda Wilder
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Kellie Shobe
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Julia Rasmussen
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Matthew I. Milowsky
- Department of Medicine, Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | - Michael Goodman
- W.G. (Bill) Hefner VA Medical Center, Salisbury, North Carolina
| | - Tian Zhang
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Michael R. Harrison
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Megan McNamara
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Dadong Zhang
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Bonnie L. LaCroix
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Rick A. Kittles
- Department of Population Sciences, Division of Health Equities, City of Hope National Medical Center, Duarte, California
| | - Brendon M. Patierno
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Alexander B. Sibley
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Steven R. Patierno
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
- Duke Cancer Institute, Duke University School of Medicine, Durham, North Carolina
| | - Terry Hyslop
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, North Carolina
| | - Jennifer A. Freedman
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| | - Andrew J. Armstrong
- Department of Medicine, Division of Medical Oncology, Duke University, Durham, North Carolina
- Center for Prostate and Urologic Cancers, Duke Cancer Institute, Duke University, Durham, North Carolina
| |
Collapse
|
9
|
Cortini M, Armirotti A, Columbaro M, Longo DL, Di Pompo G, Cannas E, Maresca A, Errani C, Longhi A, Righi A, Carelli V, Baldini N, Avnet S. Exploring Metabolic Adaptations to the Acidic Microenvironment of Osteosarcoma Cells Unveils Sphingosine 1-Phosphate as a Valuable Therapeutic Target. Cancers (Basel) 2021; 13:cancers13020311. [PMID: 33467731 PMCID: PMC7830496 DOI: 10.3390/cancers13020311] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary By studying the role of tumor acidosis in osteosarcoma, we have identified a novel lipid signaling pathway that is selectively activated in acid-induced highly metastatic cell subpopulation. Furthermore, when combined to low-serine/glycine diet, the targeting of this acid-induced lipid pathway by the FDA-approved drug FTY720 significantly impaired tumor growth. This new knowledge will provide a giant leap in the understanding of the molecular mechanisms responsible for sarcoma relapses and metastasis. Finally, we paved the way to the recognition of a novel biomarker, as our data provided evidence of significantly high circulating levels in the serum of osteosarcoma patients of S1P, a lipid member of the identified acid-driven metabolic pathway. Abstract Acidity is a key player in cancer progression, modelling a microenvironment that prevents immune surveillance and enhances invasiveness, survival, and drug resistance. Here, we demonstrated in spheroids from osteosarcoma cell lines that the exposure to acidosis remarkably caused intracellular lipid droplets accumulation. Lipid accumulation was also detected in sarcoma tissues in close proximity to tumor area that express the acid-related biomarker LAMP2. Acid-induced lipid droplets-accumulation was not functional to a higher energetic request, but rather to cell survival. As a mechanism, we found increased levels of sphingomyelin and secretion of the sphingosine 1-phosphate, and the activation of the associated sphingolipid pathway and the non-canonical NF-ĸB pathway, respectively. Moreover, decreasing sphingosine 1-phosphate levels (S1P) by FTY720 (Fingolimod) impaired acid-induced tumor survival and migration. As a confirmation of the role of S1P in osteosarcoma, we found S1P high circulating levels (30.8 ± 2.5 nmol/mL, n = 17) in the serum of patients. Finally, when we treated osteosarcoma xenografts with FTY720 combined with low-serine/glycine diet, both lipid accumulation (as measured by magnetic resonance imaging) and tumor growth were greatly inhibited. For the first time, this study profiles the lipidomic rearrangement of sarcomas under acidic conditions, suggesting the use of anti-S1P strategies in combination with standard chemotherapy.
Collapse
Affiliation(s)
- Margherita Cortini
- Biomedical Science and Technology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.); (M.C.); (G.D.P.); (S.A.)
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy; (A.A.); (E.C.)
| | - Marta Columbaro
- Biomedical Science and Technology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.); (M.C.); (G.D.P.); (S.A.)
| | - Dario Livio Longo
- Institute of Biostructures and Bioimaging, National Research Council of Italy, 10135 Torino, Italy;
| | - Gemma Di Pompo
- Biomedical Science and Technology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.); (M.C.); (G.D.P.); (S.A.)
| | - Elena Cannas
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, 16163 Genova, Italy; (A.A.); (E.C.)
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-Università di Bologna, 40125 Bologna, Italy;
| | - Alessandra Maresca
- Programma di Neurogenetica, IRCCS Istituto Delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy;
| | - Costantino Errani
- Oncologic Orthopaedic Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Alessandra Longhi
- Chemotherapy Unit for Musculoskeletal Tumors, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Alberto Righi
- Anatomy and Pathological Histology Unit, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy;
| | - Valerio Carelli
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-Università di Bologna, 40125 Bologna, Italy;
- Programma di Neurogenetica, IRCCS Istituto Delle Scienze Neurologiche di Bologna, 40139 Bologna, Italy;
| | - Nicola Baldini
- Biomedical Science and Technology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.); (M.C.); (G.D.P.); (S.A.)
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-Università di Bologna, 40125 Bologna, Italy;
- Correspondence:
| | - Sofia Avnet
- Biomedical Science and Technology Lab, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy; (M.C.); (M.C.); (G.D.P.); (S.A.)
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum-Università di Bologna, 40125 Bologna, Italy;
| |
Collapse
|
10
|
Kroll A, Cho HE, Kang MH. Antineoplastic Agents Targeting Sphingolipid Pathways. Front Oncol 2020; 10:833. [PMID: 32528896 PMCID: PMC7256948 DOI: 10.3389/fonc.2020.00833] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/28/2020] [Indexed: 12/16/2022] Open
Abstract
Emerging studies in the enigmatic area of bioactive lipids have made many exciting new discoveries in recent years. Once thought to play a strictly structural role in cellular function, it has since been determined that sphingolipids and their metabolites perform a vast variety of cellular functions beyond what was previously believed. Of utmost importance is their role in cellular signaling, for it is now well understood that select sphingolipids serve as bioactive molecules that play critical roles in both cancer cell death and survival, as well as other cellular responses such as chronic inflammation, protection from intestinal pathogens, and intrinsic protection from intestinal contents, each of which are associated with oncogenesis. Importantly, it has been demonstrated time and time again that many different tumors display dysregulation of sphingolipid metabolism, and the exact profile of said dysregulation has been proven to be useful in determining not only the presence of a tumor, but also the susceptibility to various chemotherapeutic drugs, as well as the metastasizing characteristics of the malignancies. Since these discoveries surfaced it has become apparent that the understanding of sphingolipid metabolism and profile will likely become of great importance in the clinic for both chemotherapy and diagnostics of cancer. The goal of this paper is to provide a comprehensive review of the current state of chemotherapeutic agents that target sphingolipid metabolism that are undergoing clinical trials. Additionally, we will formulate questions involving the use of sphingolipid metabolism as chemotherapeutic targets in need of further research.
Collapse
Affiliation(s)
- Alexander Kroll
- School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Hwang Eui Cho
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| | - Min H Kang
- Cancer Center, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Department of Pediatrics, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, United States
| |
Collapse
|
11
|
Schneider G. S1P Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:129-153. [PMID: 32030688 DOI: 10.1007/978-3-030-35582-1_7] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sphingosine-1-phosphate (S1P), together with other phosphosphingolipids, has been found to regulate complex cellular function in the tumor microenvironment (TME) where it acts as a signaling molecule that participates in cell-cell communication. S1P, through intracellular and extracellular signaling, was found to promote tumor growth, angiogenesis, chemoresistance, and metastasis; it also regulates anticancer immune response, modulates inflammation, and promotes angiogenesis. Interestingly, cancer cells are capable of releasing S1P and thus modifying the behavior of the TME components in a way that contributes to tumor growth and progression. Therefore, S1P is considered an important therapeutic target, and several anticancer therapies targeting S1P signaling are being developed and tested in clinics.
Collapse
Affiliation(s)
- Gabriela Schneider
- James Graham Brown Cancer Center, Division of Medical Oncology & Hematology, Department of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
12
|
Viltard M, Durand S, Pérez-Lanzón M, Aprahamian F, Lefevre D, Leroy C, Madeo F, Kroemer G, Friedlander G. The metabolomic signature of extreme longevity: naked mole rats versus mice. Aging (Albany NY) 2019; 11:4783-4800. [PMID: 31346149 PMCID: PMC6682510 DOI: 10.18632/aging.102116] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/16/2019] [Indexed: 04/11/2023]
Abstract
The naked mole-rat (Heterocephalus glaber) is characterized by a more than tenfold higher life expectancy compared to another rodent species of the same size, namely, the laboratory mouse (Mus musculus). We used mass spectrometric metabolomics to analyze circulating plasma metabolites in both species at different ages. Interspecies differences were much more pronounced than age-associated alterations in the metabolome. Such interspecies divergences affected multiple metabolic pathways involving amino, bile and fatty acids as well as monosaccharides and nucleotides. The most intriguing metabolites were those that had previously been linked to pro-health and antiaging effects in mice and that were significantly increased in the long-lived rodent compared to its short-lived counterpart. This pattern applies to α-tocopherol (also known as vitamin E) and polyamines (in particular cadaverine, N8-acetylspermidine and N1,N8-diacetylspermidine), all of which were more abundant in naked mole-rats than in mice. Moreover, the age-associated decline in spermidine and N1-acetylspermidine levels observed in mice did not occur, or is even reversed (in the case of N1-acetylspermidine) in naked mole-rats. In short, the present metabolomics analysis provides a series of testable hypotheses to explain the exceptional longevity of naked mole-rats.
Collapse
Affiliation(s)
- Mélanie Viltard
- Fondation pour la Recherche en Physiologie, Brussels, Belgium
| | - Sylvère Durand
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Maria Pérez-Lanzón
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Faculté de Médecine, Université de Paris Saclay, Kremlin Bicêtre, France
| | - Fanny Aprahamian
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Deborah Lefevre
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
| | - Christine Leroy
- INSERM UMR_S1151 CNRS UMR8253 Institut Necker-Enfants Malades (INEM), Paris, France
| | - Frank Madeo
- Institute of Molecular Biosciences, University of Graz, NAWI Graz, Graz, Austria
- BioTechMed Graz, Graz, Austria
| | - Guido Kroemer
- Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Villejuif, France
- Equipe Labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France
- Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France
- Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| | - Gérard Friedlander
- INSERM UMR_S1151 CNRS UMR8253 Institut Necker-Enfants Malades (INEM), Paris, France
- Service de Physiologie et Explorations Fonctionnelles, Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université de Paris - Paris Descartes, Faculté de Médecine, Paris, France
| |
Collapse
|
13
|
Activation of sphingosine kinase by lipopolysaccharide promotes prostate cancer cell invasion and metastasis via SphK1/S1PR4/matriptase. Oncogene 2019; 38:5580-5598. [DOI: 10.1038/s41388-019-0833-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 02/23/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
|
14
|
Chang YC, Chuang HL, Yin JH, Liao JW, Chen TH, Wang YC. Significance of sphingosine kinase 1 expression in feline mammary tumors. BMC Vet Res 2019; 15:155. [PMID: 31101115 PMCID: PMC6525354 DOI: 10.1186/s12917-019-1883-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/25/2019] [Indexed: 11/20/2022] Open
Abstract
Background Sphingosine kinase 1 (SPHK1) is an enzyme that converts pro-apoptotic ceramide and sphingosine into anti-apoptotic sphingosine-1-phosphate. There is growing evidence that SPHK1 activation promotes oncogenic transformation, tumor growth, chemotherapy resistance, and metastatic spread. High SPHK1 expression has been associated with a poor prognosis in several human cancers. Results In the present study, the expression level of SPHK1 was examined in feline mammary tumor (FMT) specimens, and the IHC expression level of SPHK1 was associated with the histological grade of FMTs. IHC analysis of 88 FMT cases revealed that the expression level of SPHK1 was upregulated in 53 tumor tissues (60.2%) compared to adjacent mammary tissues. SPHK1 expression in FMTs was significantly associated with histological grade, presence of lymphovascular invasion, and estrogen receptor negativity. Treatment of primary FMT cells with SPHK1 inhibitors reduced cell viability, indicating that SPHK1 acts to promote FMT cell survival. These results indicate that SPHK1 may play an important role in FMTs and may be a therapeutic target in cats with FMT. Conclusions SPHK1 over-expression in breast cancer tissues is associated with a poor prognosis in humans. SPHK1 over-expression in more aggressive FMTs provides support for a potential role of SPHK1 inhibitors for the treatment of FMTs. Targeting SPHK1 has potent cytotoxic effects in primary FMT cells. These findings suggest that further examination of the role SPHK1 plays in FMTs will pave the way for the investigation of SPHK1 inhibitors in future clinical applications. Electronic supplementary material The online version of this article (10.1186/s12917-019-1883-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yi-Chih Chang
- Department of Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
| | - Hsiao-Li Chuang
- National Laboratory Animal Center, National Applied Research Laboratories, Taipei, Taiwan
| | - Ji-Hang Yin
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Yu-Chih Wang, 145 Xingda Rd., South Dist, Taichung City, 402, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Yu-Chih Wang, 145 Xingda Rd., South Dist, Taichung City, 402, Taiwan
| | - Ter-Hsin Chen
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Yu-Chih Wang, 145 Xingda Rd., South Dist, Taichung City, 402, Taiwan
| | - Yu-Chih Wang
- Graduate Institute of Veterinary Pathobiology, National Chung Hsing University, Yu-Chih Wang, 145 Xingda Rd., South Dist, Taichung City, 402, Taiwan.
| |
Collapse
|
15
|
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
|
16
|
Hart PC, Chiyoda T, Liu X, Weigert M, Curtis M, Chiang CY, Loth R, Lastra R, McGregor SM, Locasale JW, Lengyel E, Romero IL. SPHK1 Is a Novel Target of Metformin in Ovarian Cancer. Mol Cancer Res 2019; 17:870-881. [PMID: 30655321 DOI: 10.1158/1541-7786.mcr-18-0409] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/17/2018] [Accepted: 01/08/2019] [Indexed: 12/12/2022]
Abstract
The role of phospholipid signaling in ovarian cancer is poorly understood. Sphingosine-1-phosphate (S1P) is a bioactive metabolite of sphingosine that has been associated with tumor progression through enhanced cell proliferation and motility. Similarly, sphingosine kinases (SPHK), which catalyze the formation of S1P and thus regulate the sphingolipid rheostat, have been reported to promote tumor growth in a variety of cancers. The findings reported here show that exogenous S1P or overexpression of SPHK1 increased proliferation, migration, invasion, and stem-like phenotypes in ovarian cancer cell lines. Likewise, overexpression of SPHK1 markedly enhanced tumor growth in a xenograft model of ovarian cancer, which was associated with elevation of key markers of proliferation and stemness. The diabetes drug, metformin, has been shown to have anticancer effects. Here, we found that ovarian cancer patients taking metformin had significantly reduced serum S1P levels, a finding that was recapitulated when ovarian cancer cells were treated with metformin and analyzed by lipidomics. These findings suggested that in cancer the sphingolipid rheostat may be a novel metabolic target of metformin. In support of this, metformin blocked hypoxia-induced SPHK1, which was associated with inhibited nuclear translocation and transcriptional activity of hypoxia-inducible factors (HIF1α and HIF2α). Further, ovarian cancer cells with high SPHK1 were found to be highly sensitive to the cytotoxic effects of metformin, whereas ovarian cancer cells with low SPHK1 were resistant. Together, the findings reported here show that hypoxia-induced SPHK1 expression and downstream S1P signaling promote ovarian cancer progression and that tumors with high expression of SPHK1 or S1P levels might have increased sensitivity to the cytotoxic effects of metformin. IMPLICATIONS: Metformin targets sphingolipid metabolism through inhibiting SPHK1, thereby impeding ovarian cancer cell migration, proliferation, and self-renewal.
Collapse
Affiliation(s)
- Peter C Hart
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois.,Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Xiaojing Liu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Melanie Weigert
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Marion Curtis
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Chun-Yi Chiang
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Rachel Loth
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Ricardo Lastra
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Stephanie M McGregor
- Department of Pathology, The University of Chicago, Chicago, Illinois.,Department of Pathology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jason W Locasale
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois
| | - Iris L Romero
- Department of Obstetrics and Gynecology, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
17
|
Alshaker H, Srivats S, Monteil D, Wang Q, Low CMR, Pchejetski D. Field template-based design and biological evaluation of new sphingosine kinase 1 inhibitors. Breast Cancer Res Treat 2018; 172:33-43. [PMID: 30043096 PMCID: PMC6208908 DOI: 10.1007/s10549-018-4900-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/19/2018] [Indexed: 11/29/2022]
Abstract
Purpose Sphingosine kinase 1 (SK1) is a protooncogenic enzyme expressed in many human tumours and is associated with chemoresistance and poor prognosis. It is a potent therapy target and its inhibition chemosensitises solid tumours. Despite recent advances in SK1 inhibitors synthesis and validation, their clinical safety and chemosensitising options are not well described. In this study, we have designed, synthesised and tested a new specific SK1 inhibitor with a low toxicity profile. Methods Field template molecular modelling was used for compound design. Lead compounds were tested in cell and mouse cancer models. Results Field template analysis of three known SK1 inhibitors, SKI-178, 12aa and SK1-I, was performed and compound screening identified six potential new SK1 inhibitors. SK1 activity assays in both cell-free and in vitro settings showed that two compounds were effective SK1 inhibitors. Compound SK-F has potently decreased cancer cell viability in vitro and sensitised mouse breast tumours to docetaxel (DTX) in vivo, without significant whole-body toxicity. Conclusion Through field template screening, we have identified a new SK1 inhibitor, SK-F, which demonstrated antitumour activity in vitro and in vivo without overt toxicity when combined with DTX. Electronic supplementary material The online version of this article (10.1007/s10549-018-4900-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Heba Alshaker
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK. .,Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.
| | - Shyam Srivats
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Danielle Monteil
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - Qi Wang
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK
| | | | - Dmitri Pchejetski
- School of Medicine, University of East Anglia, 2.53 BCRE, Norwich Research Park, Norwich, NR47UQ, UK.
| |
Collapse
|
18
|
Pirro M, Ricciuti B, Rader DJ, Catapano AL, Sahebkar A, Banach M. High density lipoprotein cholesterol and cancer: Marker or causative? Prog Lipid Res 2018; 71:54-69. [DOI: 10.1016/j.plipres.2018.06.001] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 05/15/2018] [Accepted: 06/02/2018] [Indexed: 12/11/2022]
|
19
|
Bandu R, Mok HJ, Kim KP. Phospholipids as cancer biomarkers: Mass spectrometry-based analysis. MASS SPECTROMETRY REVIEWS 2018; 37:107-138. [PMID: 27276657 DOI: 10.1002/mas.21510] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 05/19/2016] [Indexed: 05/02/2023]
Abstract
Lipids, particularly phospholipids (PLs), are key components of cellular membrane. PLs play important and diverse roles in cells such as chemical-energy storage, cellular signaling, cell membranes, and cell-cell interactions in tissues. All these cellular processes are pertinent to cells that undergo transformation, cancer progression, and metastasis. Thus, there is a strong possibility that some classes of PLs are expected to present in cancer cells and tissues in cellular physiology. The mass spectrometric soft-ionization techniques, electrospray ionization (ESI), and matrix-assisted laser desorption/ionization (MALDI) are well-established in the proteomics field, have been used for lipidomic analysis in cancer research. This review focused on the applications of mass spectrometry (MS) mainly on ESI-MS and MALDI-MS in the structural characterization, molecular composition and key roles of various PLs present in cancer cells, tissues, blood, and urine, and on their importance for cancer-related problems as well as challenges for development of novel PL-based biomarkers. The profiling of PLs helps to rationalize their functions in biological systems, and will also provide diagnostic information to elucidate mechanisms behind the control of cancer, diabetes, and neurodegenerative diseases. The investigation of cellular PLs with MS methods suggests new insights on various cancer diseases and clinical applications in the drug discovery and development of biomarkers for various PL-related different cancer diseases. PL profiling in tissues, cells and body fluids also reflect the general condition of the whole organism and can indicate the existence of cancer and other diseases. PL profiling with MS opens new prospects to assess alterations of PLs in cancer, screening specific biomarkers and provide a basis for the development of novel therapeutic strategies. © 2016 Wiley Periodicals, Inc. Mass Spec Rev 37:107-138, 2018.
Collapse
Affiliation(s)
- Raju Bandu
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yong-in City, 446-701, Korea
| | - Hyuck Jun Mok
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yong-in City, 446-701, Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yong-in City, 446-701, Korea
| |
Collapse
|
20
|
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
|
21
|
The Role of Sphingosine-1-Phosphate and Ceramide-1-Phosphate in Inflammation and Cancer. Mediators Inflamm 2017; 2017:4806541. [PMID: 29269995 PMCID: PMC5705877 DOI: 10.1155/2017/4806541] [Citation(s) in RCA: 140] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 08/01/2017] [Accepted: 08/30/2017] [Indexed: 01/02/2023] Open
Abstract
Inflammation is part of our body's response to tissue injury and pathogens. It helps to recruit various immune cells to the site of inflammation and activates the production of mediators to mobilize systemic protective processes. However, chronic inflammation can increase the risk of diseases like cancer. Apart from cytokines and chemokines, lipid mediators, particularly sphingosine-1-phosphate (S1P) and ceramide-1-phosphate (C1P), contribute to inflammation and cancer. S1P is an important player in inflammation-associated colon cancer progression. On the other hand, C1P has been recognized to be involved in cancer cell growth, migration, survival, and inflammation. However, whether C1P is involved in inflammation-associated cancer is not yet established. In contrast, few studies have also suggested that S1P and C1P are involved in anti-inflammatory pathways regulated in certain cell types. Ceramide is the substrate for ceramide kinase (CERK) to yield C1P, and sphingosine is phosphorylated to S1P by sphingosine kinases (SphKs). Biological functions of sphingolipid metabolites have been studied extensively. Ceramide is associated with cell growth inhibition and enhancement of apoptosis while S1P and C1P are associated with enhancement of cell growth and survival. Altogether, S1P and C1P are important regulators of ceramide level and cell fate. This review focuses on S1P and C1P involvement in inflammation and cancer with emphasis on recent progress in the field.
Collapse
|
22
|
Bao Y, Guo Y, Zhang C, Fan F, Yang W. Sphingosine Kinase 1 and Sphingosine-1-Phosphate Signaling in Colorectal Cancer. Int J Mol Sci 2017; 18:ijms18102109. [PMID: 28991193 PMCID: PMC5666791 DOI: 10.3390/ijms18102109] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Revised: 09/23/2017] [Accepted: 09/30/2017] [Indexed: 12/15/2022] Open
Abstract
Sphingosine kinase 1 (Sphk1) is a highly conserved lipid kinase that phosphorylates sphingosine to form sphingosine-1-phosphate (S1P). Growing studies have demonstrated that Sphk1 is overexpressed in various types of solid cancers and can be induced by growth factors, cytokines, and carcinogens, leading to the increase of S1P production. Subsequently, the increased Sphk1/S1P facilitates cancer cell proliferation, mobility, angiogenesis, invasion, and metastasis. Therefore, Sphk1/S1P signaling plays oncogenic roles. This review summarizes the features of Sphk1/S1P signaling and their functions in colorectal cancer cell growth, tumorigenesis, and metastasis, as well as the possible underlying mechanisms.
Collapse
Affiliation(s)
- Yonghua Bao
- Institute of Precision Medicine, Jining Medical University, Jining 272067, China.
| | - Yongchen Guo
- Institute of Precision Medicine, Jining Medical University, Jining 272067, China.
| | - Chenglan Zhang
- Department of Nursing, Health Professional College of Heilongjiang Province, Beian 164000, China.
| | - Fenghua Fan
- Department of Nursing, Health Professional College of Heilongjiang Province, Beian 164000, China.
| | - Wancai Yang
- Institute of Precision Medicine, Jining Medical University, Jining 272067, China.
- Department of Pathology, University of Illinois at Chicago, Chicago 60612, IL, USA.
| |
Collapse
|
23
|
The novel sphingosine-1-phosphate receptors antagonist AD2900 affects lymphocyte activation and inhibits T-cell entry into the lymph nodes. Oncotarget 2017; 8:53563-53580. [PMID: 28881832 PMCID: PMC5581131 DOI: 10.18632/oncotarget.18626] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 05/28/2017] [Indexed: 11/25/2022] Open
Abstract
Sphingolipid derivatives play key roles in immune cell migration and function. Synthetic sphingolipid analogues are used as therapeutics to intervene various inflammatory and malignant conditions. We hypothesize that different analogs have different effects on immune cells and therefore can be used as treatment for specific diseases. This study examines the properties of the novel synthetic sphingolipid analog, AD2900, and its effects on immune cell activation and lymphocyte localization in homeostasis. AD2900 is an antagonist for all sphingosine-1-phosphate (S1P) receptors. It demonstrates a significant inhibitory effect on the proliferation of activated human peripheral blood mononuclear cells, which is dependent on cAMP reduction and calcium signal transduction but not on phospholipase C activation. AD2900 causes a significant but reversible downregulation of S1P1 expression on the cell surface. AD2900 administration to C57BL/6J mice leads to the accumulation of T cells in the blood and spleen and in turn reduces T-cell number in the lymph nodes. Moreover, AD2900 treatment shows significant effects on the localization of T-cell subpopulations. These results demonstrate the key roles of S1P in T-cell trafficking in a steady state and suggest a potential clinical application for AD2900. Notably, this sphingolipid analog does not cause a severe lymphopenia. The clinical effect of AD2900 in hemato-oncologic diseases and immune-related diseases needs further investigation.
Collapse
|
24
|
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
|
25
|
Grammatikos G, Schoell N, Ferreirós N, Bon D, Herrmann E, Farnik H, Köberle V, Piiper A, Zeuzem S, Kronenberger B, Waidmann O, Pfeilschifter J. Serum sphingolipidomic analyses reveal an upregulation of C16-ceramide and sphingosine-1-phosphate in hepatocellular carcinoma. Oncotarget 2017; 7:18095-105. [PMID: 26933996 PMCID: PMC4951274 DOI: 10.18632/oncotarget.7741] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 02/02/2016] [Indexed: 01/22/2023] Open
Abstract
We have recently shown that major alterations of serum sphingolipid metabolites in chronic liver disease associate significantly with the stage of liver fibrosis in corresponding patients. In the current study we assessed via mass spectrometry serum concentrations of sphingolipid metabolites in a series of 122 patients with hepatocellular carcinoma (HCC) compared to an age- and sex-matched series of 127 patients with cirrhosis. We observed a highly significant upregulation of long and very long chain ceramides (C16-C24) in the serum of patients with HCC as compared to patients with cirrhosis (P < 0.001). Accordingly, dihydro-ceramides, synthetic precursors of ceramides and notably sphingosine, sphingosine-1-phosphate (S1P) and sphinganine-1-phosphate (SA1P) were upregulated in patients with HCC (P < 0.001). Especially the diagnostic accuracy of C16-ceramide and S1P, assessed by receiver operating curve (ROC) analysis, showed a higher area under the curve (AUC) value as compared to alpha fetoprotein (AFP) (0.999 and 0.985 versus 0.823, P < 0.001 respectively). In conclusion, serum levels of sphingolipid metabolites show a significant upregulation in patients with HCC as compared to patients with cirrhosis. Particularly C16-ceramide and S1P may serve as novel diagnostic markers for the identification of HCC in patients with liver diseases. Our data justify further investigations on the role of sphingolipids in HCC.
Collapse
Affiliation(s)
- Georgios Grammatikos
- Goethe University Hospital, Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany.,Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Niklas Schoell
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Nerea Ferreirós
- Pharmazentrum Frankfurt, Institut für klinische Pharmakologie, Goethe University Hospital, Frankfurt am Main, Germany
| | - Dimitra Bon
- Goethe University, Department of Medicine, Institute of Biostatistics and Mathematical Modelling, Frankfurt am Main, Germany
| | - Eva Herrmann
- Goethe University, Department of Medicine, Institute of Biostatistics and Mathematical Modelling, Frankfurt am Main, Germany
| | - Harald Farnik
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Verena Köberle
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Albrecht Piiper
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Stefan Zeuzem
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Bernd Kronenberger
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Oliver Waidmann
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Goethe University Hospital, Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany
| |
Collapse
|
26
|
Rodriguez YI, Campos LE, Castro MG, Aladhami A, Oskeritzian CA, Alvarez SE. Sphingosine-1 Phosphate: A New Modulator of Immune Plasticity in the Tumor Microenvironment. Front Oncol 2016; 6:218. [PMID: 27800303 PMCID: PMC5066089 DOI: 10.3389/fonc.2016.00218] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/30/2016] [Indexed: 01/01/2023] Open
Abstract
In the last 15 years, increasing evidences demonstrate a strong link between sphingosine-1-phosphate (S1P) and both normal physiology and progression of different diseases, including cancer and inflammation. Indeed, numerous studies show that tissue levels of this sphingolipid metabolite are augmented in many cancers, affecting survival, proliferation, angiogenesis, and metastatic spread. Recent insights into the possible role of S1P as a therapeutic target has attracted enormous attention and opened new opportunities in this evolving field. In this review, we will focus on the role of S1P in cancer, with particular emphasis in new developments that highlight the many functions of this sphingolipid in the tumor microenvironment. We will discuss how S1P modulates phenotypic plasticity of macrophages and mast cells, tumor-induced immune evasion, differentiation and survival of immune cells in the tumor milieu, interaction between cancer and stromal cells, and hypoxic response.
Collapse
Affiliation(s)
- Yamila I Rodriguez
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL) CONICET , San Luis , Argentina
| | - Ludmila E Campos
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL) CONICET , San Luis , Argentina
| | - Melina G Castro
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL) CONICET , San Luis , Argentina
| | - Ahmed Aladhami
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, SC , USA
| | - Carole A Oskeritzian
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine , Columbia, SC , USA
| | - Sergio E Alvarez
- Instituto Multidisciplinario de Investigaciones Biológicas San Luis (IMIBIO-SL) CONICET, San Luis, Argentina; Universidad Nacional de San Luis, San Luis, Argentina
| |
Collapse
|
27
|
Insights into the molecular roles of heparan sulfate proteoglycans (HSPGs—syndecans) in autocrine and paracrine growth factor signaling in the pathogenesis of Hodgkin’s lymphoma. Tumour Biol 2016; 37:11573-11588. [DOI: 10.1007/s13277-016-5118-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 06/09/2016] [Indexed: 12/25/2022] Open
|
28
|
Nema R, Vishwakarma S, Agarwal R, Panday RK, Kumar A. Emerging role of sphingosine-1-phosphate signaling in head and neck squamous cell carcinoma. Onco Targets Ther 2016; 9:3269-80. [PMID: 27330306 PMCID: PMC4898435 DOI: 10.2147/ott.s99989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is the sixth most frequent cancer type, with an annual incidence of approximately half a million people worldwide. It has a high recurrence rate and an extremely low survival rate. This is due to limited availability of effective therapies to reduce the rate of recurrence, resulting in high morbidity and mortality of patients with advanced stages of the disease. HNSCC often develops resistance to chemotherapy and targeted drug therapy. Thus, to overcome the problem of drug resistance, there is a need to explore novel drug targets. Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid involved in inflammation, tumor progression, and angiogenesis. S1P is synthesized intracellularly by two sphingosine kinases (SphKs). It can be exported to the extracellular space, where it can activate a family of G-protein-coupled receptors. Alternatively, S1P can act as an intracellular second messenger. SphK1 regulates tumor progression, invasion, metastasis, and chemoresistance in HNSCC. SphK1 expression is highly elevated in advanced stage HNSCC tumors and correlates with poor survival. In this article, we review current knowledge regarding the role of S1P receptors and enzymes of S1P metabolism in HNSCC carcinogenesis. Furthermore, we summarize the current perspectives on therapeutic approaches for targeting S1P pathway for treating HNSCC.
Collapse
Affiliation(s)
- Rajeev Nema
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, India
| | - Supriya Vishwakarma
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, India
| | - Rahul Agarwal
- Jawaharlal Nehru Cancer Hospital & Research Centre, Indrapuri, Bhopal, India
| | | | - Ashok Kumar
- Department of Biochemistry, All India Institute of Medical Sciences Bhopal, Bhopal, India
| |
Collapse
|
29
|
Messias CV, Santana-Van-Vliet E, Lemos JP, Moreira OC, Cotta-de-Almeida V, Savino W, Mendes-da-Cruz DA. Sphingosine-1-Phosphate Induces Dose-Dependent Chemotaxis or Fugetaxis of T-ALL Blasts through S1P1 Activation. PLoS One 2016; 11:e0148137. [PMID: 26824863 PMCID: PMC4732661 DOI: 10.1371/journal.pone.0148137] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/13/2016] [Indexed: 01/08/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive sphingolipid involved in several physiological processes including cell migration and differentiation. S1P signaling is mediated through five G protein-coupled receptors (S1P1-S1P5). S1P1 is crucial to the exit of T-lymphocytes from the thymus and peripheral lymphoid organs through a gradient of S1P. We have previously observed that T-ALL and T-LBL blasts express S1P1. Herein we analyzed the role of S1P receptors in the migratory pattern of human T-cell neoplastic blasts. S1P-triggered cell migration was directly related to S1P1 expression. T-ALL blasts expressing low levels of S1P1 mRNA (HPB-ALL) did not migrate toward S1P, whereas those expressing higher levels of S1P1 (MOLT-4, JURKAT and CEM) did migrate. The S1P ligand induced T-ALL cells chemotaxis in concentrations up to 500 nM and induced fugetaxis in higher concentrations (1000-10000 nM) through interactions with S1P1. When S1P1 was specifically blocked by the W146 compound, S1P-induced migration at lower concentrations was reduced, whereas higher concentrations induced cell migration. Furthermore, we observed that S1P/S1P1 interactions induced ERK and AKT phosphorylation, and modulation of Rac1 activity. Responding T-ALL blasts also expressed S1P3 mRNA but blockage of this receptor did not modify migratory responses. Our results indicate that S1P is involved in the migration of T-ALL/LBL blasts, which is dependent on S1P1 expression. Moreover, S1P concentrations in the given microenvironment might induce dose-dependent chemotaxis or fugetaxis of T-ALL blasts.
Collapse
Affiliation(s)
- Carolina V. Messias
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Eliane Santana-Van-Vliet
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia P. Lemos
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Otacilio C. Moreira
- Laboratory of Molecular Biology and Endemic Diseases, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Wilson Savino
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniella Arêas Mendes-da-Cruz
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
30
|
Gao Y, Gao F, Chen K, Tian ML, Zhao DL. Sphingosine kinase 1 as an anticancer therapeutic target. DRUG DESIGN DEVELOPMENT AND THERAPY 2015; 9:3239-45. [PMID: 26150697 PMCID: PMC4484649 DOI: 10.2147/dddt.s83288] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The development of chemotherapeutic resistance is a major challenge in oncology. Elevated sphingosine kinase 1 (SK1) levels is predictive of a poor prognosis, and SK1 overexpression may confer resistance to chemotherapeutics. The SK/sphingosine-1-phosphate (S1P)/sphingosine-1-phosphate receptor (S1PR) signaling pathway has been implicated in the progression of various cancers and in chemotherapeutic drug resistance. Therefore, SK1 may represent an important target for cancer therapy. Targeting the SK/S1P/S1PR signaling pathway may be an effective anticancer therapeutic strategy, particularly in the context of overcoming drug resistance. This review summarizes our current understanding of the role of SK/S1P/S1PR signaling in cancer and development of SK1 inhibitors.
Collapse
Affiliation(s)
- Ying Gao
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Fei Gao
- Department of Neurology, First Affiliated Hospital of Xi'an Medical University, Xi'an, People's Republic of China
| | - Kan Chen
- School of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, People's Republic of China
| | - Mei-li Tian
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Dong-li Zhao
- Department of Radiotherapy Oncology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
31
|
Furman D. Sexual dimorphism in immunity: improving our understanding of vaccine immune responses in men. Expert Rev Vaccines 2014; 14:461-71. [PMID: 25278153 DOI: 10.1586/14760584.2015.966694] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Weaker immune responses are often observed in males compared to females. Since female hormones have proinflammatory properties and androgens have potent immunomodulatory effects, this sexual dimorphism in the immune response seems to be hormone dependent. Despite our current knowledge about the effect of sex hormones on immune cells, definition of the factors driving the sex differences in immunoclinical outcomes, such as the diminished response to infection and vaccination observed in men or the higher rates of autoimmunity observed in females, remains elusive. Recently, systems approaches to immune function have started to suggest a way toward establishing this connection. Such studies promise to improve our understanding of the mechanisms underlying the sexual dimorphism observed in the human immune system.
Collapse
Affiliation(s)
- David Furman
- Institute for Immunity, Transplantation and Infection, Stanford University, 279 Campus Drive, B240 Beckman Center, Stanford, CA 94305-5124, USA
| |
Collapse
|
32
|
Wilkerson BA, Argraves KM. The role of sphingosine-1-phosphate in endothelial barrier function. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1841:1403-1412. [PMID: 25009123 PMCID: PMC4169319 DOI: 10.1016/j.bbalip.2014.06.012] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Revised: 06/19/2014] [Accepted: 06/26/2014] [Indexed: 02/08/2023]
Abstract
Loss of endothelial barrier function is implicated in the etiology of metastasis, atherosclerosis, sepsis and many other diseases. Studies suggest that sphingosine-1-phosphate (S1P), particularly HDL-bound S1P (HDL-S1P) is essential for endothelial barrier homeostasis and that HDL-S1P may be protective against the loss of endothelial barrier function in disease. This review summarizes evidence providing mechanistic insights into how S1P maintains endothelial barrier function, highlighting the recent findings that implicate the major S1P carrier, HDL, in the maintenance of the persistent S1P-signaling needed to maintain endothelial barrier function. We review the mechanisms proposed for HDL maintenance of persistent S1P-signaling, the evidence supporting these mechanisms and the remaining fundamental questions.
Collapse
Affiliation(s)
- Brent A Wilkerson
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA
| | - Kelley M Argraves
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, 173 Ashley Ave., BSB650, Charleston, SC 29425, USA.
| |
Collapse
|
33
|
Taniguchi M, Okazaki T. The role of sphingomyelin and sphingomyelin synthases in cell death, proliferation and migration—from cell and animal models to human disorders. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:692-703. [DOI: 10.1016/j.bbalip.2013.12.003] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 12/16/2022]
|
34
|
Grammatikos G, Mühle C, Ferreiros N, Schroeter S, Bogdanou D, Schwalm S, Hintereder G, Kornhuber J, Zeuzem S, Sarrazin C, Pfeilschifter J. Serum acid sphingomyelinase is upregulated in chronic hepatitis C infection and non alcoholic fatty liver disease. Biochim Biophys Acta Mol Cell Biol Lipids 2014; 1841:1012-20. [PMID: 24769340 DOI: 10.1016/j.bbalip.2014.04.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 04/08/2014] [Accepted: 04/17/2014] [Indexed: 02/07/2023]
Abstract
UNLABELLED Sphingolipids constitute bioactive molecules with functional implications in homeostasis and pathogenesis of various diseases. However, the role of sphingolipids as possible disease biomarkers in chronic liver disease remains largely unexplored. In the present study we used mass spectrometry and spectrofluorometry methods in order to quantify various sphingolipid metabolites and also assess the activity of an important corresponding regulating enzyme in the serum of 72 healthy volunteers as compared to 69 patients with non-alcoholic fatty liver disease and 69 patients with chronic hepatitis C virus infection. Our results reveal a significant upregulation of acid sphingomyelinase in the serum of patients with chronic liver disease as compared to healthy individuals (p<0.001). Especially in chronic hepatitis C infection acid sphingomyelinase activity correlated significantly with markers of hepatic injury (r=0.312, p=0.009) and showed a high discriminative power. Accumulation of various (dihydro-) ceramide species was identified in the serum of patients with non-alcoholic fatty liver disease (p<0.001) and correlated significantly to cholesterol (r=0.448, p<0.001) but showed a significant accumulation in patients with normal cholesterol values as well (p<0.001). Sphingosine, a further bioactive metabolite, was also upregulated in chronic liver disease (p<0.001). However, no significant correlation to markers of hepatic injury was identified. CONCLUSION Chronic hepatitis C virus infection and non-alcoholic fatty liver disease induce a significant upregulation of serum acid sphingomyelinase which appears as a novel biomarker in chronic hepatopathies. Further studies are required to elucidate the potential of the sphingolipid signaling pathway as putative therapeutic target in chronic liver disease.
Collapse
Affiliation(s)
- Georgios Grammatikos
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany; Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany.
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Nerea Ferreiros
- Pharmazentrum Frankfurt, Institut für klinische Pharmakologie, Goethe University Hospital, Frankfurt am Main, Germany
| | - Sirkka Schroeter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany; Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Dimitra Bogdanou
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Stephanie Schwalm
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany
| | - Gudrun Hintereder
- Zentrallabor, Goethe University Hospital, Frankfurt am Main, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University Erlangen-Nuremberg, Germany
| | - Stefan Zeuzem
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Christoph Sarrazin
- Goethe University Hospital, Medizinische Klinik 1, Frankfurt am Main, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie und Toxikologie, Frankfurt am Main, Germany
| |
Collapse
|
35
|
Don AS, Lim XY, Couttas TA. Re-configuration of sphingolipid metabolism by oncogenic transformation. Biomolecules 2014; 4:315-53. [PMID: 24970218 PMCID: PMC4030989 DOI: 10.3390/biom4010315] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 02/11/2014] [Accepted: 02/27/2014] [Indexed: 12/15/2022] Open
Abstract
The sphingolipids are one of the major lipid families in eukaryotes, incorporating a diverse array of structural variants that exert a powerful influence over cell fate and physiology. Increased expression of sphingosine kinase 1 (SPHK1), which catalyses the synthesis of the pro-survival, pro-angiogenic metabolite sphingosine 1-phosphate (S1P), is well established as a hallmark of multiple cancers. Metabolic alterations that reduce levels of the pro-apoptotic lipid ceramide, particularly its glucosylation by glucosylceramide synthase (GCS), have frequently been associated with cancer drug resistance. However, the simple notion that the balance between ceramide and S1P, often referred to as the sphingolipid rheostat, dictates cell survival contrasts with recent studies showing that highly potent and selective SPHK1 inhibitors do not affect cancer cell proliferation or survival, and studies demonstrating higher ceramide levels in some metastatic cancers. Recent reports have implicated other sphingolipid metabolic enzymes such as acid sphingomyelinase (ASM) more strongly in cancer pathogenesis, and highlight lysosomal sphingolipid metabolism as a possible weak point for therapeutic targeting in cancer. This review describes the evidence implicating different sphingolipid metabolic enzymes and their products in cancer pathogenesis, and suggests how newer systems-level approaches may improve our overall understanding of how oncogenic transformation reconfigures sphingolipid metabolism.
Collapse
Affiliation(s)
- Anthony S Don
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Xin Y Lim
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| | - Timothy A Couttas
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
36
|
Sphingosine kinase 1 and cancer: a systematic review and meta-analysis. PLoS One 2014; 9:e90362. [PMID: 24587339 PMCID: PMC3937388 DOI: 10.1371/journal.pone.0090362] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/29/2014] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Sphingosine kinase 1 (SK1) is a key regulator of the dynamic ceramide/sphingosine 1-phosphate rheostat balance and important in the pathological cancer genesis, progression, and metastasis processes. Many studies have demonstrated SK1 overexpressed in various cancers, but no meta-analysis has evaluated the relationship between SK1 and various cancers. METHODS We retrieved relevant articles from the PubMed, EBSCO, ISI, and OVID databases. A pooled odds ratio (OR) was used to assess the associations between SK1 expression and cancer; hazard ratios (HR) were used for 5-year and overall survival. Review Manager 5.0 was used for the meta-analysis, and publication bias was evaluated with STATA 12.0 (Egger's test). RESULTS Thirty-four eligible studies (n=4,673 patients) were identified. SK1 positivity and high expression were significantly different between cancer, non-cancer, and benign tissues. SK1 mRNA and protein expression levels were elevated in the cancer tissues, compared with the normal tissues. SK1 positivity rates differed between various cancer types (lowest [27.3%] in estrogen receptor-positive breast cancer and highest [82.2%] in tongue squamous cell carcinoma). SK1 positivity and high expression were associated with 5-year survival; the HR was 1.86 (95% confidence interval [CI], 1.18-2.94) for breast cancer, 1.58 (1.08-2.31) for gastric cancer, and 2.68 (2.10-3.44) for other cancers; the total cancer HR was 2.21 (95% CI, 1.83-2.67; P < 0.00001). The overall survival HRs were 2.09 (95% CI, 1.35-3.22), 1.56 (1.08-2.25), and 2.62 (2.05-3.35) in breast, gastric, and other cancers, respectively. The total effect HR was 2.21 (95% CI, 1.83-2.66; P < 0.00001). CONCLUSIONS SK1 positivity and high expression were significantly associated with cancer and a shorter 5-year and overall survival. SK1 positivity rates vary tremendously among the cancer types. It is necessary to further explore whether SK1 might be a predictive biomarker of outcomes in cancer patients.
Collapse
|
37
|
Huang YL, Chang CL, Tang CH, Lin YC, Ju TK, Huang WP, Lee H. Extrinsic sphingosine 1-phosphate activates S1P5 and induces autophagy through generating endoplasmic reticulum stress in human prostate cancer PC-3 cells. Cell Signal 2013; 26:611-8. [PMID: 24333325 DOI: 10.1016/j.cellsig.2013.11.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 11/27/2013] [Accepted: 11/27/2013] [Indexed: 10/25/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lysophospholipid that binds to a family of G protein-coupled receptors (GPCRs), termed S1P1-S1P5. Our previous study has reported that S1P induces autophagy in human prostate cancer PC-3 cell. In addition, S1P-induced autophagy plays a prosurvival role in PC-3 cells. Accumulating evidence has shown that the autophagy responses triggered by ER stress signaling have cytoprotective effects. Thus, we attempted to investigate whether S1P-induced autophagy is a result of triggering ER stress in PC-3 cells. By monitoring XBP-1 mRNA splicing, a characteristic of ER stress, we demonstrate that S1P triggers ER stress in a concentration-dependent and time-dependent manner. Moreover, DiH S1P, a membrane-nonpermeable S1P analog without intracellular effects also enhances ER stress. Meanwhile, we also show that S1P5 is required for S1P-induced ER stress by using RNA interference experiments. Furthermore, signaling analyses revealed that PI3K, PLC, and ROS production were involved in S1P's effects on ER stress induction. On the other hand, knockdown of XBP-1 abolished S1P-induced autophagy. In summary, our results demonstrate for the first time that the extracellular S1P-triggered ER stress is responsible for autophagy induction in PC-3 cells.
Collapse
Affiliation(s)
- Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan, ROC; Graduate Institute of Basic Medical Science, China Medical University and Hospital, Taichung, Taiwan
| | - Chi-Lun Chang
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University and Hospital, Taichung, Taiwan
| | - Yueh-Chien Lin
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Tsai-Kai Ju
- Instrumentation Center, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Technology Commons, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC
| | - Wei-Pang Huang
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Technology Commons, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC.
| | - Hsinyu Lee
- Department of Life Science, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Institute of Zoology, College of Life Science, National Taiwan University, Taipei, Taiwan, ROC; Center for Biotechnology, National Taiwan University, Taipei, Taiwan, ROC; Angiogenesis Research Center, National Taiwan University, Taipei, Taiwan, ROC; Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan, ROC.
| |
Collapse
|
38
|
Pinho FG, Frampton AE, Nunes J, Krell J, Alshaker H, Jacob J, Pellegrino L, Roca-Alonso L, de Giorgio A, Harding V, Waxman J, Stebbing J, Pchejetski D, Castellano L. Downregulation of microRNA-515-5p by the estrogen receptor modulates sphingosine kinase 1 and breast cancer cell proliferation. Cancer Res 2013; 73:5936-48. [PMID: 23928990 DOI: 10.1158/0008-5472.can-13-0158] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Sphingosine kinase 1 (SK1) plays an important role in estrogen-dependent breast tumorigenesis, but its regulation is poorly understood. A subset of microRNAs (miRNA, miR) is regulated by estrogen and contributes to cellular proliferation and cancer progression. Here, we describe that miR-515-5p is transcriptionally repressed by estrogen receptor α (ERα) and functions as a tumor suppressor in breast cancer. Its downregulation enhances cell proliferation and estrogen-dependent SK1 activity, mediated by a reduction of miR-515-5p posttranscriptional repression. Enforced expression of miR-515-5p in breast cancer cells causes a reduction in SK1 activity, reduced cell proliferation, and the induction of caspase-dependent apoptosis. Conversely, opposing effects occur with miR-515-5p inhibition and by SK1 silencing. Notably, we show that estradiol (E2) treatment downregulates miR-515-5p levels, whereas the antiestrogen tamoxifen causes a decrease in SK1, which is rescued by silencing miR-515-5p. Analysis of chromatin immunoprecipitation sequencing (ChIP-Seq) data reveals that miR-515-5p suppression is mediated by a direct interaction of ERα within its promoter. RNA-sequencing (RNA-Seq) analysis of breast cancer cells after overexpressing miR-515-5p indicates that it partly modulates cell proliferation by regulating the Wnt pathway. The clinical implications of this novel regulatory system are shown as miR-515-5p is significantly downregulated in ER-positive (n = 146) compared with ER-negative (n = 98) breast cancers. Overall, we identify a new link between ERα, miR-515-5p, proliferation, and apoptosis in breast cancer tumorigenesis.
Collapse
Affiliation(s)
- Filipa G Pinho
- Authors' Affiliations: Division of Oncology, Department of Surgery & Cancer, Imperial Centre for Translational and Experimental Medicine (ICTEM); Department of Medicine and HPB Surgical Unit, Department of Surgery & Cancer, Imperial College; Department of Oncology, Imperial College Healthcare NHS Trust, Charing Cross Hospital, London; and School of Medicine, University of East Anglia, Norwich, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Alshaker H, Sauer L, Monteil D, Ottaviani S, Srivats S, Böhler T, Pchejetski D. Therapeutic potential of targeting SK1 in human cancers. Adv Cancer Res 2013; 117:143-200. [PMID: 23290780 DOI: 10.1016/b978-0-12-394274-6.00006-6] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sphingosine kinase 1 (SK1) is a lipid enzyme with oncogenic properties that converts the proapoptotic lipids ceramide and sphingosine into the antiapoptotic lipid sphingosine-1-phosphate and activates the signal transduction pathways that lead to cell proliferation, migration, the activation of the inflammatory response, and the impairment of apoptosis. There is compelling evidence that SK1 activation contributes to cancer progression leading to increased oncogenic transformation, tumor growth, resistance to therapies, tumor neovascularization, and metastatic spread. High levels of SK1 expression or activity have been associated with a poor prognosis in several human cancers. Recent studies using cancer cell and mouse models demonstrate a significant potential for SK1-targeting therapies to synergize with the effects of chemotherapy and radiotherapy; however, until recently the absence of clinically applicable SK1 inhibitors has limited the translation of these findings into patients. With the recent discovery of SK1 inhibiting properties of a clinically approved drug FTY720 (Fingolimod), SK1 has gained significant attention from both clinicians and the pharmaceutical industry and it is hoped that trials of newly developed SK1 inhibitors may follow soon. This review provides an overview of the SK1 signaling, its relevance to cancer progression, and the potential clinical significance of targeting SK1 for improved local or systemic control of human cancers.
Collapse
Affiliation(s)
- Heba Alshaker
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
The role of sphingolipids as bioactive signaling molecules that can regulate cell fate decisions puts them at center stage for cancer treatment and prevention. While ceramide and sphingosine have been established as antigrowth molecules, sphingosine-1-phosphate (S1P) offers a progrowth message to cells. The enzymes responsible for maintaining the balance between these "stop" or "go" signals are the sphingosine kinases (SK), SK1 and SK2. While the relative contribution of SK2 is still being elucidated and may involve an intranuclear role, a substantial amount of evidence suggests that regulation of sphingolipid levels by SK1 is an important component of carcinogenesis. Here, we review the literature regarding the role of SK1 as an oncogene that can function to enhance cancer cell viability and promote tumor growth and metastasis; highlighting the importance of developing specific SK1 inhibitors to supplement current cancer therapies.
Collapse
Affiliation(s)
- Linda A Heffernan-Stroud
- Molecular and Cellular Biology and Pathobiology Program, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
41
|
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
|
42
|
Jelonek K, Ros M, Pietrowska M, Widlak P. Cancer biomarkers and mass spectrometry-based analyses of phospholipids in body fluids. ACTA ACUST UNITED AC 2013. [DOI: 10.2217/clp.12.79] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
43
|
Pyne S, Edwards J, Ohotski J, Pyne NJ. Sphingosine 1-phosphate receptors and sphingosine kinase 1: novel biomarkers for clinical prognosis in breast, prostate, and hematological cancers. Front Oncol 2012; 2:168. [PMID: 23316473 PMCID: PMC3540928 DOI: 10.3389/fonc.2012.00168] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Accepted: 10/28/2012] [Indexed: 01/15/2023] Open
Abstract
There is substantial evidence for a role in cancer of the bioactive lipid sphingosine 1-phosphate (S1P), the enzyme sphingosine kinase 1 (that catalyses S1P formation) and S1P-specific G protein-coupled receptors. This perspective highlights recent findings demonstrating that sphingosine kinase 1 and S1P receptors are new important biomarkers for detection of early cancer and progression to aggressive cancer. The impact of the sub-cellular distribution of S1P metabolizing enzymes and S1P receptors and their spatial functional interaction with oncogenes is considered with respect to prognostic outcome. These findings suggest that S1P, in addition to being a biomarker of clinical prognosis, might also be a new therapeutic target for intervention in cancer.
Collapse
Affiliation(s)
- Susan Pyne
- Cell Biology Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde Glasgow, UK
| | | | | | | |
Collapse
|
44
|
Orr Gandy KA, Obeid LM. Targeting the sphingosine kinase/sphingosine 1-phosphate pathway in disease: review of sphingosine kinase inhibitors. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:157-66. [PMID: 22801037 DOI: 10.1016/j.bbalip.2012.07.002] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 07/05/2012] [Accepted: 07/06/2012] [Indexed: 12/01/2022]
Abstract
Sphingosine 1-phosphate (S1P) is an important bioactive sphingolipid metabolite that has been implicated in numerous physiological and cellular processes. Not only does S1P play a structural role in cells by defining the components of the plasma membrane, but in the last 20 years it has been implicated in various significant cell signaling pathways and physiological processes: for example, cell migration, survival and proliferation, cellular architecture, cell-cell contacts and adhesions, vascular development, atherosclerosis, acute pulmonary injury and respiratory distress, inflammation and immunity, and tumorogenesis and metastasis [1,2]. Given the wide variety of cellular and physiological processes in which S1P is involved, it is immediately obvious why the mechanisms governing S1P synthesis and degradation, and the manner in which these processes are regulated, are necessary to understand. In gaining more knowledge about regulation of the sphingosine kinase (SK)/S1P pathway, many potential therapeutic targets may be revealed. This review explores the roles of the SK/S1P pathway in disease, summarizes available SK enzyme inhibitors and examines their potential as therapeutic agents. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
Affiliation(s)
- K Alexa Orr Gandy
- The Department of Molecular and Cellular Biology, The Medical University of South Carolina, Charleston, SC 29425, USA
| | | |
Collapse
|