1
|
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
|
2
|
Alizadeh J, Kavoosi M, Singh N, Lorzadeh S, Ravandi A, Kidane B, Ahmed N, Mraiche F, Mowat MR, Ghavami S. Regulation of Autophagy via Carbohydrate and Lipid Metabolism in Cancer. Cancers (Basel) 2023; 15:2195. [PMID: 37190124 PMCID: PMC10136996 DOI: 10.3390/cancers15082195] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 03/21/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
Metabolic changes are an important component of tumor cell progression. Tumor cells adapt to environmental stresses via changes to carbohydrate and lipid metabolism. Autophagy, a physiological process in mammalian cells that digests damaged organelles and misfolded proteins via lysosomal degradation, is closely associated with metabolism in mammalian cells, acting as a meter of cellular ATP levels. In this review, we discuss the changes in glycolytic and lipid biosynthetic pathways in mammalian cells and their impact on carcinogenesis via the autophagy pathway. In addition, we discuss the impact of these metabolic pathways on autophagy in lung cancer.
Collapse
Affiliation(s)
- Javad Alizadeh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Mahboubeh Kavoosi
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Navjit Singh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
| | - Amir Ravandi
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Institute of Cardiovascular Sciences, Albrechtsen Research Centre, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada;
| | - Biniam Kidane
- Section of Thoracic Surgery, Department of Surgery, Health Sciences Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 6C5, Canada;
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
| | - Naseer Ahmed
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
- Department of Radiology, Section of Radiation Oncology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Fatima Mraiche
- College of Pharmacy, QU Health, Qatar University, Doha 2713, Qatar;
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada
| | - Michael R. Mowat
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada; (N.A.)
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, College of Medicine, University of Manitoba, Winnipeg, MB R3E 0V9, Canada (S.L.)
- Research Institute of Oncology and Hematology, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, Academia of Silesia, 41-800 Zabrze, Poland
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada
| |
Collapse
|
3
|
Developing New Treatment Options for Castration-Resistant Prostate Cancer and Recurrent Disease. Biomedicines 2022; 10:biomedicines10081872. [PMID: 36009418 PMCID: PMC9405166 DOI: 10.3390/biomedicines10081872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/29/2022] [Indexed: 11/16/2022] Open
Abstract
Prostate cancer (PCa) is a major diagnosed cancer among men globally, and about 20% of patients develop metastatic prostate cancer (mPCa) in the initial diagnosis. PCa is a typical androgen-dependent disease; thus, hormonal therapy is commonly used as a standard care for mPCa by inhibiting androgen receptor (AR) activities, or androgen metabolism. Inevitably, almost all PCa will acquire resistance and become castration-resistant PCa (CRPC) that is associated with AR gene mutations or amplification, the presence of AR variants, loss of AR expression toward neuroendocrine phenotype, or other hormonal receptors. Treating CRPC poses a great challenge to clinicians. Research efforts in the last decade have come up with several new anti-androgen agents to prolong overall survival of CRPC patients. In addition, many potential targeting agents have been at the stage of being able to translate many preclinical discoveries into clinical practices. At this juncture, it is important to highlight the emerging strategies including small-molecule inhibitors to AR variants, DNA repair enzymes, cell survival pathway, neuroendocrine differentiation pathway, radiotherapy, CRPC-specific theranostics and immune therapy that are underway or have recently been completed.
Collapse
|
4
|
Tanaka Y, Okabe S, Ohyashiki K, Gotoh A. Potential of a sphingosine 1‑phosphate receptor antagonist and sphingosine kinase inhibitors as targets for multiple myeloma treatment. Oncol Lett 2022; 23:111. [PMID: 35251342 PMCID: PMC8850960 DOI: 10.3892/ol.2022.13231] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 01/18/2022] [Indexed: 11/12/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive lipid involved in cancer progression through its binding to S1P receptors (S1PRs). However, the association between multiple myeloma (MM) and S1P is unclear. The current study aimed to investigate the potential anti-cancer effects of fingolimod and sphingosine kinase (SK) inhibitors in myeloma cells and the effects of S1P-induced chemoresistance and neovascularization on MM cell proliferation. MM cell lines were treated with the S1PR1 antagonist fingolimod and the SK inhibitors ABC294640 and SK1-I, after which cell proliferation was measured. Protein expression was also assessed under each condition using immunoblotting. Serum S1P levels in patients with MM, monoclonal gammopathy of undetermined significance and healthy volunteers were assessed. Human umbilical vessel cells (HUVECs) were co-cultured with anti-S1P agents to assess the effect on cell migration. All treatments suppressed myeloma cell proliferation and caspase-3-mediated apoptosis by suppressing S1P activity. These findings suggest that S1P activation is associated with proliferation and survival for MM cells. S1P attenuated the proteosome inhibitor (PI) effect, while the anti-S1P agents recovered the effect. In addition, S1P promoted the migration and proliferation of HUVECs, whereas the S1P inhibitors reduced the influence of S1P. This study highlights the therapeutic potential of anti-S1P agents for MM treatment. Inhibition of S1P function may overcome resistance to PI developed by myeloma cells and inhibit the changes to the bone marrow microenvironment via neovascularization.
Collapse
Affiliation(s)
- Yuko Tanaka
- Division of Hematology, Tokyo Medical University, Tokyo 160‑0023, Japan
| | - Seiichi Okabe
- Division of Hematology, Tokyo Medical University, Tokyo 160‑0023, Japan
| | - Kazuma Ohyashiki
- Division of Hematology, Tokyo Medical University, Tokyo 160‑0023, Japan
| | - Akihiko Gotoh
- Division of Hematology, Tokyo Medical University, Tokyo 160‑0023, Japan
| |
Collapse
|
5
|
Bataller M, Sánchez-García A, Garcia-Mayea Y, Mir C, Rodriguez I, LLeonart ME. The Role of Sphingolipids Metabolism in Cancer Drug Resistance. Front Oncol 2022; 11:807636. [PMID: 35004331 PMCID: PMC8733468 DOI: 10.3389/fonc.2021.807636] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 12/07/2021] [Indexed: 12/25/2022] Open
Abstract
Drug resistance continues to be one of the major challenges to cure cancer. As research in this field evolves, it has been proposed that numerous bioactive molecules might be involved in the resistance of cancer cells to certain chemotherapeutics. One well-known group of lipids that play a major role in drug resistance are the sphingolipids. Sphingolipids are essential components of the lipid raft domains of the plasma membrane and this structural function is important for apoptosis and/or cell proliferation. Dysregulation of sphingolipids, including ceramide, sphingomyelin or sphingosine 1-phosphate, has been linked to drug resistance in different types of cancer, including breast, melanoma or colon cancer. Sphingolipid metabolism is complex, involving several lipid catabolism with the participation of key enzymes such as glucosylceramide synthase (GCS) and sphingosine kinase 1 (SPHK1). With an overview of the latest available data on this topic and its implications in cancer therapy, this review focuses on the main enzymes implicated in sphingolipids metabolism and their intermediate metabolites involved in cancer drug resistance.
Collapse
Affiliation(s)
- Marina Bataller
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Almudena Sánchez-García
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Yoelsis Garcia-Mayea
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Cristina Mir
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain
| | - Isabel Rodriguez
- Assistant Director of Nursing, Nursing Management Service Hospital Vall d'Hebron, Barcelona, Spain
| | - Matilde Esther LLeonart
- Biomedical Research in Cancer Stem Cells Group, Vall d´Hebron Research Institute (VHIR), Barcelona, Spain.,Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Madrid, Spain
| |
Collapse
|
6
|
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
|
7
|
Motono N, Ueda Y, Shimasaki M, Iwai S, Iijima Y, Usuda K, Uramoto H. Prognostic Impact of Sphingosine Kinase 1 in Nonsmall Cell Lung Cancer. CLINICAL PATHOLOGY 2021; 14:2632010X20988531. [PMID: 33623898 PMCID: PMC7879003 DOI: 10.1177/2632010x20988531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/25/2020] [Indexed: 12/02/2022]
Abstract
Bioactive sphingolipid is clearly relevant to lung physiology. The relationship of the bioactive sphingolipid pathway to pulmonary disease has been studied in cellular, tissue, and animal model, including lung cancer models. The samples of 53 patients diagnosed with nonsmall cell lung carcinoma (NSCLC) between June 2009 and May 2014 at our hospital were analyzed. Immunohistochemical (IHC) analysis was performed. The degree of immunostaining was reviewed and scored. Using this method of assessment, we evaluated the IHC score of sphingosine kinase 1 (SPHK1), vimentin, E-cadherin, and Ki-67. Both invasive adenocarcinoma cell and squamous cell carcinoma cell were well stained by SPHK1, and fibroblasts were also well stained by SPHK1. Although the IHC score of SPHK1 was not significantly differed between invasive adenocarcinoma and squamous cell carcinoma, the IHC scores of fibroblast, vimentin, and Ki-67 were higher in squamous cell carcinoma than invasive adenocarcinoma. Correlation among IHC scores in each of invasive adenocarcinoma and squamous cell carcinoma was performed. SPHK1 had positive correlation with both fibroblast and Ki-67, and fibroblast and Ki-67 had also positive correlation in invasive adenocarcinoma. On the contrary, SPHK1 had no significant correlation with fibroblast, and had negative correlation with Ki-67 in squamous cell carcinoma. Although there was not significant prognostic difference in SPHK1 score (P = .09), IHC score high group tended to be worse on relapse-free survival. SPHK1 might be prognostic factor in lung-invasive adenocarcinoma and novel target for drug against lung-invasive adenocarcinoma.
Collapse
Affiliation(s)
- Nozomu Motono
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Yoshimichi Ueda
- Department of Pathology II, Kanazawa Medical University, Uchinada, Japan
| | - Miyako Shimasaki
- Department of Pathology II, Kanazawa Medical University, Uchinada, Japan
| | - Shun Iwai
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Yoshihito Iijima
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Katsuo Usuda
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| | - Hidetaka Uramoto
- Department of Thoracic Surgery, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
8
|
Roy S, Mohammad T, Gupta P, Dahiya R, Parveen S, Luqman S, Hasan GM, Hassan MI. Discovery of Harmaline as a Potent Inhibitor of Sphingosine Kinase-1: A Chemopreventive Role in Lung Cancer. ACS OMEGA 2020; 5:21550-21560. [PMID: 32905276 PMCID: PMC7469376 DOI: 10.1021/acsomega.0c02165] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 08/10/2020] [Indexed: 06/11/2023]
Abstract
The sphingosine kinase-1/sphingosine-1-phosphate pathway is linked with the cancer progression and survival of the chemotherapy-challenged cells. Sphingosine kinase-1 (SphK1) has emerged as an attractive drug target, but their inhibitors from natural sources are limited. In this study, we have chosen harmaline, one of the β-carboline alkaloids, and report its mechanism of binding to SphK1 and subsequent inhibition. Molecular docking combined with fluorescence binding studies revealed that harmaline binds to the substrate-binding pocket of SphK1 with an appreciable binding affinity and significantly inhibits the kinase activity of SphK1 with an IC50 value in the micromolar range. The cytotoxic effect of harmaline on non-small-cell lung cancer cells by MTT assay was found to be higher for H1299 compared to A549. Harmaline induces apoptosis in non-small-cell lung carcinoma cells (H1299 and A549), possibly via the intrinsic pathway. Our findings suggest that harmaline could be implicated as a scaffold for designing potent anticancer molecules with SphK1 inhibitory potential.
Collapse
Affiliation(s)
- Sonam Roy
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Taj Mohammad
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Preeti Gupta
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Rashmi Dahiya
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Shahnaz Parveen
- Molecular
Bioprospection Department, CSIR-Central
Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Suaib Luqman
- Molecular
Bioprospection Department, CSIR-Central
Institute of Medicinal and Aromatic Plants, Lucknow 226015, Uttar Pradesh, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, Uttar Pradesh, India
| | - Gulam Mustafa Hasan
- Department
of Biochemistry, College of Medicine, Prince
Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Kingdom of Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Centre
for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| |
Collapse
|
9
|
Shi W, Zhang S, Ma D, Yan D, Zhang G, Cao Y, Wang Z, Wu J, Jiang C. Targeting SphK2 Reverses Acquired Resistance of Regorafenib in Hepatocellular Carcinoma. Front Oncol 2020; 10:694. [PMID: 32670862 PMCID: PMC7327090 DOI: 10.3389/fonc.2020.00694] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 04/14/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Regorafenib is a second-line therapy drug used for advanced hepatocellular carcinoma (HCC). Unfortunately, the survival benefit of the patients receiving this treatment is modest, which may be attributed to drug resistance. In the present study, sphingosine kinase 2 (SphK2) was targeted to reverse regorafenib resistance in HCC. Methods: The functions of SphK2 and sphingosine-1-phosphate (S1P), the catalytic product of SphK2 in regorafenib resistance of HCC cells, were evaluated by cell counting kit-8 assay, colony formation, cell cycle evaluation, and annexin V–fluorescein isothiocyanate/propidium iodide double-staining assay. The antitumor activity of combined treatment of regorafenib and the SphK2-specific inhibitor ABC294640 was examined in HCC cells in vitro and xenograft model in vivo. The molecular mechanisms of SphK2/S1P-mediating regorafenib resistance were investigated using cell line establishment and Western blot analysis. Results: Well-developed regorafenib-resistant HCC cells indicated high expression levels of SphK2. The sensitivity to regorafenib of regorafenib-resistant HCC cells was restored following SphK2 knockdown or pharmacological inhibition by ABC294640. In addition, ectopic expression of SphK2 and exogenous addition of S1P decreased the sensitivity of HCC cells to regorafenib. Furthermore, the combination treatment with ABC294640 sensitized resistant tumor to regorafenib in xenograft model of HCC. The phosphorylation levels of nuclear factor κB (NF-κB), as well as those of signal transducer and activator of transcription 3 (STAT3), were positively associated with SphK2 and S1P. Conclusions: SphK2/S1P mediates regorafenib resistance of HCC through NF-κB and STAT3 activation. Targeting SphK2 by ABC294640 potently reduces regorafenib resistance of HCC cells both in vitro and in vivo. The combination of ABC294640 and regorafenib could be developed as a novel potential treatment strategy for advanced HCC.
Collapse
Affiliation(s)
- Weiwei Shi
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Shan Zhang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Ding Ma
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Dongliang Yan
- Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Guang Zhang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.,Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Yin Cao
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.,Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Zhongxia Wang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.,Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| | - Junhua Wu
- Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Chunping Jiang
- Department of Hepatobiliary Surgery, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.,Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.,Department of Hepatobiliary Surgery, Drum Tower Clinical College of Nanjing Medical University, Nanjing, China
| |
Collapse
|
10
|
Chen P, Kuang P, Wang L, Li W, Chen B, Liu Y, Wang H, Zhao S, Ye L, Yu F, He Y, Zhou C. Mechanisms of drugs-resistance in small cell lung cancer: DNA-related, RNA-related, apoptosis-related, drug accumulation and metabolism procedure. Transl Lung Cancer Res 2020; 9:768-786. [PMID: 32676338 PMCID: PMC7354133 DOI: 10.21037/tlcr-19-547] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Small-cell lung cancer (SCLC), the highest malignant cancer amongst different types of lung cancer, has the feature of lower differentiation, rapid growth, and poor survival rate. Despite the dramatically initial sensitivity of SCLC to various types of treatment methods, including chemotherapy, radiotherapy and immunotherapy, the emergence of drugs-resistance is still a grandly clinical challenge. Therefore, in order to improve the prognosis and develop new therapeutic approaches, having a better understanding of the complex mechanisms of resistance in SCLC is of great clinical significance. This review summarized recent advances in understanding of multiple mechanisms which are involved in the resistance during SCLC treatment, including DNA-related process, RNA-related process, apoptosis-related mechanism, and the process of drug accumulation and metabolism.
Collapse
Affiliation(s)
- Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Peng Kuang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Lei Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Wei Li
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Bin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China.,Department of Medical School, Tongji University, Shanghai, China
| | - Sha Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Lingyun Ye
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Feng Yu
- Department of Medical Oncology, The First Affiliated Hospital Of Nanchang University, Nanchang, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Abstract
Sphingosine-1-phosphate (S1P) can regulate several physiological and pathological processes. S1P signaling via its cell surface receptor S1PR1 has been shown to enhance tumorigenesis and stimulate growth, expansion, angiogenesis, metastasis, and survival of cancer cells. S1PR1-mediated tumorigenesis is supported and amplified by activation of downstream effectors including STAT3, interleukin-6, and NF-κB networks. S1PR1 signaling can also trigger various other signaling pathways involved in carcinogenesis including activation of PI3K/AKT, MAPK/ERK1/2, Rac, and PKC/Ca, as well as suppression of cyclic adenosine monophosphate (cAMP). It also induces immunological tolerance in the tumor microenvironment, while the immunosuppressive function of S1PR1 can also lead to the generation of pre-metastatic niches. Some tumor cells upregulate S1PR1 signaling pathways, which leads to drug resistant cancer cells, mainly through activation of STAT3. This signaling pathway is also implicated in some inflammatory conditions leading to the instigation of inflammation-driven cancers. Furthermore, it can also increase survival via induction of anti-apoptotic pathways, for instance, in breast cancer cells. Therefore, S1PR1 and its signaling pathways can be considered as potential anti-tumor therapeutic targets, alone or in combination therapies. Given the oncogenic nature of S1PR1 and its distribution in a variety of cancer cell types along with its targeting advantages over other molecules of this family, S1PR1 should be considered a favorable target in therapeutic approaches to cancer. This review describes the role of S1PR1 in cancer development and progression, specifically addressing breast cancer, glioma, and hematopoietic malignancies. We also discuss the potential use of S1P signaling modulators as therapeutic targets in cancer therapy.
Collapse
|
12
|
Bocheńska K, Gabig-Cimińska M. Unbalanced Sphingolipid Metabolism and Its Implications for the Pathogenesis of Psoriasis. Molecules 2020; 25:E1130. [PMID: 32138315 PMCID: PMC7179243 DOI: 10.3390/molecules25051130] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 02/26/2020] [Accepted: 02/29/2020] [Indexed: 02/06/2023] Open
Abstract
Sphingolipids (SLs), which have structural and biological responsibilities in the human epidermis, are importantly involved in the maintenance of the skin barrier and regulate cellular processes, such as the proliferation, differentiation and apoptosis of keratinocytes (KCs). As many dermatologic diseases, including psoriasis (PsO), intricately characterized by perturbations in these cellular processes, are associated with altered composition and unbalanced metabolism of epidermal SLs, more education to precisely determine the role of SLs, especially in the pathogenesis of skin disorders, is needed. PsO is caused by a complex interplay between skin barrier disruption, immune dysregulation, host genetics and environmental triggers. The contribution of particular cellular compartments and organelles in SL metabolism, a process related to dysfunction of lysosomes in PsO, seems to have a significant impact on lysosomal signalling linked to a modulation of the immune-mediated inflammation accompanying this dermatosis and is not fully understood. It is also worth noting that a prominent skin disorder, such as PsO, has diminished levels of the main epidermal SL ceramide (Cer), reflecting altered SL metabolism, that may contribute not only to pathogenesis but also to disease severity and/or progression. This review provides a brief synopsis of the implications of SLs in PsO, aims to elucidate the roles of these molecules in complex cellular processes deregulated in diseased skin tissue and highlights the need for increased research in the field. The significance of SLs as structural and signalling molecules and their actions in inflammation, in which these components are factors responsible for vascular endothelium abnormalities in the development of PsO, are discussed.
Collapse
Affiliation(s)
- Katarzyna Bocheńska
- Department of Medical Biology and Genetics, University of Gdańsk, Wita Stwosza 59, 80–308 Gdańsk, Poland;
| | - Magdalena Gabig-Cimińska
- Department of Medical Biology and Genetics, University of Gdańsk, Wita Stwosza 59, 80–308 Gdańsk, Poland;
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Laboratory of Molecular Biology, Kładki 24, 80–822 Gdańsk, Poland
| |
Collapse
|
13
|
Kumar S, Kushwaha PP, Gupta S. Emerging targets in cancer drug resistance. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2019; 2:161-177. [PMID: 35582722 PMCID: PMC8992633 DOI: 10.20517/cdr.2018.27] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 03/08/2019] [Accepted: 03/14/2019] [Indexed: 02/05/2023]
Abstract
Drug resistance is a complex phenomenon that frequently develops as a failure to chemotherapy during cancer treatment. Malignant cells increasingly generate resistance to various chemotherapeutic drugs through distinct mechanisms and pathways. Understanding the molecular mechanisms involved in drug resistance remains an important area of research for identification of precise targets and drug discovery to improve therapeutic outcomes. This review highlights the role of some recent emerging targets and pathways which play critical role in driving drug resistance.
Collapse
Affiliation(s)
- Shashank Kumar
- School of Basic and Applied Sciences, Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda 151001, India
| | - Prem Prakash Kushwaha
- School of Basic and Applied Sciences, Department of Biochemistry and Microbial Sciences, Central University of Punjab, Bathinda 151001, India
| | - Sanjay Gupta
- Department of Urology, Case Western Reserve University, Cleveland, Ohio 44106, USA
- The Urology Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio 44106, USA
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Divison of General Medical Sciences, Case Comprehensive Cancer Center, Cleveland, Ohio 44106, USA
- Department of Urology, Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
14
|
Shaw J, Costa-Pinheiro P, Patterson L, Drews K, Spiegel S, Kester M. Novel Sphingolipid-Based Cancer Therapeutics in the Personalized Medicine Era. Adv Cancer Res 2018; 140:327-366. [PMID: 30060815 DOI: 10.1016/bs.acr.2018.04.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sphingolipids are bioactive lipids that participate in a wide variety of biological mechanisms, including cell death and proliferation. The myriad of pro-death and pro-survival cellular pathways involving sphingolipids provide a plethora of opportunities for dysregulation in cancers. In recent years, modulation of these sphingolipid metabolic pathways has been in the forefront of drug discovery for cancer therapeutics. About two decades ago, researchers first showed that standard of care treatments, e.g., chemotherapeutics and radiation, modulate sphingolipid metabolism to increase endogenous ceramides, which kill cancer cells. Strikingly, resistance to these treatments has also been linked to altered sphingolipid metabolism, favoring lipid species that ultimately lead to cell survival. To this end, many inhibitors of sphingolipid metabolism have been developed to further define not only our understanding of these pathways but also to potentially serve as therapeutic interventions. Therefore, understanding how to better use these new drugs that target sphingolipid metabolism, either alone or in combination with current cancer treatments, holds great potential for cancer control. While sphingolipids in cancer have been reviewed previously (Hannun & Obeid, 2018; Lee & Kolesnick, 2017; Morad & Cabot, 2013; Newton, Lima, Maceyka, & Spiegel, 2015; Ogretmen, 2018; Ryland, Fox, Liu, Loughran, & Kester, 2011) in this chapter, we present a comprehensive review on how standard of care therapeutics affects sphingolipid metabolism, the current landscape of sphingolipid inhibitors, and the clinical utility of sphingolipid-based cancer therapeutics.
Collapse
Affiliation(s)
- Jeremy Shaw
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Pedro Costa-Pinheiro
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Logan Patterson
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Kelly Drews
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville, VA, United States; University of Virginia Cancer Center, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
15
|
Stahelin RV. The unmasking of the lipid binding face of sphingosine kinase 1. J Lipid Res 2018; 59:401-403. [DOI: 10.1194/jlr.c083907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
16
|
White C, Alshaker H, Cooper C, Winkler M, Pchejetski D. The emerging role of FTY720 (Fingolimod) in cancer treatment. Oncotarget 2018; 7:23106-27. [PMID: 27036015 PMCID: PMC5029614 DOI: 10.18632/oncotarget.7145] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 02/07/2023] Open
Abstract
FTY720 (Fingolimod) is a clinically approved immunomodulating therapy for multiple sclerosis that sequesters T-cells to lymph nodes through functional antagonism of sphingosine-1-phosphate 1 receptor. FTY720 also demonstrates a proven efficacy in multiple in vitro and in vivo cancer models, suggesting a potential therapeutic role in cancer patients. A potential anticancer mechanism of FTY720 is through the inhibition of sphingosine kinase 1, a proto-oncogene with in vitro and clinical cancer association. In addition, FTY720's anticancer properties may be attributable to actions on several other molecular targets. This study focuses on reviewing the emerging evidence regarding the anticancer properties and molecular targets of FTY720. While the clinical transition of FTY720 is currently limited by its immune suppression effects, studies aiming at FTY720 delivery and release together with identifying its key synergetic combinations and relevant patient subsets may lead to its rapid introduction into the clinic.
Collapse
Affiliation(s)
| | - Heba Alshaker
- Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan.,School of Medicine, University of East Anglia, Norwich, UK
| | - Colin Cooper
- School of Medicine, University of East Anglia, Norwich, UK
| | - Matthias Winkler
- Department of Surgery and Cancer, Imperial College London, London, UK
| | | |
Collapse
|
17
|
Pulkoski-Gross MJ, Jenkins ML, Truman JP, Salama MF, Clarke CJ, Burke JE, Hannun YA, Obeid LM. An intrinsic lipid-binding interface controls sphingosine kinase 1 function. J Lipid Res 2018; 59:462-474. [PMID: 29326159 DOI: 10.1194/jlr.m081307] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 01/04/2018] [Indexed: 12/15/2022] Open
Abstract
Sphingosine kinase 1 (SK1) is required for production of sphingosine-1-phosphate (S1P) and thereby regulates many cellular processes, including cellular growth, immune cell trafficking, and inflammation. To produce S1P, SK1 must access sphingosine directly from membranes. However, the molecular mechanisms underlying SK1's direct membrane interactions remain unclear. We used hydrogen/deuterium exchange MS to study interactions of SK1 with membrane vesicles. Using the CRISPR/Cas9 technique to generate HCT116 cells lacking SK1, we explored the effects of membrane interface disruption and the function of the SK1 interaction site. Disrupting the interface resulted in reduced membrane association and decreased cellular SK1 activity. Moreover, SK1-dependent signaling, including cell invasion and endocytosis, was abolished upon mutation of the membrane-binding interface. Of note, we identified a positively charged motif on SK1 that is responsible for electrostatic interactions with membranes. Furthermore, we demonstrated that SK1 uses a single contiguous interface, consisting of an electrostatic site and a hydrophobic site, to interact with membrane-associated anionic phospholipids. Altogether, these results define a composite domain in SK1 that regulates its intrinsic ability to bind membranes and indicate that this binding is critical for proper SK1 function. This work will allow for a new line of thinking for targeting SK1 in disease.
Collapse
Affiliation(s)
- Michael J Pulkoski-Gross
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY 11790.,Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790
| | - Meredith L Jenkins
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8N 1A1, Canada
| | - Jean-Philip Truman
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790
| | - Mohamed F Salama
- Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35511, Egypt
| | - Christopher J Clarke
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790
| | - John E Burke
- Department of Biochemistry and Microbiology, University of Victoria, Victoria, British Columbia V8N 1A1, Canada
| | - Yusuf A Hannun
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790
| | - Lina M Obeid
- Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY 11790 .,Northport Veterans Affairs Medical Center, Northport, NY 11768
| |
Collapse
|
18
|
Carroll BL, Bonica J, Shamseddine AA, Hannun YA, Obeid LM. A role for caspase-2 in sphingosine kinase 1 proteolysis in response to doxorubicin in breast cancer cells - implications for the CHK1-suppressed pathway. FEBS Open Bio 2017; 8:27-40. [PMID: 29321954 PMCID: PMC5757171 DOI: 10.1002/2211-5463.12344] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/29/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Sphingosine kinase 1 (SK1) is a lipid kinase whose activity produces sphingosine 1‐phosphate, a prosurvival lipid associated with proliferation, angiogenesis, and invasion. SK1 overexpression has been observed in numerous cancers. Recent studies have demonstrated that SK1 proteolysis occurs downstream of the tumor suppressor p53 in response to several DNA‐damaging agents. Moreover, loss of SK1 in p53‐knockout mice resulted in complete protection from thymic lymphoma, providing evidence that regulation of SK1 constitutes a major tumor suppressor function of p53. Given this profound phenotype, this study aimed to investigate the mechanism by which wild‐type p53 regulates proteolysis of SK1 in response to the DNA‐damaging agent doxorubicin in breast cancer cells. We find that p53‐mediated activation of caspase‐2 was required for SK1 proteolysis and that caspase‐2 activity significantly alters the levels of endogenous sphingolipids. As p53 is mutated in 50% of all cancers, we extended our studies to investigate whether SK1 is deregulated in the context of triple‐negative breast cancer cells (TNBC) harboring a mutation in p53. Indeed, caspase‐2 was not activated in these cells and SK1 was not degraded. Moreover, caspase‐2 activation was recently shown to be downstream of the CHK1‐suppressed pathway in p53‐mutant cells, whereby inhibition of the cell cycle kinase CHK1 leads to caspase‐2 activation and apoptosis. Indeed, knockdown and inhibition of CHK1 led to the loss of SK1 in p53‐mutant TNBC cells, providing evidence that SK1 may be the first identified effector of the CHK1‐suppressed pathway.
Collapse
Affiliation(s)
- Brittany L Carroll
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Joseph Bonica
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Achraf A Shamseddine
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Yusuf A Hannun
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA
| | - Lina M Obeid
- Department of Medicine Stony Brook Cancer Center Health Sciences Center Stony Brook University NY USA.,Northport Veterans Affairs Medical Center NY USA
| |
Collapse
|
19
|
Etoposide Upregulates Survival Favoring Sphingosine-1-Phosphate in Etoposide-Resistant Retinoblastoma Cells. Pathol Oncol Res 2017; 25:391-399. [DOI: 10.1007/s12253-017-0360-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 10/27/2017] [Indexed: 10/18/2022]
|
20
|
"Dicing and Splicing" Sphingosine Kinase and Relevance to Cancer. Int J Mol Sci 2017; 18:ijms18091891. [PMID: 28869494 PMCID: PMC5618540 DOI: 10.3390/ijms18091891] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 08/29/2017] [Accepted: 08/29/2017] [Indexed: 02/06/2023] Open
Abstract
Sphingosine kinase (SphK) is a lipid enzyme that maintains cellular lipid homeostasis. Two SphK isozymes, SphK1 and SphK2, are expressed from different chromosomes and several variant isoforms are expressed from each of the isozymes, allowing for the multi-faceted biological diversity of SphK activity. Historically, SphK1 is mainly associated with oncogenicity, however in reality, both SphK1 and SphK2 isozymes possess oncogenic properties and are recognized therapeutic targets. The absence of mutations of SphK in various cancer types has led to the theory that cancer cells develop a dependency on SphK signaling (hyper-SphK signaling) or “non-oncogenic addiction”. Here we discuss additional theories of SphK cellular mislocation and aberrant “dicing and splicing” as contributors to cancer cell biology and as key determinants of the success or failure of SphK/S1P (sphingosine 1 phosphate) based therapeutics.
Collapse
|
21
|
Schissler AG, Li Q, Chen JL, Kenost C, Achour I, Billheimer DD, Li H, Piegorsch WW, Lussier YA. Analysis of aggregated cell-cell statistical distances within pathways unveils therapeutic-resistance mechanisms in circulating tumor cells. Bioinformatics 2017; 32:i80-i89. [PMID: 27307648 PMCID: PMC4908332 DOI: 10.1093/bioinformatics/btw248] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
MOTIVATION As 'omics' biotechnologies accelerate the capability to contrast a myriad of molecular measurements from a single cell, they also exacerbate current analytical limitations for detecting meaningful single-cell dysregulations. Moreover, mRNA expression alone lacks functional interpretation, limiting opportunities for translation of single-cell transcriptomic insights to precision medicine. Lastly, most single-cell RNA-sequencing analytic approaches are not designed to investigate small populations of cells such as circulating tumor cells shed from solid tumors and isolated from patient blood samples. RESULTS In response to these characteristics and limitations in current single-cell RNA-sequencing methodology, we introduce an analytic framework that models transcriptome dynamics through the analysis of aggregated cell-cell statistical distances within biomolecular pathways. Cell-cell statistical distances are calculated from pathway mRNA fold changes between two cells. Within an elaborate case study of circulating tumor cells derived from prostate cancer patients, we develop analytic methods of aggregated distances to identify five differentially expressed pathways associated to therapeutic resistance. Our aggregation analyses perform comparably with Gene Set Enrichment Analysis and better than differentially expressed genes followed by gene set enrichment. However, these methods were not designed to inform on differential pathway expression for a single cell. As such, our framework culminates with the novel aggregation method, cell-centric statistics (CCS). CCS quantifies the effect size and significance of differentially expressed pathways for a single cell of interest. Improved rose plots of differentially expressed pathways in each cell highlight the utility of CCS for therapeutic decision-making. AVAILABILITY AND IMPLEMENTATION http://www.lussierlab.org/publications/CCS/ CONTACT: yves@email.arizona.edu or piegorsch@math.arizona.edu SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- A Grant Schissler
- Center for Biomedical Informatics and Biostatistics (CB2) Graduate Interdisciplinary Program in Statistics Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Qike Li
- Center for Biomedical Informatics and Biostatistics (CB2) Graduate Interdisciplinary Program in Statistics Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - James L Chen
- Division of Bioinformatics, Departments of Biomedical Informatics Division of Medical Oncology, Department of Internal Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Colleen Kenost
- Center for Biomedical Informatics and Biostatistics (CB2) Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Ikbel Achour
- Center for Biomedical Informatics and Biostatistics (CB2) Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - D Dean Billheimer
- Center for Biomedical Informatics and Biostatistics (CB2) Graduate Interdisciplinary Program in Statistics BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Haiquan Li
- Center for Biomedical Informatics and Biostatistics (CB2) Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Walter W Piegorsch
- Graduate Interdisciplinary Program in Statistics BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA
| | - Yves A Lussier
- Center for Biomedical Informatics and Biostatistics (CB2) Graduate Interdisciplinary Program in Statistics Department of Medicine BIO5 Institute, The University of Arizona, Tucson, AZ 85721, USA The University of Arizona Cancer Center, Tucson, AZ 85719, USA Institute for Genomics and Systems Biology, The University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
22
|
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
|
23
|
Zhu YJ, You H, Tan JX, Li F, Qiu Z, Li HZ, Huang HY, Zheng K, Ren GS. Overexpression of sphingosine kinase 1 is predictive of poor prognosis in human breast cancer. Oncol Lett 2017; 14:63-72. [PMID: 28693136 PMCID: PMC5494825 DOI: 10.3892/ol.2017.6134] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2016] [Accepted: 02/22/2017] [Indexed: 01/08/2023] Open
Abstract
Sphingosine kinase 1 (SPHK1) is a bioactive lipid mediator that has been identified as a biomarker in various cancers and is considered to play an important role in tumor progression. In the present study, the expression level of SPHK1 was examined in breast cancer clinical specimens, and its association with patient survival was investigated to clarify the clinical significance of SPHK1 in breast cancer. SPHK1 mRNA expression was increased in breast cancer tissues compared with that in matched adjacent breast tissues in 19 of 32 paired tissue specimens (59.4%). Immunohistochemical analysis of 122 breast cancer cases revealed that the expression levels of SPHK1 were upregulated in 64 tumor tissues (52.5%), and increased expression levels of the protein were significantly associated with the presence of lymph node metastasis (P=0.0016), number of positive lymph nodes (P=0.0268) and presence of distant metastasis (P=0.0097). Increased SPHK1 protein expression was also associated with human epidermal growth factor receptor 2 status (P=0.0100), initial symptoms (P=0.0025) and tumor location (P=0.0457). Patients with increased SPHK1 protein expression had shorter overall survival and disease-free survival times compared with patients with lower SPHK1. Univariate and multivariate analyses indicated that high SPHK1 expression may be a poor prognostic factor. These results indicated that SPHK1 may perform an important role in breast cancer and may be a predictive factor in patients with breast cancer.
Collapse
Affiliation(s)
- Ya-Jing Zhu
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Hua You
- Department of Lymphoma, Head and Neck Oncology, Affiliated Hospital of Academy of Military Medical Sciences, Fengtai, Beijing 100071, P.R. China
| | - Jin-Xiang Tan
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Fan Li
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Zhu Qiu
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Hong-Zhong Li
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Hong-Yan Huang
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Ke Zheng
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Guo-Sheng Ren
- Molecular Oncology and Epigenetics Laboratory, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China.,Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|
24
|
Aoyama Y, Sobue S, Mizutani N, Inoue C, Kawamoto Y, Nishizawa Y, Ichihara M, Kyogashima M, Suzuki M, Nozawa Y, Murate T. Modulation of the sphingolipid rheostat is involved in paclitaxel resistance of the human prostate cancer cell line PC3-PR. Biochem Biophys Res Commun 2017; 486:551-557. [PMID: 28322796 DOI: 10.1016/j.bbrc.2017.03.084] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 03/17/2017] [Indexed: 01/01/2023]
Abstract
Taxoids are anti-cancer drugs frequently used to treat solid tumors, but they are sometimes ineffective and tumors may become resistant to their action. Here, we examined the involvement of sphingolipid metabolic enzymes in paclitaxel (PTX) resistance using a human prostate cancer cell line, PC3, and its PTX-resistant subline, PC3-PR. PTX (20 nM) suppressed cell proliferation and increased various ceramide species in PC3, but not PC3-PR, cells. PC3-PR contained higher S1P levels than did PC3, regardless of PTX treatment. Western blotting revealed that PC3-PR cells expressed higher levels of sphingosine kinase 1 (SPHK1) and glucosylceramide synthase (GCS) but lower levels of acid sphingomyelinase (ASMase) and neutral sphingomyelinase 2 than did PC3 cells. Inhibition of SPHK1 using siRNA or a pharmacological inhibitor decreased S1P levels in PC3-PR cells and inhibited proliferation in the presence or absence of PTX, suggesting that SPHK1 is at least partially responsible for PTX resistance. Similarly, GCS inhibitors (PDMP and PPMP) increased cellular ceramides and suppressed the proliferation of PC3-PR. However, inhibition of proteasome function or histone deacetylase activity increased SMase and ceramide levels and suppressed PC3-PR proliferation. These results suggest that modulation of metabolic enzyme expression and alteration of the sphingolipid rheostat protects cancer cells against PTX.
Collapse
Affiliation(s)
- Yuka Aoyama
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Sayaka Sobue
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Naoki Mizutani
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Chisato Inoue
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Yoshiyuki Kawamoto
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Yuji Nishizawa
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Masatoshi Ichihara
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan
| | - Mamoru Kyogashima
- Division of Molecular Carcinogenesis, Nagoya University Graduate School of Medicine, Nagoya, 466-0064, Japan
| | - Motoshi Suzuki
- Department of Microbiology and Molecular Biology, Nihon Pharmaceutical University, Saitama, 362-0806, Japan
| | | | - Takashi Murate
- College of Life and Health Sciences, Chubu University, Kasugai, 487-8501, Japan.
| |
Collapse
|
25
|
Liang W, Xie Z, Cui W, Guo Y, Xu L, Wu J, Guan H. Comprehensive gene and microRNA expression profiling reveals a role for miRNAs in the oncogenic roles of SphK1 in papillary thyroid cancer. J Cancer Res Clin Oncol 2017; 143:601-611. [PMID: 27942860 DOI: 10.1007/s00432-016-2315-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 11/28/2016] [Indexed: 01/17/2023]
Abstract
PURPOSE The oncogenic roles of sphingosine kinase 1 (SphK1) in various cancers, including thyroid cancer, have been well demonstrated. However, the microRNAs (miRNAs) associated with the oncogenic roles of SphK1 remain largely unknown. METHODS Global gene and miRNA expression in TPC1-Vector and TPC1-SphK1 cells was analyzed using digital gene expression (DGE) analysis and small RNA-seq, respectively. miRNA-mRNA interactions were explored by microT-CDS, and the predicted networks were visualized using CytoScape®. Cell invasion and migration were assessed by performing Transwell invasion and wound-healing assays. Luciferase reporter and immunoblot assays were used to evaluate the targeting of fibronectin 1 (FN1) by miR-144-3p. RESULTS In this study, we found that overexpression of SphK1 differentially regulates the expression of 46 miRNAs and 506 mRNAs in papillary thyroid cancer (PTC) TPC1 cells. Combining bioinformatics predictions of mRNA targets with DGE data on mRNA expression allowed us to identify the mRNA targets of deregulated miRNAs. The direct interaction between miR-144-3p and FN1, which mediates the pro-invasive role of SphK1 in PTC cells, was experimentally validated. CONCLUSIONS Our results demonstrated that SphK1 overexpression drives a regulatory network governing miRNA and mRNA expression in PTC cells. We also demonstrated the roles played by miR-144-3p and FN1 in mediating the oncogenic function of SphK1, which enhanced the understanding of the etiology of PTC.
Collapse
Affiliation(s)
- Weiwei Liang
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Zhiwei Xie
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Weiling Cui
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Yan Guo
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Lijuan Xu
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China
| | - Jueheng Wu
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, 74 Zhongshan Road II, Guangzhou, 510080, Guangdong, China.
| | - Hongyu Guan
- Department of Endocrinology and Diabetes Center, The First Affiliated Hospital of Sun Yat-sen University, 58 Zhongshan Road II, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
26
|
Bernacchioni C, Ghini V, Cencetti F, Japtok L, Donati C, Bruni P, Turano P. NMR metabolomics highlights sphingosine kinase-1 as a new molecular switch in the orchestration of aberrant metabolic phenotype in cancer cells. Mol Oncol 2017; 11:517-533. [PMID: 28258651 PMCID: PMC5527469 DOI: 10.1002/1878-0261.12048] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/17/2017] [Accepted: 02/21/2017] [Indexed: 12/11/2022] Open
Abstract
Strong experimental evidence in animal and cellular models supports a pivotal role of sphingosine kinase‐1 (SK1) in oncogenesis. In many human cancers, SK1 levels are upregulated and these increases are linked to poor prognosis in patients. Here, by employing untargeted NMR‐based metabolomic profiling combined with functional validations, we report the crucial role of SK1 in the metabolic shift known as the Warburg effect in A2780 ovarian cancer cells. Indeed, expression of SK1 induced a high glycolytic rate, characterized by increased levels of lactate along with increased expression of the proton/monocarboxylate symporter MCT1, and decreased oxidative metabolism, associated with the accumulation of intermediates of the tricarboxylic acid cycle and reduction in CO2 production. Additionally, SK1‐expressing cells displayed a significant increase in glucose uptake paralleled by GLUT3 transporter upregulation. The role of SK1 is not limited to the induction of aerobic glycolysis, affecting metabolic pathways that appear to support the biosynthesis of macromolecules. These findings highlight the role of SK1 signaling axis in cancer metabolic reprogramming, pointing out innovative strategies for cancer therapies.
Collapse
Affiliation(s)
- Caterina Bernacchioni
- Department of Biomedical, Clinical and Experimental Sciences, University of Florence, Italy
| | - Veronica Ghini
- CERM and Department of Chemistry, University of Florence, Italy
| | - Francesca Cencetti
- Department of Biomedical, Clinical and Experimental Sciences, University of Florence, Italy
| | - Lukasz Japtok
- Department of Toxicology, Faculty of Mathematics and Natural Science, Institute of Nutritional Science, University of Potsdam, Germany
| | - Chiara Donati
- Department of Biomedical, Clinical and Experimental Sciences, University of Florence, Italy
| | - Paola Bruni
- Department of Biomedical, Clinical and Experimental Sciences, University of Florence, Italy
| | - Paola Turano
- CERM and Department of Chemistry, University of Florence, Italy
| |
Collapse
|
27
|
Vogt D, Stark H. Therapeutic Strategies and Pharmacological Tools Influencing S1P Signaling and Metabolism. Med Res Rev 2016; 37:3-51. [PMID: 27480072 DOI: 10.1002/med.21402] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 06/01/2016] [Accepted: 06/28/2016] [Indexed: 02/06/2023]
Abstract
During the last two decades the study of the sphingolipid anabolic, catabolic, and signaling pathways has attracted enormous interest. Especially the introduction of fingolimod into market as first p.o. therapeutic for the treatment of multiple sclerosis has boosted this effect. Although the complex regulation of sphingosine-1-phosphate (S1P) and other catabolic and anabolic sphingosine-related compounds is not fully understood, the influence on different (patho)physiological states from inflammation to cytotoxicity as well as the availability of versatile pharmacological tools that represent new approaches to study these states are described. Here, we have summarized various aspects concerning the many faces of sphingolipid function modulation by different pharmacological tools up to clinical candidates. Due to the immense heterogeneity of physiological or pharmacological actions and complex cross regulations, it is difficult to predict their role in upcoming therapeutic approaches. Currently, inflammatory, immunological, and/or antitumor aspects are discussed.
Collapse
Affiliation(s)
- Dominik Vogt
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, Max-von-Laue-Straße 9, D-60438, Frankfurt, Germany
| | - Holger Stark
- Institute of Pharmaceutical and Medicinal Chemistry, Heinrich Heine University Düsseldorf, Universitätsstraße 1, D-40225, Düsseldorf, Germany
| |
Collapse
|
28
|
Aurelio L, Scullino CV, Pitman MR, Sexton A, Oliver V, Davies L, Rebello RJ, Furic L, Creek DJ, Pitson SM, Flynn BL. From Sphingosine Kinase to Dihydroceramide Desaturase: A Structure-Activity Relationship (SAR) Study of the Enzyme Inhibitory and Anticancer Activity of 4-((4-(4-Chlorophenyl)thiazol-2-yl)amino)phenol (SKI-II). J Med Chem 2016; 59:965-84. [PMID: 26780304 DOI: 10.1021/acs.jmedchem.5b01439] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The sphingosine kinase (SK) inhibitor, SKI-II, has been employed extensively in biological investigations of the role of SK1 and SK2 in disease and has demonstrated impressive anticancer activity in vitro and in vivo. However, interpretations of results using this pharmacological agent are complicated by several factors: poor SK1/2 selectivity, additional activity as an inducer of SK1-degradation, and off-target effects, including its recently identified capacity to inhibit dihydroceramide desaturase-1 (Des1). In this study, we have delineated the structure-activity relationship (SAR) for these different targets and correlated them to that required for anticancer activity and determined that Des1 inhibition is primarily responsible for the antiproliferative effects of SKI-II and its analogues. In the course of these efforts, a series of novel SK1, SK2, and Des1 inhibitors have been generated, including compounds with significantly greater anticancer activity.
Collapse
Affiliation(s)
- Luigi Aurelio
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Carmen V Scullino
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Melissa R Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Anna Sexton
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Victoria Oliver
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lorena Davies
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Richard J Rebello
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Luc Furic
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
29
|
Yang L, Hu H, Deng Y, Bai Y. [Role of SPHK1 regulates multi-drug resistance of small cell lung cancer
and its clinical significance]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 17:769-77. [PMID: 25404266 PMCID: PMC6000353 DOI: 10.3779/j.issn.1009-3419.2014.11.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
背景与目的 小细胞肺癌约占全部肺癌的15%,化疗是其主要的治疗方法之一,虽然早期对一线化疗方案敏感,但极易出现多药耐药而导致治疗失败。前期基因芯片发现SPHK1与小细胞肺癌的耐药性相关,本研究进一步探讨SPHK1在小细胞肺癌多药耐药中的作用。 方法 首先通过QRT-PCR和Western blot从基因和蛋白水平检测化疗敏感细胞株H69及多药耐药细胞株H69AR中SPHK1的差异表达;转染siRNA下调H69AR细胞中的SPHK1的表达,通过CCK8检测细胞对各种化疗药物(ADM, DDP, VP-16)的敏感性变化,流式细胞仪检测细胞周期及凋亡的变化。同时收集小细胞肺癌化疗前组织和血液标本,将其分为化疗敏感组和耐药组,QRT-PCR检测小细胞肺癌患者血液标本中SPHK1的表达,免疫组化法检测小细胞肺癌患者组织标本中SPHK1的表达,分析SPHK1与小细胞肺癌患者预后相关性。 结果 SPHK1在耐药细胞H69AR中的表达明显高于H69,下调H69AR中SPHK1的表达能够增加细胞对化疗药物的敏感性,促进细胞的凋亡,细胞周期发生G0/G1期阻滞,SPHK1在小细胞肺癌耐药患者中的表达较敏感患者明显增加,SPHK1的表达与患者的性别、年龄无关,与疾病的分期、对化疗的敏感性及生存时间密切相关,差异具有统计学意义(P<0.05)。 结论 SPHK1参与调节小细胞肺癌多药耐药,SPHK1可作为评估小细胞肺癌化疗敏感性及临床预后的潜在靶基因。
Collapse
Affiliation(s)
- Lan Yang
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| | - Honglin Hu
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| | - Ying Deng
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| | - Yifeng Bai
- Department of Oncology, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu 610000, China
| |
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
|
Mizutani N, Inoue M, Omori Y, Ito H, Tamiya-Koizumi K, Takagi A, Kojima T, Nakamura M, Iwaki S, Nakatochi M, Suzuki M, Nozawa Y, Murate T. Increased acid ceramidase expression depends on upregulation of androgen-dependent deubiquitinases, USP2, in a human prostate cancer cell line, LNCaP. J Biochem 2015; 158:309-19. [PMID: 25888580 DOI: 10.1093/jb/mvv039] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 03/25/2015] [Indexed: 01/03/2023] Open
Abstract
Acid ceramidase (ACDase) metabolizes ceramide to sphingosine, leading to sphingosine 1-phosphate production. Reportedly, ACDase has been upregulated in prostate cancer. However, its regulatory mechanism remains unclear. LNCaP (androgen-sensitive prostate cancer cell line) but not PC3 and DU-145, (androgen-unresponsive cell lines) exhibited the highest ACDase protein. Among three cell lines, ASAH1 mRNA level was not correlated with ACDase protein expression, and the 5'-promoter activity did not show androgen dependency, suggesting the post-transcriptional regulation of ACDase in LNCaP cells. Based on these results, LNCaP was analysed further. Casodex, androgen receptor antagonist, and charcoal-stripped FCS (CS-FCS) decreased ACDase protein and activity, whereas dihydrotestosterone in CS-FCS culture increased ACDase protein and enzyme activity. MG132, a proteasome inhibitor, prevented the decrease of ACDase protein when cultured in CS-FCS, suggesting the involvement of ubiquitin/proteasome system. Reportedly, USP2, a deubiquitinase, plays an important role in LNCaP cells. USP2 siRNA decreased ACDase protein, whereas USP2 overexpression increased ACDase protein of LNCaP cells. However, SKP2, an ubiquitin E3 ligase known to be active in prostate cancer, did not affect androgen-dependent ACDase expression in LNCaP cells. Thus, ACDase regulation by androgen in androgen-sensitive LNCaP cells is mainly due to its prolonged protein half-life by androgen-stimulated USP2 expression.
Collapse
Affiliation(s)
- Naoki Mizutani
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Minami Inoue
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Yukari Omori
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Hiromi Ito
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Keiko Tamiya-Koizumi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Akira Takagi
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Tetsuhito Kojima
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673
| | - Mitsuhiro Nakamura
- Department of Drug Information, Gifu Pharmaceutical University, Gifu 501-1196
| | - Soichiro Iwaki
- Department of Molecular and Cellular Pathophysiology and Therapeutics, Graduate School of Pharmaceutical Science, Nagoya City University, Nagoya 467-8603
| | - Masahiro Nakatochi
- Bioinformatics Section, Center for Advanced Medicine and Clinical Research, Nagoya University Hospital, Nagoya 466-8550
| | - Motoshi Suzuki
- Division of Molecular Carcinogenesis, Nagoya University Graduate School of Medicine, Nagoya 466-8560; and
| | - Yoshinori Nozawa
- Department of Food and Health Science, Tokai Gakuin University, Kakamigahara 504-8511, Japan
| | - Takashi Murate
- Department of Pathophysiological Laboratory Science, Nagoya University Graduate School of Medicine, Nagoya 461-8673;
| |
Collapse
|
32
|
Li W, Tian Z, Qin H, Li N, Zhou X, Li J, Ni B, Ruan Z. High expression of sphingosine kinase 1 is associated with poor prognosis in nasopharyngeal carcinoma. Biochem Biophys Res Commun 2015; 460:341-7. [PMID: 25778867 DOI: 10.1016/j.bbrc.2015.03.036] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 03/08/2015] [Indexed: 11/25/2022]
Abstract
It has been reported that sphingosine kinase 1 (SPHK1), an oncogenic enzyme, was involved in the development and progression of a number of human cancers. However, the role of SPHK1 in nasopharyngeal carcinoma (NPC) is largely unknown. The present study aimed to characterize the expression of SPHK1 in human NPC and evaluate its clinical significance. Real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) and Western blot analyses showed that the expression of SPHK1 mRNA and protein in NPC specimens was significantly higher than that in non-tumorous nasopharyngeal mucosa biopsies. Immunohistochemistry (IHC) was conducted to characterize the expression pattern of SPHK1 in 142 archived paraffin-embedded NPC specimens. Statistical analyses revealed that high levels of SPHK1 expression were associated with the clinical stages, locoregional recurrence and distant metastasis of NPC. NPC patients with high levels of SPHK1 expression had shorter survival time, whereas those with lower levels of SPHK1 expression survived longer. Moreover, multivariate analysis suggested that SPHK1 up-regulation was an independent prognostic factor for NPC. Our results suggest for the first time that SPHK1 is involved in the development and progression of NPC, which can be used as a useful prognostic marker for NPC patients and may be an effective target for treating NPC.
Collapse
Affiliation(s)
- Wenhua Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Zhiqiang Tian
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China
| | - Hong Qin
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Ni Li
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Xiaoqing Zhou
- Department of Otolaryngology, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China
| | - Jian Li
- Institute of Hepatopancreatobiliary Surgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China.
| | - Bing Ni
- Institute of Immunology, PLA, Third Military Medical University, Chongqing 400038, PR China.
| | - Zhihua Ruan
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University, Chongqing 400038, PR China.
| |
Collapse
|
33
|
Park JS, Kwon JK, Kim HR, Kim HJ, Kim BS, Jung JY. Farnesol induces apoptosis of DU145 prostate cancer cells through the PI3K/Akt and MAPK pathways. Int J Mol Med 2014; 33:1169-76. [PMID: 24584843 DOI: 10.3892/ijmm.2014.1679] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Accepted: 02/13/2014] [Indexed: 11/06/2022] Open
Abstract
The aim of this study was to investigate the effect of farnesol on the induction of apoptosis in DU145 prostate cancer cells. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay showed that cell proliferation decreased significantly in a dose- and time-dependent manner. 4',6-Diamidino-2-phenylindole staining showed that chromatin condensation in cells treated with 60 µM of farnesol was markedly higher than in the control groups. Farnesol increased the expression of p53, p-c-Jun N-terminal kinase, cleaved-caspase-3, Bax, and cleaved-caspase-9, but decreased the expression of p-phosphatidylinositol-3-kinase (PI3K), p-Akt, p-p38, Bcl-2, and p-extracellular signal-regulated protein kinase, in a dose-dependent manner. The apoptotic cell ratio increased in a dose-dependent manner. The tumor growth inhibitory effect of farnesol was investigated in a mouse model. Compared to the control group, tumor volume decreased significantly in the group administered 50 mg/kg farnesol. Apoptosis was frequently detected in this same group by terminal deoxynucleotidyl transferase dUTP nick-end labeling assay. The results indicated that farnesol induced apoptosis of DU145 prostate cancer cells through the PI3K/Akt and mitogen-activated protein kinase signaling pathways.
Collapse
Affiliation(s)
- Jin Soo Park
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| | - Jung Ki Kwon
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| | - Hye Ri Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| | - Hyeong Jin Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| | - Byeong Soo Kim
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| | - Ji Youn Jung
- Department of Companion and Laboratory Animal Science, Kongju National University, Yesan, Chungnam 340702, Republic of Korea
| |
Collapse
|
34
|
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
|
35
|
Isoforms of protein kinase C involved in phorbol ester-induced sphingosine 1-phosphate receptor 1 phosphorylation and desensitization. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1843:327-34. [DOI: 10.1016/j.bbamcr.2013.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 11/04/2013] [Accepted: 11/07/2013] [Indexed: 12/11/2022]
|
36
|
Chen K, Pan Q, Gao Y, Yang X, Wang S, Peppelenbosch MP, Kong X. DMS triggers apoptosis associated with the inhibition of SPHK1/NF-κB activation and increase in intracellular Ca2+ concentration in human cancer cells. Int J Mol Med 2013; 33:17-24. [PMID: 24173614 PMCID: PMC3868491 DOI: 10.3892/ijmm.2013.1541] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 10/09/2013] [Indexed: 01/04/2023] Open
Abstract
N,N-Dimethyl-D-erythro-sphingosine (DMS) is known to induce cell apoptosis by specifically inhibiting sphingosine kinase 1 (SPHK1) and modulating the activity of cellular ceramide levels. The present study investigated the effects and the mechanism(s) of action of DMS in human lung cancer cells. We found that DMS dose-dependently suppressed cell proliferation and induced cell apoptosis in the human lung cancer cell line, A549. Mechanistically, treatment with DMS suppressed the activation of SPHK1 and nuclear factor-κB (NF-κB) p65, but increased intracellular [Ca2+]i in A549 cells. This study demonstrates that DMS triggers the apoptosis of human lung cancer cells through the modulation of SPHK1, NF-κB and calcium signaling. These molecules may represent targets for anticancer drug design.
Collapse
Affiliation(s)
- Kan Chen
- Bio-X Center, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, Zhejiang 310018, P.R. China
| | | | | | | | | | | | | |
Collapse
|
37
|
Meng XD, Zhou ZS, Qiu JH, Shen WH, Wu Q, Xiao J. Increased SPHK1 expression is associated with poor prognosis in bladder cancer. Tumour Biol 2013; 35:2075-80. [PMID: 24092575 DOI: 10.1007/s13277-013-1275-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 09/25/2013] [Indexed: 12/25/2022] Open
Abstract
Upregulation of sphingosine kinase 1 (SPHK1) protein has been reported to be associated with a poor prognosis in a variety of malignant tumors. However, the role of SPHK1 in bladder cancer (BC) has not been thoroughly elucidated. The purpose of this study was to assess SPHK1 expression and to explore its contribution to BC. Real-time quantitative reverse transcriptase-polymerase chain reaction (qRT-PCR) was conducted to detect SPHK1 mRNA expression in 37 pairs of fresh-frozen BC tissues and corresponding noncancerous tissues. Results showed that SPHK1 mRNA expression level in BC tissues was significantly higher than that in corresponding noncancerous tissues. To investigate the association between SPHK1 protein expression and clinicopathological characteristics of BC, immunohistochemistry (IHC) was performed in 153 archived paraffin-embedded BC samples. Interestingly, high SPHK1 expression was significantly associated with histologic grade (P = 0.045) and tumor stage (P < 0.001) of patients with BC. The Kaplan-Meier survival curve showed that patients with high SPHK1 expression had significantly reduced overall 5-year survival rates (P < 0.001). Multivariate Cox regression analysis further suggested that the increased expression of SPHK1 was an independent poor prognostic factor for this disease. In conclusion, our data offer the convincing evidence for the first time that the increased expression of SPHK1 may be involved in the pathogenesis and progression of BC. SPHK1 might be a potential marker to predict the prognosis in BC.
Collapse
Affiliation(s)
- Xiao-Dong Meng
- Department of Urology, Southwest Hospital, The Third Military Medical University, No. 30, Gaotanyanzheng Street, Shapingba District, Chongqing, 40038, China,
| | | | | | | | | | | |
Collapse
|
38
|
Tonelli F, Alossaimi M, Williamson L, Tate RJ, Watson DG, Chan E, Bittman R, Pyne NJ, Pyne S. The sphingosine kinase inhibitor 2-(p-hyroxyanilino)-4-(p-chlorophenyl)thiazole reduces androgen receptor expression via an oxidative stress-dependent mechanism. Br J Pharmacol 2013; 168:1497-505. [PMID: 23113536 DOI: 10.1111/bph.12035] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2012] [Revised: 10/01/2012] [Accepted: 10/15/2012] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND AND PURPOSE Sphingosine kinase catalyses the formation of sphingosine 1-phosphate and is linked with androgen receptor signalling in prostate cancer cells. Therefore, we investigated the effect of sphingosine kinase inhibitors on androgen receptor expression. EXPERIMENTAL APPROACH Androgen-sensitive LNCaP cells were treated with SKi (2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole), which inhibits sphingosine kinases 1 and 2 activity, and the effect on androgen receptor expression was measured. KEY RESULTS Treatment of cells with SK1 inhibitors reduced the expression of the androgen receptor and prostate-specific antigen, while (R)-FTY720 methyl ether (a sphingosine-kinase-2-selective inhibitor), at a concentration that eliminates sphingosine kinase 2 from cells, had no significant effect on androgen receptor expression. The effect of SKi on androgen receptor expression was independent of the SKi-induced proteasomal degradation of SK1 and was post translational, although androgen receptor mRNA transcript was reduced. Fumonisin B1 (a ceramide synthase inhibitor) also failed to reverse the effect of SKi on androgen receptor expression, thereby excluding a role for ceramide derived from the salvage pathway. The effect of SKi on androgen receptor expression was reversed by N-acetylcysteine, which was used to scavenge reactive oxygen species. CONCLUSION AND IMPLICATIONS Inhibition of sphingosine kinase 1 activity abrogates androgen receptor signalling via an oxidative stress-induced, p53-independent mechanism in prostate cancer cells. Therefore, SK1 inhibitors may offer therapeutic potential in promoting the removal of AR receptors from prostate cancer cells, resulting in an increased efficacy, which is likely to be superior to inhibitors that simply reversibly inhibit AR signalling.
Collapse
Affiliation(s)
- Francesca Tonelli
- Cell Biology and Pharmaceutical Analysis and Metabolomics Groups, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Morad SAF, Levin JC, Tan SF, Fox TE, Feith DJ, Cabot MC. Novel off-target effect of tamoxifen--inhibition of acid ceramidase activity in cancer cells. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1657-64. [PMID: 23939396 DOI: 10.1016/j.bbalip.2013.07.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 07/18/2013] [Accepted: 07/30/2013] [Indexed: 10/26/2022]
Abstract
Acid ceramidase (AC), EC 3.5.1.23, a lysosomal enzyme, catalyzes the hydrolysis of ceramide to constituent sphingoid base, sphingosine, and fatty acid. Because AC regulates the levels of pro-apoptotic ceramide and mitogenic sphingosine-1-phosphate, it is considered an apt target in cancer therapy. The present study reveals, for the first time, that the prominent antiestrogen, tamoxifen, is a pan-effective AC inhibitor in the low, single digit micromolar range, as demonstrated in a wide spectrum of cancer cell types, prostate, pancreatic, colorectal, and breast. Prostate cancer cells were chosen for the detailed investigations. Treatment of intact PC-3 cells with tamoxifen produced time- and dose-dependent inhibition of AC activity. Tamoxifen did not impact cell viability nor did it inhibit AC activity in cell-free assays. In pursuit of mechanism of action, we demonstrate that tamoxifen induced time-, as early as 5min, and dose-dependent, as low as 5μM, increases in lysosomal membrane permeability (LMP), and time- and dose-dependent downregulation of AC protein expression. Assessing various protease inhibitors revealed that a cathepsin B inhibitor blocked tamoxifen-elicited downregulation of AC protein; however, this action failed to restore AC activity unless assayed in a cell-free system at pH4.5. In addition, pretreatment with tamoxifen inhibited PC-3 cell migration. Toremifene, an antiestrogen structurally similar to tamoxifen, was also a potent inhibitor of AC activity. This study reveals a new, off-target action of tamoxifen that may be of benefit to enhance anticancer therapies that either incorporate ceramide or target ceramide metabolism.
Collapse
Affiliation(s)
- Samy A F Morad
- John Wayne Cancer Institute at Saint John's Health Center, Department of Experimental Therapeutics, Santa Monica, CA 90404, USA
| | | | | | | | | | | |
Collapse
|
40
|
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
|
41
|
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
|
42
|
Abstract
Non-surgical therapies for human malignancies must negotiate complex cell signaling pathways to impede cancer cell growth, ideally promoting death of cancer cells while sparing healthy tissue. For most of the past half century, medical approaches for treating cancer have relied primarily on cytotoxic chemotherapeutics that interfere with DNA replication and cell division, susceptibilities of rapidly dividing cancer cells. As a consequence, these therapies exert considerable cell stress, promoting the generation of ceramide through de novo synthesis and recycling of complex glycosphingolipids and sphingomyelin into apoptotic ceramide. Radiotherapy of cancer exerts similar geno- and cytotoxic cell stresses, and generation of ceramide following ionizing radiation therapy is a well-described feature of radiation-induced cell death. Emerging evidence now describes sphingolipids as mediators of death in response to newer targeted therapies, cementing ceramide generation as a common mechanism of cell death in response to cancer therapy. Many studies have now shown that dysregulation of ceramide accumulation-whether by reduced generation or accelerated metabolism-is a common mechanism of resistance to standard cancer therapies. The aims of this chapter will be to discuss described mechanisms of cancer resistance to therapy related to dysregulation of sphingolipid metabolism and to explore clinical and preclinical approaches to interdict sphingolipid metabolism to improve outcomes of standard cancer therapies.
Collapse
|
43
|
Sphingosine kinase 1 plays a role in the upregulation of CD44 expression through extracellular signal-regulated kinase signaling in human colon cancer cells. Anticancer Drugs 2013; 24:473-83. [DOI: 10.1097/cad.0b013e32835f705f] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
44
|
Watson DG, Tonelli F, Osaimi MA, Williamson L, Chan E, Gorshkova I, Berdyshev E, Bittman R, Pyne NJ, Pyne S. The roles of sphingosine kinases 1 and 2 in regulating the Warburg effect in prostate cancer cells. Cell Signal 2013; 25:1011-7. [PMID: 23314175 PMCID: PMC3595369 DOI: 10.1016/j.cellsig.2013.01.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2012] [Accepted: 01/06/2013] [Indexed: 12/30/2022]
Abstract
Two isoforms of sphingosine kinase, SK1 and SK2, catalyze the formation of the bioactive lipid sphingosine 1-phosphate (S1P) in mammalian cells. We have previously shown that treatment of androgen-sensitive LNCaP prostate cancer cells with a non-selective SK isoform inhibitor, 2-(p-hydroxyanilino)-4-(p-chlorophenyl)thiazole (SKi), induces the proteasomal degradation of SK1. This is concomitant with a significant increase in C22:0-ceramide and sphingosine levels and a reduction in S1P levels, resulting in the apoptosis of LNCaP cells. In contrast, we show here that a SK2-selective inhibitor, (R)-FTY720 methyl ether (ROME), increases sphingosine and decreases S1P levels but has no effect on ceramide levels and does not induce apoptosis in LNCaP cells. We also show that several glycolytic metabolites and (R)-S-lactoylglutathione are increased upon treatment of LNCaP cells with SKi, which induces the proteasomal degradation of c-Myc. These changes reflect an indirect antagonism of the Warburg effect. LNCaP cells also respond to SKi by diverting glucose 6-phosphate into the pentose phosphate pathway to provide NADPH, which serves as an antioxidant to counter an oxidative stress response. SKi also promotes the formation of a novel pro-apoptotic molecule called diadenosine 5',5'''-P(1),P(3)-triphosphate (Ap3A), which binds to the tumor suppressor fragile histidine triad protein (FHIT). In contrast, the SK2-selective inhibitor, ROME, induces a reduction in some glycolytic metabolites and does not affect oxidative stress. We conclude that SK1 functions to increase the stability of c-Myc and suppresses Ap3A formation, which might maintain the Warburg effect and cell survival, while SK2 exhibits a non-overlapping function.
Collapse
Affiliation(s)
| | | | | | | | | | - Irina Gorshkova
- The University of Illinois at Chicago Section of Pulmonary, Critical Care, Sleep, and Allergy, 909 S. Wolcott Ave., Chicago, IL 60612, USA
| | - Evgeny Berdyshev
- The University of Illinois at Chicago Section of Pulmonary, Critical Care, Sleep, and Allergy, 909 S. Wolcott Ave., Chicago, IL 60612, USA
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York 11367-1597, USA
| | | | - Susan Pyne
- To whom correspondence should be addressed (Tel: 441415482012; Fax: 441415522562; )
| |
Collapse
|
45
|
Baek DJ, MacRitchie N, Pyne NJ, Pyne S, Bittman R. Synthesis of selective inhibitors of sphingosine kinase 1. Chem Commun (Camb) 2013; 49:2136-8. [PMID: 23388656 DOI: 10.1039/c3cc00181d] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Sphingosine kinase isoform 1 (SK1) inhibitors may serve as therapeutic agents for proliferative diseases, including hypertension. We synthesized a series of sphingosine-based SK1-selective inhibitors, the most potent of which is RB-005 (IC(50) = 3.6 μM), which also induced proteasomal degradation of SK1 in human pulmonary arterial smooth muscle cells.
Collapse
Affiliation(s)
- Dong Jae Baek
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York 11367-1597, USA
| | | | | | | | | |
Collapse
|
46
|
Kim YM, Park TS, Kim SG. The role of sphingolipids in drug metabolism and transport. Expert Opin Drug Metab Toxicol 2013; 9:319-31. [PMID: 23289866 DOI: 10.1517/17425255.2013.748749] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Sphingolipids represent a diverse class of lipid molecules. In addition to their function as membrane structural components, they serve as signaling molecules involved in various biological processes such as cell metabolism, growth, differentiation, stress and inflammatory responses and apoptosis. Sphingolipids may modulate the activity and/or expression of cytochrome P450s (CYPs) and transporters, which suggests that they may affect drug metabolism and excretion. AREAS COVERED In this review, the authors provide an overview of the properties of sphingolipid structures and metabolism. They also describe the effects of sphingolipids on the activity and expression of CYPs and transporters. In addition, the authors discuss the pathologic conditions where the sphingolipid metabolism is dysregulated particularly in association with inflammation and cancer. EXPERT OPINION Sphingolipidomic approaches have become accessible with the aid of advances in analytical technology. Sphingolipid profiles are modified by diseases, genetic disorders or certain drug treatment. The consequent changes in sphingolipid contents may alter the activities of detoxifying enzymes and those associated with cell viability. Since CYPs and transporters play roles in xenobiotics metabolism and excretion, sphingolipidomic information may be of use in understanding drug effect and toxicity.
Collapse
Affiliation(s)
- Young Mi Kim
- Seoul National University, Research Institute of Pharmaceutical Sciences, College of Pharmacy, San 56-1, Sillim-dong, Gwanak-gu, Seoul 151-742, Korea
| | | | | |
Collapse
|
47
|
Abstract
There are several well-established mechanisms involved in radiation-induced cell death in mammalian cell systems. The p53-mediated apoptotic pathway is the most widely recognized mechanism (Lowe et al. Nature 362:847-849, 1993), although apoptosis has long been considered a less relevant mechanism of radiation-induced cell death (Steel, Acta Oncol 40:968-975, 2001; Brown and Wouters, Cancer Res 59:1391-1399, 1999; Olive and Durand, Int J Radiat Biol 71:695-707, 1997). We and others have recently focused instead on the emerging links between radiation, apoptosis, and ceramide and showed that ceramide is a sphingolipid-derived second messenger capable of initiating apoptotic cascades in response to various stress stimuli, including radiation.Ceramide, the backbone of all sphingolipids, is synthesized by a family of ceramide synthases (CerS), each using acyl-CoAs of defined chain length for N-acylation of the sphingoid long-chain base. Six mammalian CerS homologs have been cloned that demonstrated high selectivity towards acyl-CoAs (Lahiri et al. FEBS Lett 581:5289-5294, 2007), and more recently, it was shown that their activity can be modulated by dimer formation (Mesicek et al. Cell Signal 22:1300-1307, 2010; Laviad et al. J Biol Chem 283:5677-5684, 2008).This de novo ceramide synthesis has been observed in irradiated cells through a pathway normally suppressed by ataxia telangiectasia-mutated (ATM) protein, a key component of the cellular response to DNA double-strand breaks (Liao et al. J Biol Chem 274:17908-17917, 1999). ATM is not the sole factor known to affect apoptotic potential by modulating CerS activity. Recent work has also implicated protein kinase Cα (PKCα) as a potential CerS activator (Truman et al. Cancer Biol Ther 8:54-63, 2009).In this review, we summarize involvement of CerS in sphingolipid-mediated apoptosis in irradiated human prostate cancer cells and discuss future directions in this field.
Collapse
Affiliation(s)
- Carla Hajj
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, USA.
| | | |
Collapse
|
48
|
Selvam SP, Ogretmen B. Sphingosine kinase/sphingosine 1-phosphate signaling in cancer therapeutics and drug resistance. Handb Exp Pharmacol 2013:3-27. [PMID: 23563649 DOI: 10.1007/978-3-7091-1511-4_1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this chapter, roles of bioactive sphingolipids, specifically sphingosine kinase 1 (SK1) and 2 (SK2) and their product-sphingosine 1-phosphate (S1P)-will be reviewed with respect to regulation of cancer growth, metastasis, chemotherapeutics, and drug resistance. Sphingolipids are known to be key bioeffector molecules that regulate cancer proliferation, angiogenesis, and cell death. Sphingolipid molecules such as ceramide and S1P have been shown to control cancer cell death and proliferation, respectively. Roles of S1P have been described with respect to their intracellular and extracellular pro-survival and drug resistance functions mostly through S1P receptor (S1PR1-5) engagement. Identification of novel intracellular SK/S1P targets has broadened the existing complex regulatory roles of bioactive sphingolipids in cancer pathogenesis and therapeutics. Thus, deciphering the biochemical and molecular regulation of SK/S1P/S1PR signaling could permit development of novel therapeutic interventions to improve cancer therapy and/or overcome drug resistance.
Collapse
Affiliation(s)
- Shanmugam Panneer Selvam
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | | |
Collapse
|
49
|
Delgado A, Fabriàs G, Casas J, Abad JL. Natural products as platforms for the design of sphingolipid-related anticancer agents. Adv Cancer Res 2013; 117:237-81. [PMID: 23290782 DOI: 10.1016/b978-0-12-394274-6.00008-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Modulation of sphingolipid metabolism is a promising strategy for cancer therapy that has already opened innovative approaches for the development of pharmacological tools and rationally designed new drugs. On the other hand, natural products represent a classical and well-established source of chemical diversity that has guided medicinal chemists on the development of new chemical entities with potential therapeutic use. Based on these premises, the aim of this chapter is to provide the reader with a general overview of some of the most representative families of sphingolipid-related natural products that have been described in the recent literature as lead compounds for the design of new modulators of sphingolipid metabolism. Special emphasis is placed on the structural aspects of natural sphingoids and synthetic analogs that have found application as anticancer agents. In addition, their cellular targets and/or their mode of action are also considered.
Collapse
Affiliation(s)
- Antonio Delgado
- Spanish National Research Council, Consejo Superior de Investigaciones Científicas, Research Unit on Bioactive Molecules, Jordi Girona 18-26, Barcelona, Spain.
| | | | | | | |
Collapse
|
50
|
Abstract
Chemotherapy is frequently used to treat primary or metastatic cancers, but intrinsic or acquired drug resistance limits its efficiency. Sphingolipids are important regulators of various cellular processes including proliferation, apoptosis, differentiation, angiogenesis, stress, and inflammatory responses which are linked to various aspects of cancer, like tumor growth, neoangiogenesis, and response to chemotherapy. Ceramide, the central molecule of sphingolipid metabolism, generally mediates antiproliferative and proapoptotic functions, whereas sphingosine-1-phosphate and other derivatives have opposing effects. Among the variety of enzymes that control ceramide generation, acid or neutral sphingomyelinases and ceramide synthases are important targets to allow killing of cancer cells by chemotherapeutic drugs. On the contrary, glucosylceramide synthase, ceramidase, and sphingosine kinase are other targets driving cancer cell resistance to chemotherapy. This chapter focuses on ceramide-based mechanisms leading to cancer therapy sensitization or resistance which could have some impacts on the development of novel cancer therapeutic strategies.
Collapse
|