1
|
Kirmpaki MA, Saied EM, Schumacher F, Prause K, Kleuser B, Arenz C. Live-Cell Identification of Inhibitors of the Lipid Transfer Protein CERT Using Nanoluciferase Bioluminescence Resonance Energy Transfer (NanoBRET). Angew Chem Int Ed Engl 2024; 63:e202413562. [PMID: 39450584 DOI: 10.1002/anie.202413562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024]
Abstract
A BRET system is described, in which Nanoluciferase was fused to the lipid transfer protein CERT for efficient energy transfer to a Nile red-labeled ceramide, which is either directly bound to CERT or transported to the adjacent Golgi membrane. Bulk formation of sphingomyelin, a major plasma membrane component in mammals, is dependent on CERT-mediated transfer of its predecessor ceramide. CERT is considered a promising drug target but no direct cell-based methods exist to efficiently identify inhibitors. The utility of the method was demonstrated by a library of 140 derivatives of the CERT inhibitor HPA-12. These were obtained in a combinatorial synthesis using solid-phase transacylation. Screening of the library led to six compounds that were picked and confirmed to be superior to HPA-12 in a subsequent dose-response study and also in an orthogonal lipidomics analysis.
Collapse
Affiliation(s)
- Maria-Anna Kirmpaki
- Institute for Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| | - Essa M Saied
- Institute for Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
- Chemistry Department, Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| | - Fabian Schumacher
- Freie Universität Berlin, Institute of Pharmacy, Königin-Luise-Str. 2+4, Berlin, Germany
| | - Kevin Prause
- Institute for Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| | - Burkhard Kleuser
- Freie Universität Berlin, Institute of Pharmacy, Königin-Luise-Str. 2+4, Berlin, Germany
| | - Christoph Arenz
- Institute for Chemistry, Humboldt-Universität zu Berlin, Brook-Taylor-Str. 2, 12489, Berlin, Germany
| |
Collapse
|
2
|
Ung J, Kassai M, Tan SF, Loughran TP, Feith DJ, Cabot MC. The Drug Transporter P-Glycoprotein and Its Impact on Ceramide Metabolism-An Unconventional Ally in Cancer Treatment. Int J Mol Sci 2024; 25:9825. [PMID: 39337312 PMCID: PMC11432138 DOI: 10.3390/ijms25189825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
The tumor-suppressor sphingolipid ceramide is recognized as a key participant in the cytotoxic mechanism of action of many types of chemotherapy drugs, including anthracyclines, Vinca alkaloids, the podophyllotoxin etoposide, taxanes, and the platinum drug oxaliplatin. These drugs can activate de novo synthesis of ceramide or stimulate the production of ceramide via sphingomyelinases to limit cancer cell survival. On the contrary, dysfunctional sphingolipid metabolism, a prominent factor in cancer survival and therapy resistance, blunts the anticancer properties of ceramide-orchestrated cell death pathways, especially apoptosis. Although P-glycoprotein (P-gp) is famous for its role in chemotherapy resistance, herein, we propose alternate interpretations and discuss the capacity of this multidrug transporter as a "ceramide neutralizer", an unwelcome event, highlighting yet another facet of P-gp's versatility in drug resistance. We introduce sphingolipid metabolism and its dysfunctional regulation in cancer, present a summary of factors that contribute to chemotherapy resistance, explain how P-gp "neutralizes" ceramide by hastening its glycosylation, and consider therapeutic applications of the P-gp-ceramide connection in the treatment of cancer.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA;
| | - Miki Kassai
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| | - Su-Fern Tan
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Thomas P. Loughran
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - David J. Feith
- University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA; (S.-F.T.); (D.J.F.)
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Myles C. Cabot
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, The East Carolina Diabetes and Obesity Institute, Greenville, NC 27834, USA;
| |
Collapse
|
3
|
Reddi KK, Chava S, Chabattula SC, Edwards YJK, Singh K, Gupta R. ASAH1 facilitates TNBC by DUSP5 suppression-driven activation of MAP kinase pathway and represents a therapeutic vulnerability. Cell Death Dis 2024; 15:452. [PMID: 38926346 PMCID: PMC11208621 DOI: 10.1038/s41419-024-06831-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that is prone to metastasis and therapy resistance. Owing to its aggressive nature and limited availability of targeted therapies, TNBC is associated with higher mortality as compared to other forms of breast cancer. In order to develop new therapeutic options for TNBC, we characterized the factors involved in TNBC growth and progression. Here, we demonstrate that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in TNBC cells and is regulated via p53 and PI3K-AKT signaling pathways. Genetic knockdown or pharmacological inhibition of ASAH1 suppresses TNBC growth and progression. Mechanistically, ASAH1 inhibition stimulates dual-specificity phosphatase 5 (DUSP5) expression, suppressing the mitogen-activated protein kinase (MAPK) pathway. Furthermore, pharmacological cotargeting of the ASAH1 and MAPK pathways inhibits TNBC growth. Collectively, we unmasked a novel role of ASAH1 in driving TNBC and identified dual targeting of the ASAH1 and MAPK pathways as a potential new therapeutic approach for TNBC treatment.
Collapse
Affiliation(s)
- Kiran Kumar Reddi
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suresh Chava
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Siva Chander Chabattula
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Yvonne J K Edwards
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kamaljeet Singh
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center at The University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
4
|
Muley H, Dowdy T, Zaibaq F, Karadimov G, Li A, Song H, Zhang M, Zhang W, Wong Z, Zhang L, Lita A, Larion M. Targeting IDH1-Mutated Oligodendroglioma with Acid Ceramidase Inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.27.591426. [PMID: 38903086 PMCID: PMC11188094 DOI: 10.1101/2024.04.27.591426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Oligodendroglioma is genetically defined as a tumor harboring isocitrate dehydrogenase 1 or 2 mutations (IDH1 mut /IDH2 mut ) and 1p/19q co-deletions. Previously, we reported that in IDH1 mut gliomas, D-2HG, the product of IDH1 mutant enzyme produces an increase in monounsaturated fatty acid levels that are incorporated into ceramides, tilting the S1P-to-ceramide rheostat toward apoptosis. Herein, we exploited this imbalance to further induce and IDH mut -specific glioma cell death. We report for the first time that the inhibition of acid ceramidase (AC) induces apoptosis and provides a benefit in mice survival in IDH1 mut oligodendroglioma. We demonstrated an IDH1 mut -specific cytotoxicity of SABRAC, an irreversible inhibitor of AC, in patient-derived oligodendroglioma cells. Exploring the mechanism of action of this drug, we found that SABRAC activates both extrinsic and intrinsic apoptosis in an ER stress-independent manner, pointing to a direct action of AC-related ceramides in mitochondria permeability. The activation of apoptosis detected under SABRAC treatment was associated with up to 30-fold increase in some ceramide levels and its derivatives from the salvage pathway. We propose that this novel enzyme, AC, has the potential to increase survival in oligodendroglioma with IDH1 mut and should be considered in the future.
Collapse
|
5
|
Casasampere M, Ung J, Iñáñez A, Dufau C, Tsuboi K, Casas J, Tan SF, Feith DJ, Andrieu-Abadie N, Segui B, Loughran TP, Abad JL, Fabrias G. A fluorogenic substrate for the detection of lipid amidases in intact cells. J Lipid Res 2024; 65:100520. [PMID: 38369184 PMCID: PMC10956054 DOI: 10.1016/j.jlr.2024.100520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/25/2024] [Accepted: 02/12/2024] [Indexed: 02/20/2024] Open
Abstract
Lipid amidases of therapeutic relevance include acid ceramidase (AC), N-acylethanolamine-hydrolyzing acid amidase, and fatty acid amide hydrolase (FAAH). Although fluorogenic substrates have been developed for the three enzymes and high-throughput methods for screening have been reported, a platform for the specific detection of these enzyme activities in intact cells is lacking. In this article, we report on the coumarinic 1-deoxydihydroceramide RBM1-151, a 1-deoxy derivative and vinilog of RBM14-C12, as a novel substrate of amidases. This compound is hydrolyzed by AC (appKm = 7.0 μM; appVmax = 99.3 nM/min), N-acylethanolamine-hydrolyzing acid amidase (appKm = 0.73 μM; appVmax = 0.24 nM/min), and FAAH (appKm = 3.6 μM; appVmax = 7.6 nM/min) but not by other ceramidases. We provide proof of concept that the use of RBM1-151 in combination with reported irreversible inhibitors of AC and FAAH allows the determination in parallel of the three amidase activities in single experiments in intact cells.
Collapse
Affiliation(s)
- Mireia Casasampere
- Department of Biological Chemistry, Research Unit on BioActive Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Johnson Ung
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA; Department of Microbiology, Immunology and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Alejandro Iñáñez
- Department of Biological Chemistry, Research Unit on BioActive Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Carine Dufau
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France; Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Kazuhito Tsuboi
- Department of Pharmacology, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Josefina Casas
- Department of Biological Chemistry, Research Unit on BioActive Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain; CIBEREHD, Madrid, Spain
| | - Su-Fern Tan
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA; University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - David J Feith
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA; University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Nathalie Andrieu-Abadie
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France; Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France
| | - Bruno Segui
- INSERM UMR 1037, Cancer Research Center of Toulouse (CRCT), Toulouse, France; Equipe Labellisée Fondation ARC pour la recherche sur le cancer, Toulouse, France; Université Toulouse III - Paul Sabatier, Toulouse, France
| | - Thomas P Loughran
- Division of Hematology and Oncology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, USA; University of Virginia Cancer Center, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - José Luis Abad
- Department of Biological Chemistry, Research Unit on BioActive Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.
| | - Gemma Fabrias
- Department of Biological Chemistry, Research Unit on BioActive Molecules, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain; CIBEREHD, Madrid, Spain; Spanish National Research Council (CSIC)'s Cancer Hub, Madrid, Spain.
| |
Collapse
|
6
|
Afrin F, Mateen S, Oman J, Lai JCK, Barrott JJ, Pashikanti S. Natural Products and Small Molecules Targeting Cellular Ceramide Metabolism to Enhance Apoptosis in Cancer Cells. Cancers (Basel) 2023; 15:4645. [PMID: 37760612 PMCID: PMC10527029 DOI: 10.3390/cancers15184645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Molecular targeting strategies have been used for years in order to control cancer progression and are often based on targeting various enzymes involved in metabolic pathways. Keeping this in mind, it is essential to determine the role of each enzyme in a particular metabolic pathway. In this review, we provide in-depth information on various enzymes such as ceramidase, sphingosine kinase, sphingomyelin synthase, dihydroceramide desaturase, and ceramide synthase which are associated with various types of cancers. We also discuss the physicochemical properties of well-studied inhibitors with natural product origins and their related structures in terms of these enzymes. Targeting ceramide metabolism exhibited promising mono- and combination therapies at preclinical stages in preventing cancer progression and cemented the significance of sphingolipid metabolism in cancer treatments. Targeting ceramide-metabolizing enzymes will help medicinal chemists design potent and selective small molecules for treating cancer progression at various levels.
Collapse
Affiliation(s)
- Farjana Afrin
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Sameena Mateen
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Jordan Oman
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - James C. K. Lai
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Jared J. Barrott
- Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Srinath Pashikanti
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| |
Collapse
|
7
|
Targeting the Sphingolipid Rheostat in Gliomas. Int J Mol Sci 2022; 23:ijms23169255. [PMID: 36012521 PMCID: PMC9408832 DOI: 10.3390/ijms23169255] [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: 07/30/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/26/2022] Open
Abstract
Gliomas are highly aggressive cancer types that are in urgent need of novel drugs and targeted therapies. Treatment protocols have not improved in over a decade, and glioma patient survival remains among the worst of all cancer types. As a result, cancer metabolism research has served as an innovative approach to identifying novel glioma targets and improving our understanding of brain tumors. Recent research has uncovered a unique metabolic vulnerability in the sphingolipid pathways of gliomas that possess the IDH1 mutation. Sphingolipids are a family of lipid signaling molecules that play a variety of second messenger functions in cellular regulation. The two primary metabolites, sphingosine-1-phosphate (S1P) and ceramide, maintain a rheostat balance and play opposing roles in cell survival and proliferation. Altering the rheostat such that the pro-apoptotic signaling of the ceramides outweighs the pro-survival S1P signaling in glioma cells diminishes the hallmarks of cancer and enhances tumor cell death. Throughout this review, we discuss the sphingolipid pathway and identify the enzymes that can be most effectively targeted to alter the sphingolipid rheostat and enhance apoptosis in gliomas. We discuss each pathway’s steps based on their site of occurrence in the organelles and postulate novel targets that can effectively exploit this vulnerability.
Collapse
|
8
|
DiPasquale M, Deering TG, Desai D, Sharma AK, Amin S, Fox TE, Kester M, Katsaras J, Marquardt D, Heberle FA. Influence of ceramide on lipid domain stability studied with small-angle neutron scattering: The role of acyl chain length and unsaturation. Chem Phys Lipids 2022; 245:105205. [PMID: 35483419 PMCID: PMC9320172 DOI: 10.1016/j.chemphyslip.2022.105205] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/11/2022] [Indexed: 12/13/2022]
Abstract
Ceramides and diacylglycerols are groups of lipids capable of nucleating and stabilizing ordered lipid domains, structures that have been implicated in a range of biological processes. Previous studies have used fluorescence reporter molecules to explore the influence of ceramide acyl chain structure on sphingolipid-rich ordered phases. Here, we use small-angle neutron scattering (SANS) to examine the ability of ceramides and diacylglycerols to promote lipid domain formation in the well-characterized domain-forming mixture DPPC/DOPC/cholesterol. SANS is a powerful, probe-free technique for interrogating membrane heterogeneity, as it is differentially sensitive to hydrogen's stable isotopes protium and deuterium. Specifically, neutron contrast is generated through selective deuteration of lipid species, thus enabling the detection of nanoscopic domains enriched in deuterated saturated lipids dispersed in a matrix of protiated unsaturated lipids. Using large unilamellar vesicles, we found that upon replacing 10 mol% DPPC with either C16:0 or C18:0 ceramide, or 16:0 diacylglycerol (dag), lipid domains persisted to higher temperatures. However, when DPPC was replaced with short chain (C6:0 or C12:0) or very long chain (C24:0) ceramides, or ceramides with unsaturated acyl chains of any length (C6:1(3), C6:1(5), C18:1, and C24:1), as well as C18:1-dag, lipid domains were destabilized, melting at lower temperatures than those in the DPPC/DOPC/cholesterol system. These results show how ceramide acyl chain length and unsaturation influence lipid domains and have implications for how cell membranes might modify their function through the generation of different ceramide species.
Collapse
Affiliation(s)
- Mitchell DiPasquale
- Department of Chemistry and Biochemistry, University of Windsor, Windsor N9B 3P4, ON, Canada
| | - Tye G Deering
- Department of Pharmacology, University of Virginia, Charlottesville 22908, VA, USA
| | - Dhimant Desai
- Department of Pharmacology, Penn State University, University Park 16801, PA, USA
| | - Arun K Sharma
- Department of Pharmacology, Penn State University, University Park 16801, PA, USA
| | - Shantu Amin
- Department of Pharmacology, Penn State University, University Park 16801, PA, USA
| | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville 22908, VA, USA
| | - Mark Kester
- Department of Pharmacology, University of Virginia, Charlottesville 22908, VA, USA; Department of Molecular Physiology and Biophysics, University of Virginia, Charlottesville 22908, VA, USA
| | - John Katsaras
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge 37831, TN, USA; Joint Institute for Neutron Sciences, Oak Ridge National Laboratory, Oak Ridge 37831, TN, USA; Department of Physics and Astronomy, University of Tennessee, Knoxville 37996, TN, USA.
| | - Drew Marquardt
- Department of Chemistry and Biochemistry, University of Windsor, Windsor N9B 3P4, ON, Canada; Department of Physics, University of Windsor, Windsor N9B 3P4, ON, Canada.
| | | |
Collapse
|
9
|
Application of Proteogenomics to Urine Analysis towards the Identification of Novel Biomarkers of Prostate Cancer: An Exploratory Study. Cancers (Basel) 2022; 14:cancers14082001. [PMID: 35454907 PMCID: PMC9031064 DOI: 10.3390/cancers14082001] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/12/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Prostate cancer (PCa) is one of the most common cancers. Due to the limited and invasive approaches for PCa diagnosis, it is crucial to identify more accurate and non-invasive biomarkers for its detection. The aim of our study was to non-invasively uncover new protein targets for detecting PCa using a proteomics and proteogenomics approach. This work identified several dysregulated mutant protein isoforms in urine from PCa patients, some of them predicted to have a protective or an adverse role in these patients. These results are promising given urine’s non-invasive nature and offers an auspicious opportunity for research and development of PCa biomarkers. Abstract To identify new protein targets for PCa detection, first, a shotgun discovery experiment was performed to characterize the urinary proteome of PCa patients. This revealed 18 differentially abundant urinary proteins in PCa patients. Second, selected targets were clinically tested by immunoblot, and the soluble E-cadherin fragment was detected for the first time in the urine of PCa patients. Third, the proteogenome landscape of these PCa patients was characterized, revealing 1665 mutant protein isoforms. Statistical analysis revealed 6 differentially abundant mutant protein isoforms in PCa patients. Analysis of the likely effects of mutations on protein function and PPIs involving the dysregulated mutant protein isoforms suggests a protective role of mutations HSPG2*Q1062H and VASN*R161Q and an adverse role of AMBP*A286G and CD55*S162L in PCa patients. This work originally characterized the urinary proteome, focusing on the proteogenome profile of PCa patients, which is usually overlooked in the analysis of PCa and body fluids. Combined analysis of mass spectrometry data using two different software packages was performed for the first time in the context of PCa, which increased the robustness of the data analysis. The application of proteogenomics to urine proteomic analysis can be very enriching in mutation-related diseases such as cancer.
Collapse
|
10
|
Trapika IGMGSC, Liu XT, Chung LH, Lai F, Xie C, Zhao Y, Cui S, Chen J, Tran C, Wang Q, Zhang S, Don AS, Li GQ, Hanrahan JR, Qi Y. Ceramide Regulates Anti-Tumor Mechanisms of Erianin in Androgen-Sensitive and Castration-Resistant Prostate Cancers. Front Oncol 2021; 11:738078. [PMID: 34604081 PMCID: PMC8484793 DOI: 10.3389/fonc.2021.738078] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/02/2021] [Indexed: 01/09/2023] Open
Abstract
Prostate cancer is the second most prevalent malignancy worldwide. In the early stages, the development of prostate cancer is dependent on androgens. Over time with androgen deprivation therapy, 20% of prostate cancers progress to a castration-resistant form. Novel treatments for prostate cancers are still urgently needed. Erianin is a plant-derived bibenzyl compound. We report herein that erianin exhibits anti-tumor effects in androgen-sensitive and castration-resistant prostate cancer cells through different mechanisms. Erianin induces endoplasmic reticulum stress-associated apoptosis in androgen-sensitive prostate cancer cells. It also triggers pro-survival autophagic responses, as inhibition of autophagy predisposes to apoptosis. In contrast, erianin fails to induce apoptosis in castration-resistant prostate cancer cells. Instead, it results in cell cycle arrest at the M phase. Mechanistically, C16 ceramide dictates differential responses of androgen-sensitive and castration-resistant prostate cancer cells to erianin. Erianin elevates C16 ceramide level in androgen-sensitive but not castration-resistant prostate cancer cells. Overexpression of ceramide synthase 5 that specifically produces C16 ceramide enables erianin to induce apoptosis in castration-resistant prostate cancer cells. Our study provides both experimental evidence and mechanistic data showing that erianin is a potential treatment option for prostate cancers.
Collapse
Affiliation(s)
- I Gusti Md Gde Surya C. Trapika
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Xin Tracy Liu
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Long Hoa Chung
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Felcia Lai
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Chanlu Xie
- Department of Endocrinology, Royal Prince Alfred Hospital, Sydney, NSW, Australia,Chinese Medicine Anti-Cancer Evaluation Program, Central Clinical School, University of Sydney, Camperdown, NSW, Australia
| | - Yang Zhao
- Department of Biochemistry and Molecular Biology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shaohui Cui
- Key Laboratory of Biotechnology and Biorescources Utilization of Ministry of Education, Dalian Minzu University, Dalian, China
| | - Jinbiao Chen
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Collin Tran
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia
| | - Qian Wang
- Translational Cancer Metabolism Laboratory, School of Medical Sciences and Prince of Wales Clinical School, UNSW, Sydney, NSW, Australia
| | - Shubiao Zhang
- Key Laboratory of Biotechnology and Biorescources Utilization of Ministry of Education, Dalian Minzu University, Dalian, China
| | - Anthony S. Don
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,School of Medical Sciences, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - George Qian Li
- School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Jane R. Hanrahan
- School of Pharmacy, Faculty of Health and Medicine, University of Sydney, Camperdown, NSW, Australia,*Correspondence: Yanfei Qi, ; Jane R. Hanrahan,
| | - Yanfei Qi
- Centenary Institute of Cancer Medicine and Cell Biology, University of Sydney, Camperdown, NSW, Australia,*Correspondence: Yanfei Qi, ; Jane R. Hanrahan,
| |
Collapse
|
11
|
Munk R, Anerillas C, Rossi M, Tsitsipatis D, Martindale JL, Herman AB, Yang JH, Roberts JA, Varma VR, Pandey PR, Thambisetty M, Gorospe M, Abdelmohsen K. Acid ceramidase promotes senescent cell survival. Aging (Albany NY) 2021; 13:15750-15769. [PMID: 34102611 PMCID: PMC8266329 DOI: 10.18632/aging.203170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 05/18/2021] [Indexed: 01/18/2023]
Abstract
Cellular senescence is linked to chronic age-related diseases including atherosclerosis, diabetes, and neurodegeneration. Compared to proliferating cells, senescent cells express distinct subsets of proteins. In this study, we used cultured human diploid fibroblasts rendered senescent through replicative exhaustion or ionizing radiation to identify proteins differentially expressed during senescence. We identified acid ceramidase (ASAH1), a lysosomal enzyme that cleaves ceramide into sphingosine and fatty acid, as being highly elevated in senescent cells. This increase in ASAH1 levels in senescent cells was associated with a rise in the levels of ASAH1 mRNA and a robust increase in ASAH1 protein stability. Furthermore, silencing ASAH1 in pre-senescent fibroblasts decreased the levels of senescence proteins p16, p21, and p53, and reduced the activity of the senescence-associated β-galactosidase. Interestingly, depletion of ASAH1 in pre-senescent cells sensitized these cells to the senolytics Dasatinib and Quercetin (D+Q). Together, our study indicates that ASAH1 promotes senescence, protects senescent cells, and confers resistance against senolytic drugs. Given that inhibiting ASAH1 sensitizes cells towards senolysis, this enzyme represents an attractive therapeutic target in interventions aimed at eliminating senescent cells.
Collapse
Affiliation(s)
- Rachel Munk
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Carlos Anerillas
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Martina Rossi
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Dimitrios Tsitsipatis
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Jennifer L Martindale
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Allison B Herman
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Jen-Hao Yang
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Jackson A Roberts
- Laboratory of Behavioral Neuroscience, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Vijay R Varma
- Laboratory of Behavioral Neuroscience, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Poonam R Pandey
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Madhav Thambisetty
- Laboratory of Behavioral Neuroscience, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging-Intramural Research Program, National Institutes of Health, Baltimore, Maryland 21224, USA
| |
Collapse
|
12
|
Malvi P, Janostiak R, Nagarajan A, Zhang X, Wajapeyee N. N-acylsphingosine amidohydrolase 1 promotes melanoma growth and metastasis by suppressing peroxisome biogenesis-induced ROS production. Mol Metab 2021; 48:101217. [PMID: 33766731 PMCID: PMC8081993 DOI: 10.1016/j.molmet.2021.101217] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/02/2021] [Accepted: 03/17/2021] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE Metabolic deregulation is a key hallmark of cancer cells and has been shown to drive cancer growth and metastasis. However, not all metabolic drivers of melanoma are known. Based on our finding that N-acylsphingosine amidohydrolase 1 (ASAH1) is overexpressed in melanoma, the objective of these studies was to establish its role in melanoma tumor growth and metastasis, understand its mechanism of action, and evaluate ASAH1 targeting for melanoma therapy. METHODS We used publicly available melanoma datasets and patient-derived samples of melanoma and normal skin tissue and analyzed them for ASAH1 mRNA expression and ASAH1 protein expression using immunohistochemistry. ASAH1 was knocked down using short-hairpin RNAs in multiple melanoma cell lines that were tested in a series of cell culture-based assays and mouse-based melanoma xenograft assays to monitor the effect of ASAH1 knockdown on melanoma tumor growth and metastasis. An unbiased metabolomics analysis was performed to identify the mechanism of ASAH1 action. Based on the metabolomics findings, the role of peroxisome-mediated reactive oxygen species (ROS) production was explored in regard to mediating the effect of ASAH1. The ASAH1 inhibitor was used alone or in combination with a BRAFV600E inhibitor to evaluate the therapeutic value of ASAH1 targeting for melanoma therapy. RESULTS We determined that ASAH1 was overexpressed in a large percentage of melanoma cells and regulated by transcription factor E2F1 in a mitogen-activated protein (MAP) kinase pathway-dependent manner. ASAH1 expression was necessary to maintain melanoma tumor growth and metastatic attributes in cell cultures and mouse models of melanoma tumor growth and metastasis. To identify the mechanism by which ASAH1 facilitates melanoma tumor growth and metastasis, we performed a large-scale and unbiased metabolomics analysis of melanoma cells expressing ASAH1 short-hairpin RNAs (shRNAs). We found that ASAH1 inhibition increased peroxisome biogenesis through ceramide-mediated PPARγ activation. ASAH1 loss increased ceramide and peroxisome-derived ROS, which in turn inhibited melanoma growth. Pharmacological inhibition of ASAH1 also attenuated melanoma growth and enhanced the effectiveness of BRAF kinase inhibitor in the cell cultures and mice. CONCLUSIONS Collectively, these results demonstrate that ASAH1 is a druggable driver of melanoma tumor growth and metastasis that functions by suppressing peroxisome biogenesis, thereby inhibiting peroxisome-derived ROS production. These studies also highlight the therapeutic utility of ASAH1 inhibitors for melanoma therapy.
Collapse
Affiliation(s)
- Parmanand Malvi
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA
| | - Radoslav Janostiak
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA; Institute for Research in Biomedicine (IRB Barcelona), The Barcelona Institute of Science and Technology, Barcelona, 08028, Spain
| | - Arvindhan Nagarajan
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Xuchen Zhang
- Department of Pathology, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Narendra Wajapeyee
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Alabama, 35233, USA.
| |
Collapse
|
13
|
Quinville BM, Deschenes NM, Ryckman AE, Walia JS. A Comprehensive Review: Sphingolipid Metabolism and Implications of Disruption in Sphingolipid Homeostasis. Int J Mol Sci 2021; 22:ijms22115793. [PMID: 34071409 PMCID: PMC8198874 DOI: 10.3390/ijms22115793] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/23/2021] [Accepted: 05/24/2021] [Indexed: 12/16/2022] Open
Abstract
Sphingolipids are a specialized group of lipids essential to the composition of the plasma membrane of many cell types; however, they are primarily localized within the nervous system. The amphipathic properties of sphingolipids enable their participation in a variety of intricate metabolic pathways. Sphingoid bases are the building blocks for all sphingolipid derivatives, comprising a complex class of lipids. The biosynthesis and catabolism of these lipids play an integral role in small- and large-scale body functions, including participation in membrane domains and signalling; cell proliferation, death, migration, and invasiveness; inflammation; and central nervous system development. Recently, sphingolipids have become the focus of several fields of research in the medical and biological sciences, as these bioactive lipids have been identified as potent signalling and messenger molecules. Sphingolipids are now being exploited as therapeutic targets for several pathologies. Here we present a comprehensive review of the structure and metabolism of sphingolipids and their many functional roles within the cell. In addition, we highlight the role of sphingolipids in several pathologies, including inflammatory disease, cystic fibrosis, cancer, Alzheimer’s and Parkinson’s disease, and lysosomal storage disorders.
Collapse
|
14
|
Ablation of Acid Ceramidase Impairs Autophagy and Mitochondria Activity in Melanoma Cells. Int J Mol Sci 2021; 22:ijms22063247. [PMID: 33806766 PMCID: PMC8004726 DOI: 10.3390/ijms22063247] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/15/2021] [Accepted: 03/18/2021] [Indexed: 12/12/2022] Open
Abstract
Cutaneous melanoma is often resistant to therapy due to its high plasticity, as well as its ability to metabolise chemotherapeutic drugs. Sphingolipid signalling plays a pivotal role in its progression and metastasis. One of the ways melanoma alters sphingolipid rheostat is via over-expression of lysosomal acid ceramidase (AC), which catalyses the hydrolysis of pro-apoptotic long-chain ceramides into sphingosine and fatty acid. In this report, we examine the role of acid ceramidase in maintaining cellular homeostasis through the regulation of autophagy and mitochondrial activity in melanoma cell lines. We show that under baseline conditions, wild-type melanoma cells had 3-fold higher levels of the autophagy marker, microtubule-associated proteins 1A/1B light chain 3B (LC3 II), compared to AC-null cells. This difference was further magnified after cell starvation. Moreover, we noticed autophagy impairment in A375 AC-null cells, possibly due to local accumulation of non-metabolized ceramides. Nonetheless, we observed that AC-null cells exhibited a significant increase in mitochondrial membrane potential compared to control cells. Consistent with this observation, we found that, after total starvation, ~30% of AC-null cells undergo apoptosis compared to ~6% of wild-type cells. As expected, AC transfection restored viability in A375 AC-null cells. Together, these findings suggest that AC-null melanoma cells change and adapt their metabolism to survive in the absence of AC, although in a way that does not allow them to cope with the stress of nutrient deprivation.
Collapse
|
15
|
Bielsa N, Casasampere M, Aseeri M, Casas J, Delgado A, Abad JL, Fabriàs G. Discovery of deoxyceramide analogs as highly selective ACER3 inhibitors in live cells. Eur J Med Chem 2021; 216:113296. [PMID: 33677352 DOI: 10.1016/j.ejmech.2021.113296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/27/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
Acid (AC), neutral (NC) and alkaline ceramidase 3 (ACER3) are the most ubiquitous ceramidases and their therapeutic interest as targets in cancer diseases has been well sustained. This supports the importance of discovering potent and specific inhibitors for further use in combination therapies. Although several ceramidase inhibitors have been reported, most of them target AC and a few focus on NC. In contrast, well characterized ACER3 inhibitors are lacking. Here we report on the synthesis and screening of two series of 1-deoxy(dihydro)ceramide analogs on the three enzymes. Activity was determined using fluorogenic substrates in recombinant human NC (rhNC) and both lysates and intact cells enriched in each enzyme. None of the molecules elicited a remarkable AC inhibitory activity in either experimental setup, while using rhNC, several compounds of both series were active as non-competitive inhibitors with Ki values between 1 and 5 μM. However, a dramatic loss of potency occurred in NC-enriched cell lysates and no activity was elicited in intact cells. Interestingly, several compounds of Series 2 inhibited ACER3 dose-dependently in both cell lysates and intact cells with IC50's around 20 μM. In agreement with their activity in live cells, they provoked a significant increase in the amounts of ceramides. Overall, this study identifies highly selective ACER3 activity blockers in intact cells, opening the door to further medicinal chemistry efforts aimed at developing more potent and specific compounds.
Collapse
Affiliation(s)
- Núria Bielsa
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Mireia Casasampere
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Mazen Aseeri
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII, 28029, Madrid, Spain
| | - Antonio Delgado
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Department of Pharmacology, Toxicology and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC). Faculty of Pharmacy. University of Barcelona (UB). Avda. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - José Luis Abad
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII, 28029, Madrid, Spain.
| |
Collapse
|
16
|
Cholesterol and Sphingolipid Enriched Lipid Rafts as Therapeutic Targets in Cancer. Int J Mol Sci 2021; 22:ijms22020726. [PMID: 33450869 PMCID: PMC7828315 DOI: 10.3390/ijms22020726] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023] Open
Abstract
Lipid rafts are critical cell membrane lipid platforms enriched in sphingolipid and cholesterol content involved in diverse cellular processes. They have been proposed to influence membrane properties and to accommodate receptors within themselves by facilitating their interaction with ligands. Over the past decade, technical advances have improved our understanding of lipid rafts as bioactive structures. In this review, we will cover the more recent findings about cholesterol, sphingolipids and lipid rafts located in cellular and nuclear membranes in cancer. Collectively, the data provide insights on the role of lipid rafts as biomolecular targets in cancer with good perspectives for the development of innovative therapeutic strategies.
Collapse
|
17
|
Ren R, Pang B, Han Y, Li Y. A Glimpse of the Structural Biology of the Metabolism of Sphingosine-1-Phosphate. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2021; 4:2515256421995601. [PMID: 37366379 PMCID: PMC10243590 DOI: 10.1177/2515256421995601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/28/2021] [Accepted: 01/28/2021] [Indexed: 06/28/2023]
Abstract
As a key sphingolipid metabolite, sphingosine-1-phosphate (S1P) plays crucial roles in vascular and immune systems. It regulates angiogenesis, vascular integrity and homeostasis, allergic responses, and lymphocyte trafficking. S1P is interconverted with sphingosine, which is also derived from the deacylation of ceramide. S1P levels and the ratio to ceramide in cells are tightly regulated by its metabolic pathways. Abnormal S1P production causes the occurrence and progression of numerous severe diseases, such as metabolic syndrome, cancers, autoimmune disorders such as multiple sclerosis, and kidney and cardiovascular diseases. In recent years, huge advances on the structure of S1P metabolic pathways have been accomplished. In this review, we have got a glimpse of S1P metabolism through structural and biochemical studies of: sphingosine kinases, S1P transporters and S1P receptors, and the development of therapeutics targeting S1P signaling. The progress we summarize here could provide fresh perspectives to further the exploration of S1P functions and facilitate the development of therapeutic molecules targeting S1P signaling with improved specificity and therapeutic effects.
Collapse
Affiliation(s)
- Ruobing Ren
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Bin Pang
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yufei Han
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| | - Yihao Li
- Kobilka Institute of Innovative Drug
Discovery, School of Life and Health Sciences, the Chinese University
of Hong Kong, Shenzhen, China
| |
Collapse
|
18
|
Vethakanraj HS, Chandrasekaran N, Sekar AK. Acid ceramidase, a double-edged sword in cancer aggression: A minireview. Curr Cancer Drug Targets 2020; 21:CCDT-EPUB-112652. [PMID: 33357194 DOI: 10.2174/1568009620666201223154621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 10/18/2020] [Accepted: 10/30/2020] [Indexed: 11/22/2022]
Abstract
Acid ceramidase (AC), the key enzyme of the ceramide metabolic pathway hydrolyzes pro-apoptotic ceramide to sphingosine, which by the action of sphingosine-1-kinase is metabolized to mitogenic sphingosine-1-phosphate. The intracellular level of AC determines ceramide/sphingosine-1-phosphate rheostat which in turn decides the cell fate. The upregulated AC expression during cancerous condition acts as a "double-edged sword" by converting pro-apoptotic ceramide to anti-apoptotic sphingosine-1-phosphate, wherein on one end, the level of ceramide is decreased and on the other end, the level of sphingosine-1-phosphate is increased, thus altogether aggravating the cancer progression. In addition, cancer cells with upregulated AC expression exhibited increased cell proliferation, metastasis, chemoresistance, radioresistance and numerous strategies were developed in the past to effectively target the enzyme. Gene silencing and pharmacological inhibition of AC sensitized the resistant cells to chemo/radiotherapy thereby promoting cell death. The core objective of this review is to explore AC mediated tumour progression and the potential role of AC inhibitors in various cancer cell lines/models.
Collapse
|
19
|
Caputo S, Di Martino S, Cilibrasi V, Tardia P, Mazzonna M, Russo D, Penna I, Summa M, Bertozzi SM, Realini N, Margaroli N, Migliore M, Ottonello G, Liu M, Lansbury P, Armirotti A, Bertorelli R, Ray SS, Skerlj R, Scarpelli R. Design, Synthesis, and Biological Evaluation of a Series of Oxazolone Carboxamides as a Novel Class of Acid Ceramidase Inhibitors. J Med Chem 2020; 63:15821-15851. [PMID: 33290061 PMCID: PMC7770833 DOI: 10.1021/acs.jmedchem.0c01561] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
![]()
Acid
ceramidase (AC) is a cysteine hydrolase that plays a crucial
role in the metabolism of lysosomal ceramides, important members of
the sphingolipid family, a diversified class of bioactive molecules
that mediate many biological processes ranging from cell structural
integrity, signaling, and cell proliferation to cell death. In the
effort to expand the structural diversity of the existing collection
of AC inhibitors, a novel class of substituted oxazol-2-one-3-carboxamides
were designed and synthesized. Herein, we present the chemical optimization
of our initial hits, 2-oxo-4-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 8a and 2-oxo-5-phenyl-N-(4-phenylbutyl)oxazole-3-carboxamide 12a, which resulted in the identification of 5-[4-fluoro-2-(1-methyl-4-piperidyl)phenyl]-2-oxo-N-pentyl-oxazole-3-carboxamide 32b as a potent
AC inhibitor with optimal physicochemical and metabolic properties,
showing target engagement in human neuroblastoma SH-SY5Y cells and
a desirable pharmacokinetic profile in mice, following intravenous
and oral administration. 32b enriches the arsenal of
promising lead compounds that may therefore act as useful pharmacological
tools for investigating the potential therapeutic effects of AC inhibition
in relevant sphingolipid-mediated disorders.
Collapse
Affiliation(s)
- Samantha Caputo
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Simona Di Martino
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Vincenzo Cilibrasi
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Piero Tardia
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Marco Mazzonna
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Debora Russo
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,D3-Pharma Chemistry, Via Morego 30, I-16163 Genova, Italy
| | - Ilaria Penna
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,D3-Pharma Chemistry, Via Morego 30, I-16163 Genova, Italy
| | - Maria Summa
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Sine Mandrup Bertozzi
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Natalia Realini
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Natasha Margaroli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Marco Migliore
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| | - Giuliana Ottonello
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Min Liu
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Peter Lansbury
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Andrea Armirotti
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Rosalia Bertorelli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Analytical Chemistry and Translational Pharmacology, Via Morego 30, I-16163 Genova, Italy
| | - Soumya S Ray
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Renato Skerlj
- Lysosomal Therapeutics Inc., 19 Blackstone Street, Cambridge, Massachusetts 02139, United States
| | - Rita Scarpelli
- Fondazione Istituto Italiano di Tecnologia, Via Morego 30, I-16163 Genova, Italy.,Drug Discovery and Development (D3)-Validation, Via Morego 30, I-16163 Genova, Italy
| |
Collapse
|
20
|
Fisher-Wellman KH, Hagen JT, Neufer PD, Kassai M, Cabot MC. On the nature of ceramide-mitochondria interactions - Dissection using comprehensive mitochondrial phenotyping. Cell Signal 2020; 78:109838. [PMID: 33212155 DOI: 10.1016/j.cellsig.2020.109838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 02/06/2023]
Abstract
Sphingolipids are a unique class of lipids owing to their non-glycerol-containing backbone, ceramide, that is constructed from a long-chain aliphatic amino alcohol, sphinganine, to which a fatty acid is attached via an amide bond. Ceramide plays a star role in the initiation of apoptosis by virtue of its interactions with mitochondria, a control point for a downstream array of signaling cascades culminating in apoptosis. Many pathways converge on mitochondria to elicit mitochondrial outer membrane permeabilization (MOMP), a step that corrupts bioenergetic service. Although much is known regarding ceramides interaction with mitochondria and the ensuing cell signal transduction cascades, how ceramide impacts the elements of mitochondrial bioenergetic function is poorly understood. The objective of this review is to introduce the reader to sphingolipid metabolism, present a snapshot of mitochondrial respiration, elaborate on ceramides convergence on mitochondria and the upstream players that collaborate to elicit MOMP, and introduce a mitochondrial phenotyping platform that can be of utility in dissecting the fine-points of ceramide impact on cellular bioenergetics.
Collapse
Affiliation(s)
- Kelsey H Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States of America.
| | - James T Hagen
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States of America
| | - P Darrell Neufer
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States of America
| | - Miki Kassai
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States of America
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC, United States of America.
| |
Collapse
|
21
|
Casasampere M, Izquierdo E, Casas J, Abad JL, Liu X, Xu R, Mao C, Chang YT, Delgado A, Fabrias G. Click and count: specific detection of acid ceramidase activity in live cells. Chem Sci 2020; 11:13044-13051. [PMID: 34094488 PMCID: PMC8163297 DOI: 10.1039/d0sc03166f] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Accepted: 10/16/2020] [Indexed: 01/02/2023] Open
Abstract
The use of intact cells in medical research offers a number of advantages over employing cell-free systems. In diagnostics, cells isolated from liquid biopsies can be directly used, speeding up the time of analysis and diminishing the risk of protein degradation by sample manipulation. In drug discovery, studies in live cells take into account aspects neglected in cell-free systems, such as uptake, metabolization, and subcellular concentration by compartmentalization of potential drug candidates. Therefore, probes for studies in cellulo are of paramount importance. Acid ceramidase (AC) is a lysosomal enzyme that hydrolyses ceramides into sphingoid bases and fatty acids. The essential role of this enzyme in the outburst and progress of several diseases, some of them still incurable, is well sustained. Despite the great clinical relevance of AC as a biomarker and therapeutic target, the specific monitoring of AC activity in live cells has remained elusive due to the concomitant existence of neutral and alkaline ceramidases. In this work, we report that 1-deoxydihydroceramides are exclusively hydrolysed by AC. Using N-octanoyl-18-azidodeoxysphinganine as a probe and a BODIPY-substituted bicyclononyne, we show the click-reliant predominant staining of lysosomes, with extra-lysosomal labeling also occurring in some cells. Importantly, using pharmacological and genetic tools together with high resolution mass spectrometry, we demonstrate that both lysosomal and extra-lysosomal staining are AC-dependent. These findings are translated into the specific flow cytometry monitoring of AC activity in intact cells, which fills an important gap in the field of diseases linked to altered AC activity.
Collapse
Affiliation(s)
- Mireia Casasampere
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
| | - Eduardo Izquierdo
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII 28029 Madrid Spain
| | - José Luís Abad
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
| | - Xiao Liu
- Department of Chemistry, Pohang University of Science and Technology (POSTECH) Pohang Gyeongbuk 37673 Korea
| | - Ruijuan Xu
- Department of Medicine and Cancer Center, The State University of New York at Stony Brook Room 9M-0834, MART Building, 100 Nicolls Road, Stony Brook NY 11794 USA
| | - Cungui Mao
- Department of Medicine and Cancer Center, The State University of New York at Stony Brook Room 9M-0834, MART Building, 100 Nicolls Road, Stony Brook NY 11794 USA
| | - Young-Tae Chang
- Department of Chemistry, Pohang University of Science and Technology (POSTECH) Pohang Gyeongbuk 37673 Korea
- Center for Self-assembly and Complexity, Institute for Basic Science (IBS) Pohang Gyeongbuk 37673 Korea
| | - Antonio Delgado
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
- Department of Pharmacology, Toxicology and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), Faculty of Pharmacy, University of Barcelona Avda. Joan XXIII s/n 08028 Barcelona Spain
| | - Gemma Fabrias
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) Jordi Girona 18 08034-Barcelona Spain
- Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII 28029 Madrid Spain
| |
Collapse
|
22
|
Synthesis and HPLC-ECD Study of Cytostatic Condensed O,N-Heterocycles Obtained from 3-Aminoflavanones. Biomolecules 2020; 10:biom10101462. [PMID: 33092128 PMCID: PMC7593906 DOI: 10.3390/biom10101462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 11/16/2022] Open
Abstract
Racemic chiral O,N-heterocycles containing 2-arylchroman or 2-aryl-2H-chromene subunit condensed with morpholine, thiazole, or pyrrole moieties at the C-3-C-4 bond were synthesized with various substitution patterns of the aryl group by the cyclization of cis- or trans-3-aminoflavanone analogues. The 3-aminoflavanone precursors were obtained in a Neber rearrangement of oxime tosylates of flavanones, which provided the trans diastereomer as the major product and enabled the isolation of both the cis- and trans-diastereomers. The cis- and trans-aminoflavanones were utilized to prepare three diastereomers of 5-aryl-chromeno[4,3-b][1,4]oxazines. Antiproliferative activity of the condensed heterocycles and precursors was evaluated against A2780 and WM35 cancer cell lines. For a 3-(N-chloroacetylamino)-flavan-4-ol derivative, showing structural analogy with acyclic acid ceramidase inhibitors, 0.15 μM, 3.50 μM, and 6.06 μM IC50 values were measured against A2780, WM35, and HaCat cell lines, and apoptotic mechanism was confirmed. Low micromolar IC50 values down to 2.14 μM were identified for the thiazole- and pyrrole-condensed 2H-chromene derivatives. Enantiomers of the condensed heterocycles were separated by HPLC using chiral stationary phase, HPLC-ECD spectra were recorded and TDDFT-ECD calculations were performed to determine the absolute configuration and solution conformation. Characteristic ECD transitions of the separated enantiomers were correlated with the absolute configuration and effect of substitution pattern on the HPLC elution order was determined.
Collapse
|
23
|
Clifford RE, Bowden D, Blower E, Kirwan CC, Vimalachandran D. Does tamoxifen have a therapeutic role outside of breast cancer? A systematic review of the evidence. Surg Oncol 2020; 33:100-107. [PMID: 32561074 DOI: 10.1016/j.suronc.2020.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 02/08/2020] [Indexed: 02/04/2023]
Abstract
INTRODUCTION Tamoxifen is a widely used hormonal based therapy for breast cancer in the adjuvant and metastatic setting, prolonging overall and recurrence-free survival. There has been increasing interest in the potential for novel "off-target" effects of tamoxifen and its metabolite N-desmethyltamoxifen across a number of cancer types. We aim to review the current literature regarding the potential use of tamoxifen in other primary malignancies. METHOD A qualitative systematic review was performed according to the PRISMA guidelines using pre-set search criteria across the PubMed, Cochrane and Scopus databases from 1985 to 2019. Additional results were generated from included papers references. RESULTS A total of 324 papers were identified, of which 47 were included; a further 29 articles were obtained from additional referencing to give a total of 76 articles. Clinical trials have demonstrated benefits with the use of tamoxifen in isolation and combination, specifically in patients with advanced non-resectable malignancy, however results are not consistent across the literature. In vivo data consistently suggests that off target effects of tamoxifen are mediated through the ceramide pathway or through inhibition of protein kinase C (PKC). CONCLUSIONS With increased focus upon the potential of repurposing drugs, tamoxifen may be a candidate for repurposing in the wider cancer setting. There is evidence to suggest that the ceramide or PKC pathway could act as a therapeutic target for tamoxifen or alternative chemotherapeutics and merits further investigation.
Collapse
Affiliation(s)
- R E Clifford
- Institute of Cancer Medicine, The University of Liverpool, UK.
| | - D Bowden
- Institute of Cancer Medicine, The University of Liverpool, UK
| | - E Blower
- Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - C C Kirwan
- Cancer Research UK Manchester Institute, The University of Manchester, UK
| | - D Vimalachandran
- Institute of Cancer Medicine, The University of Liverpool, UK; The Countess of Chester Foundation Trust, UK
| |
Collapse
|
24
|
White-Gilbertson S, Voelkel-Johnson C. Giants and monsters: Unexpected characters in the story of cancer recurrence. Adv Cancer Res 2020; 148:201-232. [PMID: 32723564 DOI: 10.1016/bs.acr.2020.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Polyploid giant cancer cells (PGCC) constitute a dangerous subpopulation of cancer cells and are a driving force in cancer recurrence. These unique cells arise from diploid tumor cells in response to stress encountered in the tumor microenvironment or during cancer therapy. PGCC are greatly dedifferentiated, acquire pluripotency, and are able to replicate through a form of asymmetric division called neosis, which results in new populations that are themselves able to differentiate into new cell types or to re-establish tumors. Progeny tend to be more genetically unstable than the founding population due to the dysregulation required to transition through a PGCC state. Therefore, cancers that escape stressors through this mechanism tend to re-emerge with a more aggressive phenotype that is therapy resistant. This review focuses on the clinical significance of PGCC, the need for standardized nomenclature and molecular markers, as well as possible avenues to develop therapies aimed at PGCC and the process of neosis. The biology underlying the development of PGCC including cell cycle checkpoint dysregulation, stress responses, dedifferentiation, stemness and epithelial-mesenchymal transition is discussed.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States.
| |
Collapse
|
25
|
Li Y, He CL, Li WX, Zhang RX, Duan Y. Transcriptome analysis reveals gender-specific differences in overall metabolic response of male and female patients in lung adenocarcinoma. PLoS One 2020; 15:e0230796. [PMID: 32236130 PMCID: PMC7112214 DOI: 10.1371/journal.pone.0230796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 03/08/2020] [Indexed: 12/23/2022] Open
Abstract
Background Evidence from multiple studies suggests metabolic abnormalities play an important role in lung cancer. Lung adenocarcinoma (LUAD) is the most common subtype of lung cancer. The present study aimed to explore differences in the global metabolic response between male and female patients in LUAD and to identify the metabolic genes associated with lung cancer susceptibility. Methods Transcriptome and clinical LUAD data were acquired from The Cancer Genome Atlas (TCGA) database. Information on metabolic genes and metabolic subsystems were collected from the Recon3D human metabolic model. Two validation datasets (GSE68465 and GSE72094) were downloaded from the Gene Expression Omnibus (GEO) database. Differential expression analysis, gene set enrichment analysis and protein-protein interaction networks were used to identified key metabolic pathways and genes. Functional experiments were used to verify the effects of genes on proliferation, migration, and invasion in lung cancer cells in vitro. Results Samples of tumors and adjacent non-tumor tissue from both male and female patients exhibited distinct global patterns of gene expression. In addition, we found large differences in methionine and cysteine metabolism, pyruvate metabolism, cholesterol metabolism, nicotinamide adenine dinucleotide (NAD) metabolism, and nuclear transport between male and female LUAD patients. We identified 34 metabolic genes associated with lung cancer susceptibility in males and 15 in females. Most of the metabolic cancer-susceptibility genes had high prediction accuracy for lung cancer (AUC > 0.9). Furthermore, both bioinformatics analysis and experimental results showed that TAOK2 was down-regulated and ASAH1 was up-regulated in male tumor tissue and female tumor tissue in LUAD. Functional experiments showed that inhibiting ASAH1 suppressed the proliferation, migration, and invasion of lung cancer cells. Conclusions Metabolic cancer-susceptibility genes may be used alone or in combination as diagnostic markers for LUAD. Further studies are required to elucidate the functions of these genes in LUAD.
Collapse
Affiliation(s)
- Ya Li
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, Yunnan, China
- Yunnan Institute of Laboratory Diagnosis, Kunming, Yunnan, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Cheng-Lu He
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, Yunnan, China
- Yunnan Institute of Laboratory Diagnosis, Kunming, Yunnan, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wen-Xing Li
- Key Laboratory of Animal Models and Human Disease Mechanisms, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Rui-Xian Zhang
- Yunnan Center for Disease Control and Prevention, Kunming, Yunnan, China
| | - Yong Duan
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, Yunnan, China
- Yunnan Institute of Laboratory Diagnosis, Kunming, Yunnan, China
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
- * E-mail:
| |
Collapse
|
26
|
Tomizawa S, Tamori M, Tanaka A, Utsumi N, Sato H, Hatakeyama H, Hisaka A, Kohama T, Yamagata K, Honda T, Nakamura H, Murayama T. Inhibitory effects of ceramide kinase on Rac1 activation, lamellipodium formation, cell migration, and metastasis of A549 lung cancer cells. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158675. [PMID: 32112978 DOI: 10.1016/j.bbalip.2020.158675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 01/30/2020] [Accepted: 02/22/2020] [Indexed: 01/01/2023]
Abstract
Ceramide kinase (CerK) phosphorylates ceramide to ceramide-1-phosphate (C1P), a bioactive sphingolipid. Since the mechanisms responsible for regulating the proliferation and migration/metastasis of cancer cells by the CerK/C1P pathway remain unclear, we conducted the present study. The knockdown of CerK in A549 lung and MCF-7 breast cancer cells (shCerK cells) increased the formation of lamellipodia, which are membrane protrusions coupled with cell migration. Mouse embryonic fibroblasts prepared from CerK-null mice also showed an enhanced formation of lamellipodia. The overexpression of CerK inhibited lamellipodium formation in A549 cells. The knockdown of CerK increased the number of cells having lamellipodia with Rac1 and the levels of active Rac1-GTP form, whereas the overexpression of CerK decreased them. CerK was located in lamellipodia after the epidermal growth factor treatment, indicating that CerK functioned there to inhibit Rac1. The migration of A549 cells was negatively regulated by CerK. An intravenous injection of A549-shCerK cells into nude mice resulted in markedly stronger metastatic responses in the lungs than an injection of control cells. The in vitro growth of A549 cells and in vivo expansion after the injection into mouse flanks were not affected by the CerK knockdown. These results suggest that the activation of CerK/C1P pathway has inhibitory roles on lamellipodium formation, migration, and metastasis of A549 lung cancer cells.
Collapse
Affiliation(s)
- Satoshi Tomizawa
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Mizuki Tamori
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Ai Tanaka
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Naoya Utsumi
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hiromi Sato
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hiroto Hatakeyama
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Akihiro Hisaka
- Laboratory of Clinical Pharmacology and Pharmacometrics, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Takafumi Kohama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan; Research Coordination Group, Research Management Department, Daiichi Sankyo RD Novare Co., Ltd., 1016-13 Kitakasai, Edogawa-ku, Tokyo 134-8630, Japan
| | - Kazuyuki Yamagata
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Takuya Honda
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan.
| | - Toshihiko Murayama
- Laboratory of Chemical Pharmacology, Graduate School of Pharmaceutical Sciences, Chiba University, 1-8-1 Inohana, Chuo-ku, Chiba 260-8675, Japan
| |
Collapse
|
27
|
Pearson JM, Tan SF, Sharma A, Annageldiyev C, Fox TE, Abad JL, Fabrias G, Desai D, Amin S, Wang HG, Cabot MC, Claxton DF, Kester M, Feith DJ, Loughran TP. Ceramide Analogue SACLAC Modulates Sphingolipid Levels and MCL-1 Splicing to Induce Apoptosis in Acute Myeloid Leukemia. Mol Cancer Res 2019; 18:352-363. [PMID: 31744877 DOI: 10.1158/1541-7786.mcr-19-0619] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/30/2019] [Accepted: 11/15/2019] [Indexed: 02/06/2023]
Abstract
Acute myeloid leukemia (AML) is a disease characterized by uncontrolled proliferation of immature myeloid cells in the blood and bone marrow. The 5-year survival rate is approximately 25%, and recent therapeutic developments have yielded little survival benefit. Therefore, there is an urgent need to identify novel therapeutic targets. We previously demonstrated that acid ceramidase (ASAH1, referred to as AC) is upregulated in AML and high AC activity correlates with poor patient survival. Here, we characterized a novel AC inhibitor, SACLAC, that significantly reduced the viability of AML cells with an EC50 of approximately 3 μmol/L across 30 human AML cell lines. Treatment of AML cell lines with SACLAC effectively blocked AC activity and induced a decrease in sphingosine 1-phosphate and a 2.5-fold increase in total ceramide levels. Mechanistically, we showed that SACLAC treatment led to reduced levels of splicing factor SF3B1 and alternative MCL-1 mRNA splicing in multiple human AML cell lines. This increased proapoptotic MCL-1S levels and contributed to SACLAC-induced apoptosis in AML cells. The apoptotic effects of SACLAC were attenuated by SF3B1 or MCL-1 overexpression and by selective knockdown of MCL-1S. Furthermore, AC knockdown and exogenous C16-ceramide supplementation induced similar changes in SF3B1 level and MCL-1S/L ratio. Finally, we demonstrated that SACLAC treatment leads to a 37% to 75% reduction in leukemic burden in two human AML xenograft mouse models. IMPLICATIONS: These data further emphasize AC as a therapeutic target in AML and define SACLAC as a potent inhibitor to be further optimized for future clinical development.
Collapse
Affiliation(s)
- Jennifer M Pearson
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, Virginia
| | - Su-Fern Tan
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, Virginia
| | - Arati Sharma
- Penn State Cancer Institute, Hershey, Pennsylvania.,Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | | | - Todd E Fox
- Department of Pharmacology, University of Virginia, Charlottesville, Virginia
| | - Jose Luis Abad
- Department of Biological Chemistry, Networking Biomedical Research Centre on Liver and Digestive Diseases (CIBER-EHD), Institute for Advanced Chemistry of Catalonia, Spanish National Research Council (IQAC-CSIC), Barcelona, Spain
| | - Gemma Fabrias
- Department of Biological Chemistry, Networking Biomedical Research Centre on Liver and Digestive Diseases (CIBER-EHD), Institute for Advanced Chemistry of Catalonia, Spanish National Research Council (IQAC-CSIC), Barcelona, Spain
| | - Dhimant Desai
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Shantu Amin
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania
| | - Hong-Gang Wang
- Penn State Cancer Institute, Hershey, Pennsylvania.,Department of Pediatrics, Penn State College of Medicine, Hershey, Pennsylvania
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina Diabetes and Obesity Institute, Brody School of Medicine, East Carolina University, Greenville, North Carolina
| | | | - Mark Kester
- Department of Pharmacology, Penn State College of Medicine, Hershey, Pennsylvania.,University of Virginia Cancer Center, Charlottesville, Virginia
| | - David J Feith
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, Virginia.,University of Virginia Cancer Center, Charlottesville, Virginia
| | - Thomas P Loughran
- Department of Medicine, Division of Hematology & Oncology, University of Virginia, Charlottesville, Virginia. .,University of Virginia Cancer Center, Charlottesville, Virginia
| |
Collapse
|
28
|
Vijayan Y, Lankadasari MB, Harikumar KB. Acid Ceramidase: A Novel Therapeutic Target in Cancer. Curr Top Med Chem 2019; 19:1512-1520. [PMID: 30827244 DOI: 10.2174/1568026619666190227222930] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/22/2019] [Accepted: 02/18/2019] [Indexed: 12/15/2022]
Abstract
Sphingolipids are important constituents of the eukaryotic cell membrane which govern various signaling pathways related to different aspects of cell survival. Ceramide and Sphingosine are interconvertible sphingolipid metabolites, out of which Ceramide is pro-apoptotic and sphingosine is anti-apoptotic in nature. The conversion of ceramide to sphingosine is mediated by Acid Ceramidase (ASAH1) thus maintaining a rheostat between a tumor suppressor and a tumor promoter. This rheostat is completely altered in many tumors leading to uncontrolled proliferation. This intriguing property of ASAH1 can be used by cancer cells to their advantage, by increasing the expression of the tumor promoter, sphingosine inside cells, thus creating a favorable environment for cancer growth. The different possibilities through which this enzyme serves its role in formation, progression and resistance of different types of cancers will lead to the possibility of making Acid Ceramidase a promising drug target. This review discusses the current understanding of the role of acid ceramidase in cancer progression, metastasis and resistance, strategies to develop novel natural and synthetic inhibitors of ASAH1 and their usefulness in cancer therapy.
Collapse
Affiliation(s)
- Yadu Vijayan
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Manendra Babu Lankadasari
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India.,Manipal Academy of Higher Education (MAHE), Manipal, India
| | - Kuzhuvelil B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology (RGCB), Thiruvananthapuram, Kerala State 695014, India
| |
Collapse
|
29
|
Ordóñez YF, Abad JL, Aseeri M, Casas J, Garcia V, Casasampere M, Schuchman EH, Levade T, Delgado A, Triola G, Fabrias G. Activity-Based Imaging of Acid Ceramidase in Living Cells. J Am Chem Soc 2019; 141:7736-7742. [PMID: 31030513 DOI: 10.1021/jacs.8b11687] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Acid ceramidase (AC) hydrolyzes ceramides into sphingoid bases and fatty acids. The enzyme is overexpressed in several types of cancer and Alzheimer's disease, and its genetic defect causes different incurable disorders. The availability of a method for the specific visualization of catalytically active AC in intracellular compartments is crucial for diagnosis and follow-up of therapeutic strategies in diseases linked to altered AC activity. This work was undertaken to develop activity-based probes for the detection of AC. Several analogues of the AC inhibitor SABRAC were synthesized and found to act as very potent (two-digit nM range) irreversible AC inhibitors by reaction with the active site Cys143. Detection of active AC in cell-free systems was achieved either by using fluorescent SABRAC analogues or by click chemistry with an azide-substituted analogue. The compound affording the best features allowed the unprecedented labeling of active AC in living cells.
Collapse
Affiliation(s)
- Yadira F Ordóñez
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain
| | - José Luís Abad
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain
| | - Mazen Aseeri
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain
| | - Josefina Casas
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain.,Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD) , 28029 Madrid , Spain
| | | | - Mireia Casasampere
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain
| | - Edward H Schuchman
- Department of Genetics and Genomic Sciences , Icahn School of Medicine at Mount Sinai , New York 10029 , United States
| | - Thierry Levade
- INSERM, UMR1037 CRCT , 31037 Toulouse , France.,Laboratoire de Biochimie Métabolique , Institut Fédératif de Biologie, CHU Purpan , 31300 Toulouse , France
| | - Antonio Delgado
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain.,Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), Department of Pharmacology, Toxicology and Medicinal Chemistry, Faculty of Pharmacy and Food Sciences , University of Barcelona , 08028 Barcelona , Spain
| | - Gemma Triola
- Chemical Biology group, Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain
| | - Gemma Fabrias
- Research Unit on Bioactive Molecules (RUBAM), Department of Biological Chemistry , Institute for Advanced Chemistry of Catalonia (IQAC-CSIC) , Jordi Girona, 18 , 08034 Barcelona , Spain.,Liver and Digestive Diseases Networking Biomedical Research Centre (CIBER-EHD) , 28029 Madrid , Spain
| |
Collapse
|
30
|
White-Gilbertson S, Lu P, Norris JS, Voelkel-Johnson C. Genetic and pharmacological inhibition of acid ceramidase prevents asymmetric cell division by neosis. J Lipid Res 2019; 60:1225-1235. [PMID: 30988134 DOI: 10.1194/jlr.m092247] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 03/27/2019] [Indexed: 12/17/2022] Open
Abstract
Radiation treatment failure or relapse after initial response to chemotherapy presents significant clinical challenges in cancer patients. Escape from initial courses of treatment can involve reactivation of embryonic developmental stages, with the formation of polynuclear giant cancer cells (PGCCs). This strategy of dedifferentiation can insulate cancer cells from a variety of treatments and allows a residual subpopulation to reestablish tumors after treatment. Using radiation or docetaxel chemotherapy, we generated PGCCs from prostate cancer cells. Here, we show that expression of acid ceramidase (ASAH1), an enzyme in the sphingolipid pathway linked to therapy resistance and poor outcomes, is elevated in PGCCs. Targeting ASAH1 with shRNA or treatment with the ASAH1 inhibitor, LCL-521, did not impair the formation of PGCCs, but prevented the formation of PGCC progeny that arise through an asymmetric cell division called neosis. Similar results were obtained in lung cancer cells that had been exposed to radiation or cisplatin chemotherapy as stressors. In summary, our data suggest that endoreplication occurs independent of ASAH1 while neosis is ASAH1-dependent in both prostate and lung cancer cells. Because ASAH1 knockout is embryonic lethal but not deleterious to adult animals, targeting this enzyme has the potential to be highly specific to cells undergoing the dedifferentiation process to escape cancer treatments. Pharmacological inhibition of ASAH1 is a potentially powerful strategy to eliminate cells that could otherwise serve as seed populations for recurrence.
Collapse
Affiliation(s)
- Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Ping Lu
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - James S Norris
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| | - Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425
| |
Collapse
|
31
|
Govindarajah N, Clifford R, Bowden D, Sutton PA, Parsons JL, Vimalachandran D. Sphingolipids and acid ceramidase as therapeutic targets in cancer therapy. Crit Rev Oncol Hematol 2019; 138:104-111. [PMID: 31092365 DOI: 10.1016/j.critrevonc.2019.03.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/27/2019] [Accepted: 03/30/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Sphingolipids have been shown to play a key part in cancer cell growth and death and have increasingly become the subject of novel anti-cancer therapies. Acid ceramidase, a sphingolipid enzyme, has an important role in the regulation of apoptosis. In this review we aim to assess the current evidence supporting the role of sphingolipids in cancer and the potential role that acid ceramidase may play in cancer treatment. METHODS A literature search was performed for published full text articles using the PubMed, Cochrane and Scopus databases using the search criteria string "acid ceramidase", "sphingolipid", "cancer". Additional papers were detected by scanning the references of relevant papers. A summary of the evidence for each cancer subgroup was then formed. Given the nature of the data extracted, no meta-analysis was performed. RESULTS Over expression of acid ceramidase has been demonstrated in a number of human cancers. In vitro data demonstrate that manipulation of acid ceramidase may present a useful therapeutic target. In the clinical setting, a number of drugs have been investigated with the ability to target acid ceramidase, with the most promising of those being small molecular inhibitors, such as LCL521. CONCLUSION The role of the sphingolipid pathway in cancer is becoming very clearly established by promoting ceramide accumulation in response to cancer or cellular stress. Acid ceramidase is over expressed in a variety of cancers and has a role as a potential target for inhibition by novel specific inhibitors or off-target effects of traditional anti-cancer agents. Further work is required to develop acid ceramidase inhibitors safe for progression to clinical trials.
Collapse
Affiliation(s)
- N Govindarajah
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom; Department of General Surgery, The Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom
| | - R Clifford
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom; Department of General Surgery, The Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom
| | - D Bowden
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom; Department of General Surgery, The Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom
| | - P A Sutton
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom; Department of General Surgery, The Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom
| | - J L Parsons
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom
| | - D Vimalachandran
- Institute of Translational Medicine, The University of Liverpool, Liverpool, United Kingdom; Department of General Surgery, The Countess of Chester Hospital NHS Foundation Trust, Chester, United Kingdom.
| |
Collapse
|
32
|
Wang SB, Ma YY, Chen XY, Zhao YY, Mou XZ. Ceramide-Graphene Oxide Nanoparticles Enhance Cytotoxicity and Decrease HCC Xenograft Development: A Novel Approach for Targeted Cancer Therapy. Front Pharmacol 2019; 10:69. [PMID: 30800068 PMCID: PMC6376252 DOI: 10.3389/fphar.2019.00069] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/21/2019] [Indexed: 12/19/2022] Open
Abstract
Despite substantial efforts to develop novel therapeutic strategies for treating hepatocellular carcinoma (HCC), the effectiveness and specificity of available drugs still require further improvement. Previous work has shown that exogenous ceramide can play a key role in inducing the apoptotic death of cancer cells, however, the poor water-solubility of this compound has hampered its use for cancer treatment. In the present study, we used polyethylene glycol (PEG) and polyethylenimine (PEI) co-conjugated ultra-small nano-GO (NGO-PEG-PEI) loaded with C6-ceramide (NGO-PEG-PEI/Cer) as a strategy for HCC treatment. We assessed the biological role of NGO-PEG-PEI/Cer, and we assessed its antitumor efficacy against HCC both in vitro and in vivo in combination with the chemotherapeutic drug sorafenib. We found that NGO-PEG-PEI significantly enhanced the cellular uptake of C6-ceramide. By investigating the mechanism of cellular delivery, we determined that the internalization of NGO-PEG-PEI/Cer progressed primarily via a clathrin-mediated mechanism. The combination of NGO-PEG-PEI/Cer and sorafenib exhibited synergy between these two drugs. Further work revealed that NGO-PEG-PEI/Cer may play a role in subverting multidrug resistance (MDR) in HCC cells by inactivating MDR and Akt signaling. NGO-PEG-PEI/Cer also significantly inhibited tumor growth and improved survival times in vivo, and the synergetic effect of NGO-PEG-PEI/Cer combined with sorafenib was also observed in drug-resistant HCC xenografts. In conclusion, our NGO-PEG-PEI nanocomposite is an effective nano-platform for loading C6-ceramide for therapeutic use in treating HCC, exhibiting high cancer cell killing potency in this tumor model. The NGO-PEG-PEI/Cer/sorafenib combination additionally represents a promising potential therapeutic strategy for the treatment of drug-resistant HCC.
Collapse
Affiliation(s)
- Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiao-Yi Chen
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Yuan-Yuan Zhao
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China.,Clinical Research Institute, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
33
|
Cho SM, Lee HK, Liu Q, Wang MW, Kwon HJ. A Guanidine-Based Synthetic Compound Suppresses Angiogenesis via Inhibition of Acid Ceramidase. ACS Chem Biol 2019; 14:11-19. [PMID: 30507149 DOI: 10.1021/acschembio.8b00558] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Angiogenesis generates new blood vessels from pre-existing vessels. Tumors induce the formation of new blood vessels to ensure sufficient oxygen and nutrients for their growth. Normally, angiogenesis is induced by various pro-angiogenesis factors, including vascular endothelial growth factor (VEGF). Inhibition of VEGF is a promising approach to cancer treatment. A guanidine-based synthetic compound, E2, was identified as a potent hit from 68 guanidine-based derivatives by screening for angiogenesis inhibitors showing antiproliferative activity in human umbilical vein endothelial cells (HUVECs). To explore the mode of action of E2, target proteins were investigated using phage display biopanning, and acid ceramidase 1 (ASAH1) was identified as an E2-binding protein. Drug affinity responsive target stability (DARTS) and ASAH1 activity assays revealed the direct binding of E2 to ASAH1. Moreover, siRNA knockdown of ASAH1 demonstrated its role as an angiogenesis factor. Consequently, E2 inhibited chemoinvasion and tube formation of HUVECs in a dose-dependent manner. E2 also potently suppressed neo-vascularization of chorioallantoic membranes in vivo. Collectively, these data suggest that E2 is a novel angiogenesis inhibitor and ASAH1 is proposed to be a new antiangiogenesis target.
Collapse
Affiliation(s)
- Sung Min Cho
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Hyung Keun Lee
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| | - Qing Liu
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Ming-Wei Wang
- The National Center for Drug Screening and CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ho Jeong Kwon
- Chemical Genomics Global Research Laboratory, Department of Biotechnology, College of Life Science & Biotechnology, Yonsei University, Seoul 120-749, Republic of Korea
| |
Collapse
|
34
|
Bai A, Bielawska A, Rahmaniyan M, Kraveka JM, Bielawski J, Hannun YA. Dose dependent actions of LCL521 on acid ceramidase and key sphingolipid metabolites. Bioorg Med Chem 2018; 26:6067-6075. [PMID: 30448190 PMCID: PMC6323005 DOI: 10.1016/j.bmc.2018.11.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 10/30/2018] [Accepted: 11/09/2018] [Indexed: 12/17/2022]
Abstract
The function of acid ceramidase (ACDase), whose congenital deficiency leads to Farber disease, has been recognized to be vital to tumor cell biology, and inhibition of its activity may be beneficial in cancer therapy. Therefore, manipulation of the activity of this enzyme may have significant effect, especially on cancer cells. LCL521, Di-DMG-B13, is a lysosomotropic inhibitor of ACDase. Here we define complexities in the actions of LCL521 on ACDase. Systematic studies in MCF7 cells showed dose and time divergent action of LCL521 on ACDase protein expression and sphingolipid levels. Low dose of LCL521 (1 µM) effectively inhibited ACDase in cells, but the effects were transient. A higher dose of LCL521 (10 µM) caused a profound decrease of sphingosine and increase of ceramide, but additionally affected the processing and regeneration of the ACDase protein, with biphasic and reversible effects on the expression of ACDase, which paralleled the long term changes of cellular sphingosine and ceramide. Finally, the higher concentrations of LCL521 also inhibited Dihydroceramide desaturase (DES-1). In summary, LCL521 exhibits significant effects on ACDase in a dose and time dependent manner, but dose range and treatment time need to be paid attention to specify its future exploration on ACDase targeted cancer treatment.
Collapse
Affiliation(s)
- Aiping Bai
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 294255, United States; Lipidomics Shared Resources, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, United States
| | - Alicja Bielawska
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 294255, United States; Lipidomics Shared Resources, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, United States
| | - Mehrdad Rahmaniyan
- Department of Pediatrics-Hematology/Oncology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 294255, United States
| | - Jacqueline M Kraveka
- Department of Pediatrics-Hematology/Oncology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 294255, United States
| | - Jacek Bielawski
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 294255, United States; Lipidomics Shared Resources, Medical University of South Carolina, 173 Ashley Ave., Charleston, SC 29425, United States
| | - Yusuf A Hannun
- Departments of Medicine, Biochemistry and Cell Biology, and Pharmacology and the Stony Brook Cancer Center at Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
35
|
Mohamed ZH, Rhein C, Saied EM, Kornhuber J, Arenz C. FRET probes for measuring sphingolipid metabolizing enzyme activity. Chem Phys Lipids 2018; 216:152-161. [DOI: 10.1016/j.chemphyslip.2018.09.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 09/21/2018] [Indexed: 12/31/2022]
|
36
|
Click chemistry in sphingolipid research. Chem Phys Lipids 2018; 215:71-83. [DOI: 10.1016/j.chemphyslip.2018.07.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 01/17/2023]
|
37
|
Voelkel-Johnson C, Norris JS, White-Gilbertson S. Interdiction of Sphingolipid Metabolism Revisited: Focus on Prostate Cancer. Adv Cancer Res 2018; 140:265-293. [PMID: 30060812 PMCID: PMC6460930 DOI: 10.1016/bs.acr.2018.04.014] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Sphingolipid metabolism is known to play a role in cell death, survival, and therapy resistance in cancer. Sphingolipids, particularly dihydroceramide and ceramide, are associated with antiproliferative or cell death responses, respectively, and are central to effective cancer therapy. Within the last decade, strides have been made in elucidating many intricacies of sphingolipid metabolism. New information has emerged on the mechanisms by which sphingolipid metabolism is dysregulated during malignancy and how cancer cells survive and/or escape therapeutic interventions. This chapter focuses on three main themes: (1) sphingolipid enzymes that are dysregulated in cancer, particularly in prostate cancer; (2) inhibitors of sphingolipid metabolism that antagonize prosurvival responses; and (3) sphingolipid-driven escape mechanisms that allow cancer cells to evade therapies. We explore clinical and preclinical approaches to interdict sphingolipid metabolism and provide a rationale for combining strategies to drive the generation of antiproliferative ceramides with prevention of ceramide clearance.
Collapse
Affiliation(s)
- Christina Voelkel-Johnson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - James S. Norris
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Shai White-Gilbertson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| |
Collapse
|
38
|
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
|
39
|
Li YH, Liu HT, Xu J, Xing AY, Zhang J, Wang YW, Yin G, Gao P. The value of detection of S100A8 and ASAH1 in predicting the chemotherapy response for breast cancer patients. Hum Pathol 2018; 74:156-163. [PMID: 29320752 DOI: 10.1016/j.humpath.2018.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/28/2017] [Accepted: 01/02/2018] [Indexed: 12/14/2022]
Abstract
Chemotherapy plays an important role in the treatment of breast cancer. However, chemoresistance remains the main obstacle for effective treatment, leading to poor prognosis. This study aims to investigate the value of detection of S100A8 and ASAH1 in predicting the chemotherapy response. Miller and Payne grades were used to assess the chemotherapy response in breast cancers. The expression of S100A8 and ASAH1, as well as ER, PR, HER2 and Ki-67 were assessed by immunohistochemical staining in 120 cases of non-special type invasive ductal carcinoma (IDC-NOS). S100A8 expression was higher in chemosensitive breast cancers than chemoresistant ones. Moreover, S100A8 expression was significantly correlated with the molecular subtypes and histological grade, but not with patients' age, tumor size and lymph nodes status. However, there was no significant difference in ASAH1 expression between chemoresistant and chemosensitive group. We also found that higher ASAH1 expression was correlated with positive lymph nodes status, but not with age, tumor size, molecular subtypes and histological grade. S100A8 was valuable in predicting chemotherapy response in breast cancers. The expression of ASAH1 was associated significantly with lymph nodes metastasis, indicating that ASAH1 may serve as a biomarker to predict patients' lymph nodes status in breast cancers.
Collapse
Affiliation(s)
- Yu-Hong Li
- Department of Pathology, The People's Hospital of Liaocheng, Liaocheng, P.R. China; Department of Surgery, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Hai-Ting Liu
- Department of Pathology, QiLu Hospital, Shandong University, Jinan, P.R. China
| | - Jing Xu
- Department of Pathology, The People's Hospital of Liaocheng, Liaocheng, P.R. China; Department of Pathology, Qingdao Central Hospital, Qingdao, P.R. China
| | - Ai-Yan Xing
- Department of Pathology, QiLu Hospital, Shandong University, Jinan, P.R. China
| | - Jie Zhang
- Department of Surgery, Qilu Hospital, Shandong University, Jinan, P.R. China
| | - Ya-Wen Wang
- Department of Pathology, QiLu Hospital, Shandong University, Jinan, P.R. China
| | - Gang Yin
- Department of Surgery, Qilu Hospital, Shandong University, Jinan, P.R. China.
| | - Peng Gao
- Department of Pathology, QiLu Hospital, Shandong University, Jinan, P.R. China.
| |
Collapse
|
40
|
Marín de Mas I, Aguilar E, Zodda E, Balcells C, Marin S, Dallmann G, Thomson TM, Papp B, Cascante M. Model-driven discovery of long-chain fatty acid metabolic reprogramming in heterogeneous prostate cancer cells. PLoS Comput Biol 2018; 14:e1005914. [PMID: 29293497 PMCID: PMC5766231 DOI: 10.1371/journal.pcbi.1005914] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Revised: 01/12/2018] [Accepted: 12/01/2017] [Indexed: 12/17/2022] Open
Abstract
Epithelial-mesenchymal-transition promotes intra-tumoral heterogeneity, by enhancing tumor cell invasiveness and promoting drug resistance. We integrated transcriptomic data for two clonal subpopulations from a prostate cancer cell line (PC-3) into a genome-scale metabolic network model to explore their metabolic differences and potential vulnerabilities. In this dual cell model, PC-3/S cells express Epithelial-mesenchymal-transition markers and display high invasiveness and low metastatic potential, while PC-3/M cells present the opposite phenotype and higher proliferative rate. Model-driven analysis and experimental validations unveiled a marked metabolic reprogramming in long-chain fatty acids metabolism. While PC-3/M cells showed an enhanced entry of long-chain fatty acids into the mitochondria, PC-3/S cells used long-chain fatty acids as precursors of eicosanoid metabolism. We suggest that this metabolic reprogramming endows PC-3/M cells with augmented energy metabolism for fast proliferation and PC-3/S cells with increased eicosanoid production impacting angiogenesis, cell adhesion and invasion. PC-3/S metabolism also promotes the accumulation of docosahexaenoic acid, a long-chain fatty acid with antiproliferative effects. The potential therapeutic significance of our model was supported by a differential sensitivity of PC-3/M cells to etomoxir, an inhibitor of long-chain fatty acid transport to the mitochondria. The coexistence within the same tumor of a variety of subpopulations, featuring different phenotypes (intra-tumoral heterogeneity) represents a challenge for diagnosis, prognosis and targeted therapies. In this work, we have explored the metabolic differences underlying tumor heterogeneity by building cell-type-specific genome-scale metabolic models that integrate transcriptome and metabolome data of two clonal subpopulations derived from the same prostate cancer cell line (PC-3). These subpopulations display either highly proliferative, cancer stem cell (PC-3/M) or highly invasive, epithelial-mesenchymal-transition-like phenotypes (PC-3/S). Our model-driven analysis and experimental validations have unveiled a differential utilization of the long-chain fatty acids pool in both subpopulations. More specifically, our findings show an enhanced entry of long-chain fatty acids into the mitochondria in PC-3/M cells, while in PC-3/S cells, long-chain fatty acids are used as precursors of eicosanoid metabolism. The different utilization of long-chain fatty acids between subpopulations endows PC-3/M cells with a highly proliferative phenotype while enhances PC-3/S invasive phenotype. The present work provides a tool to unveil key metabolic nodes associated with tumor heterogeneity and highlights potential subpopulation-specific targets with important therapeutic implications.
Collapse
Affiliation(s)
- Igor Marín de Mas
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
| | - Esther Aguilar
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
| | - Erika Zodda
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
- Department of Cell Biology, Barcelona Institute for Molecular Biology (IBMB), National Research Council (CSIC), Barcelona, Spain
| | - Cristina Balcells
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
| | - Silvia Marin
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
| | | | - Timothy M. Thomson
- Department of Cell Biology, Barcelona Institute for Molecular Biology (IBMB), National Research Council (CSIC), Barcelona, Spain
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Center of the Hungarian Academy of Sciences, Szeged, Hungary
- * E-mail: (BP); (MC)
| | - Marta Cascante
- Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- Institute of Biomedicine of University of Barcelona (IBUB) and Associated Unit with CSIC, Barcelona, Spain
- * E-mail: (BP); (MC)
| |
Collapse
|
41
|
Kurokawa H, Ito H, Matsui H. Monascus purpureus induced apoptosis on gastric cancer cell by scavenging mitochondrial reactive oxygen species. J Clin Biochem Nutr 2017; 61:189-195. [PMID: 29203960 PMCID: PMC5703783 DOI: 10.3164/jcbn.17-27] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022] Open
Abstract
Monascus purpureus is a red dye derived from yeast rice and has been used as color additives for food in East Asia. Monascus purpureus consists of several bioactive components. Some of these components work as a radical scavenger, thus monascus purpureus would also eliminate reactive oxygen species. Cancer cells maintain the high level of reactive oxygen species than normal cell and are death by imbalance in pro-oxidant/antioxidant homeostasis. In this study, we investigated whether monascus purpureus induced cancer specific cell death by scavenging reactive oxygen species. Compared to normal cell, monascus purpureus had cancer specific cytotoxicity. Monascus purpureus and lovastatin, its component, scavenged free radicals caused by a xanthine/xanthine oxidase system, thus Monascus purpureus is likely to scavenge reactive oxygen species by a synergistic effect between lovastatin and other components. Monascus purpureus also decreased reactive oxygen species derived from mitochondria in cancer cells, and cellular apoptosis was induced via activation of caspase-9. Induction of apoptosis by reduction of reactive oxygen species generation decreased acid ceramidase, and this mechanism could be involved with increasing ceramide accumulation in cells.
Collapse
Affiliation(s)
- Hiromi Kurokawa
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
| | - Hiromu Ito
- Graduate School of Medical and Dental Sciences, Kagoshima University, 8-35-1 Sakuragaoka, Kagoshima 890-8544, Japan
| | - Hirofumi Matsui
- Faculty of Medicine, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8575, Japan
- Kyoto Prefectural University of Medicine, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
42
|
Complete Acid Ceramidase ablation prevents cancer-initiating cell formation in melanoma cells. Sci Rep 2017; 7:7411. [PMID: 28785021 PMCID: PMC5547127 DOI: 10.1038/s41598-017-07606-w] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Accepted: 06/30/2017] [Indexed: 12/22/2022] Open
Abstract
Acid ceramidase (AC) is a lysosomal cysteine hydrolase that catalyzes the conversion of ceramide into fatty acid and sphingosine. This reaction lowers intracellular ceramide levels and concomitantly generates sphingosine used for sphingosine-1-phosphate (S1P) production. Since increases in ceramide and consequent decreases of S1P reduce proliferation of various cancers, AC might offer a new target for anti-tumor therapy. Here we used CrispR-Cas9-mediated gene editing to delete the gene encoding for AC, ASAH1, in human A375 melanoma cells. ASAH1-null clones show significantly greater accumulation of long-chain saturated ceramides that are substrate for AC. As seen with administration of exogenous ceramide, AC ablation blocks cell cycle progression and accelerates senescence. Importantly, ASAH1-null cells also lose the ability to form cancer-initiating cells and to undergo self-renewal, which is suggestive of a key role for AC in maintaining malignancy and self-renewal of invasive melanoma cells. The results suggest that AC inhibitors might find therapeutic use as adjuvant therapy for advanced melanoma.
Collapse
|
43
|
Tan SF, Pearson JM, Feith DJ, Loughran TP. The emergence of acid ceramidase as a therapeutic target for acute myeloid leukemia. Expert Opin Ther Targets 2017; 21:583-590. [PMID: 28434262 DOI: 10.1080/14728222.2017.1322065] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Acute myeloid leukemia (AML) is the most common adult leukemia. Only a fraction of AML patients will survive with existing chemotherapy regimens. Hence, there is an urgent and unmet need to identify novel targets and develop better therapeutics in AML. In the past decade, the field of sphingolipid metabolism has emerged into the forefront of cancer biology due to its importance in cancer cell proliferation and survival. In particular, acid ceramidase (AC) has emerged as a promising therapeutic target due to its role in neutralizing the pro-death effects of ceramide. Areas covered: This review highlights key information about AML biology as well as current knowledge on dysregulated sphingolipid metabolism in cancer and AML. We describe AC function and dysregulation in cancer, followed by a review of studies that report elevated AC in AML and compounds known to inhibit the enzyme. Expert opinion: AML has a great need for new drug targets and better therapeutic agents. The finding of elevated AC in AML supports the concept that this enzyme represents a novel and realistic therapeutic target for this common leukemia. More effort is needed towards developing better AC inhibitors for clinical use and combination treatment with existing AML therapies.
Collapse
Affiliation(s)
- Su-Fern Tan
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA
| | - Jennifer M Pearson
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA
| | - David J Feith
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA.,b University of Virginia Cancer Center , Charlottesville , VA , USA
| | - Thomas P Loughran
- a Department of Medicine , University of Virginia , Charlottesville , VA , USA.,b University of Virginia Cancer Center , Charlottesville , VA , USA
| |
Collapse
|
44
|
Ma YY, Mou XZ, Ding YH, Zou H, Huang DS. Delivery systems of ceramide in targeted cancer therapy: ceramide alone or in combination with other anti-tumor agents. Expert Opin Drug Deliv 2016; 13:1397-406. [PMID: 27168034 DOI: 10.1080/17425247.2016.1188803] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Ying-Yu Ma
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Medical School and Jiangsu Laboratory of Molecular Medicine, Nanjing University, Nanjing, China
| | - Xiao-Zhou Mou
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Ya-Hui Ding
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People’s Hospital, Hangzhou, China
| | - Dong-Sheng Huang
- Clinical Research Institute, Zhejiang Provincial People’s Hospital, Hangzhou, China
- Department of Hepatobiliary Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, China
| |
Collapse
|
45
|
Thangavel S, Yoshitomi T, Sakharkar MK, Nagasaki Y. Redox nanoparticle increases the chemotherapeutic efficiency of pioglitazone and suppresses its toxic side effects. Biomaterials 2016; 99:109-23. [PMID: 27235996 DOI: 10.1016/j.biomaterials.2016.05.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Revised: 04/26/2016] [Accepted: 05/02/2016] [Indexed: 02/03/2023]
Abstract
Pioglitazone is a widely used anti-diabetic drug that induces cytotoxicity in cancer cells; however, its clinical use is questioned due to its associated liver toxicity caused by increased oxidative stress. We therefore employed nitroxide-radical containing nanoparticle, termed redox nanoparticle (RNP(N)) which is an effective scavenger of reactive oxygen species (ROS) as a drug carrier. RNP(N) encapsulation increased pioglitazone solubility, thus increasing cellular uptake of encapsulated pioglitazone which reduced the dose required to induce toxicity in prostate cancer cell lines. Investigation of in vitro molecular mechanism of pioglitazone revealed that both apoptosis and cell cycle arrest were involved in tumor cell death. In addition, intravenously administered pioglitazone-loaded RNP(N) produced significant tumor volume reduction in vivo due to enhanced permeation and retention effect. Most importantly, oxidative damage caused by pioglitazone in the liver was significantly suppressed by pioglitazone-loaded RNP(N) due to the presence of nitroxide radicals. It is interesting to note that oral administration of encapsulated pioglitazone, and co-administration of RNP(N) and pioglitazone, i.e., no encapsulation of pioglitazone in RNP(N) also significantly contributed to suppression of the liver injury. Therefore, use of RNP(N) either as an adjuvant or as a carrier for drugs with severe side effects is a promising chemotherapeutic strategy.
Collapse
Affiliation(s)
- Sindhu Thangavel
- Department of Materials Sciences, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan
| | - Toru Yoshitomi
- Department of Materials Sciences, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan
| | - Meena Kishore Sakharkar
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Tennodai 1-1-1, Ibaraki 305-8572, Japan
| | - Yukio Nagasaki
- Department of Materials Sciences, Graduate School of Pure and Applied Sciences, University of Tsukuba, Tennodai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Master's School of Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan; Satellite Laboratory, International Center for Materials Nanoarchitectonics (WPI-MANA), National Institute for Materials Science (NIMS), University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki 305-8573, Japan.
| |
Collapse
|
46
|
Quantitative proteomic analysis of anticancer drug RH1 resistance in liver carcinoma. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2016; 1864:219-32. [DOI: 10.1016/j.bbapap.2015.11.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/26/2015] [Accepted: 11/16/2015] [Indexed: 01/18/2023]
|
47
|
Roh JL, Park JY, Kim EH, Jang HJ. Targeting acid ceramidase sensitises head and neck cancer to cisplatin. Eur J Cancer 2015; 52:163-72. [PMID: 26687835 DOI: 10.1016/j.ejca.2015.10.056] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 10/20/2015] [Accepted: 10/23/2015] [Indexed: 10/22/2022]
Abstract
BACKGROUND Acid ceramidase (AC), a key enzyme in ceramide metabolism, plays a role in cancer progression and resistance to therapy. However, the role of AC in head and neck cancer (HNC) has not been addressed. Here, we investigate the effect of AC inhibition on the response to cisplatin-based chemotherapy for HNC. METHODS AC protein and messenger RNA (mRNA) expression were examined in primary tumours and paired normal tissues, and in HNC cell lines. The effects of genetic and pharmacological AC inhibition using small hairpin RNA (shRNA) and N-oleoyl-ethanolamine (NOE), alone and in combination with cisplatin, were assessed in human HNC cells by measuring cell viability, cell cycle progression, apoptosis, mRNA, and protein expression, and in preclinical tumour xenograft mouse models. FINDINGS AC overexpression was observed in four of six primary tumour tissues and six of nine HNC cell lines. Cisplatin sensitivity was significantly decreased by AC overexpression and significantly increased by AC downregulation in HNC cells (P<0.01). NOE or AC shRNA-mediated AC inhibition enhanced cisplatin-induced HNC cell death by increasing ceramide production and activating pro-apoptotic proteins, and these effects were abrogated by PUMA small interfering RNA transfection. AC inhibition promoted cisplatin-induced apoptosis of HNC cells in vitro and in vivo. INTERPRETATIONS AC overexpression is associated with cisplatin sensitivity, suggesting its potential role as a chemotherapeutic target for HNC. Genetic or pharmacological AC inhibition promotes cisplatin cytotoxicity in HNC cells.
Collapse
Affiliation(s)
- Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | - Jin Young Park
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye Jin Jang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
48
|
Bach A, Pizzirani D, Realini N, Vozella V, Russo D, Penna I, Melzig L, Scarpelli R, Piomelli D. Benzoxazolone Carboxamides as Potent Acid Ceramidase Inhibitors: Synthesis and Structure-Activity Relationship (SAR) Studies. J Med Chem 2015; 58:9258-72. [PMID: 26560855 DOI: 10.1021/acs.jmedchem.5b01188] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Ceramides are lipid-derived intracellular messengers involved in the control of senescence, inflammation, and apoptosis. The cysteine amidase, acid ceramidase (AC), hydrolyzes these substances into sphingosine and fatty acid and, by doing so, regulates their signaling activity. AC inhibitors may be useful in the treatment of pathological conditions, such as cancer, in which ceramide levels are abnormally reduced. Here, we present a systematic SAR investigation of the benzoxazolone carboxamides, a recently described class of AC inhibitors that display high potency and systemic activity in mice. We examined a diverse series of substitutions on both benzoxazolone ring and carboxamide side chain. Several modifications enhanced potency and stability, and one key compound with a balanced activity-stability profile (14) was found to inhibit AC activity in mouse lungs and cerebral cortex after systemic administration. The results expand our arsenal of AC inhibitors, thereby facilitating the use of these compounds as pharmacological tools and their potential development as drug leads.
Collapse
Affiliation(s)
- Anders Bach
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Daniela Pizzirani
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Natalia Realini
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Valentina Vozella
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Debora Russo
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Ilaria Penna
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Laurin Melzig
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Rita Scarpelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy
| | - Daniele Piomelli
- Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia , Via Morego 30, I-16163 Genova, Italy.,Departments of Anatomy and Neurobiology, Biological Chemistry, and Pharmacology, University of California , Irvine, California 92697-4625, United States
| |
Collapse
|
49
|
Realini N, Palese F, Pizzirani D, Pontis S, Basit A, Bach A, Ganesan A, Piomelli D. Acid Ceramidase in Melanoma: EXPRESSION, LOCALIZATION, AND EFFECTS OF PHARMACOLOGICAL INHIBITION. J Biol Chem 2015; 291:2422-34. [PMID: 26553872 DOI: 10.1074/jbc.m115.666909] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Indexed: 11/06/2022] Open
Abstract
Acid ceramidase (AC) is a lysosomal cysteine amidase that controls sphingolipid signaling by lowering the levels of ceramides and concomitantly increasing those of sphingosine and its bioactive metabolite, sphingosine 1-phosphate. In the present study, we evaluated the role of AC-regulated sphingolipid signaling in melanoma. We found that AC expression is markedly elevated in normal human melanocytes and proliferative melanoma cell lines, compared with other skin cells (keratinocytes and fibroblasts) and non-melanoma cancer cells. High AC expression was also observed in biopsies from human subjects with Stage II melanoma. Immunofluorescence studies revealed that the subcellular localization of AC differs between melanocytes (where it is found in both cytosol and nucleus) and melanoma cells (where it is primarily localized to cytosol). In addition to having high AC levels, melanoma cells generate lower amounts of ceramides than normal melanocytes do. This down-regulation in ceramide production appears to result from suppression of the de novo biosynthesis pathway. To test whether AC might contribute to melanoma cell proliferation, we blocked AC activity using a new potent (IC50 = 12 nM) and stable inhibitor. AC inhibition increased cellular ceramide levels, decreased sphingosine 1-phosphate levels, and acted synergistically with several, albeit not all, antitumoral agents. The results suggest that AC-controlled sphingolipid metabolism may play an important role in the control of melanoma proliferation.
Collapse
Affiliation(s)
- Natalia Realini
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Francesca Palese
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Daniela Pizzirani
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Silvia Pontis
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Abdul Basit
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy
| | - Anders Bach
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy, the Department of Drug Design and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen 2100, Denmark, and
| | | | - Daniele Piomelli
- From the Department of Drug Discovery and Development, Fondazione Istituto Italiano di Tecnologia, Genova 16163, Italy, Anatomy and Neurobiology, University of California, Irvine, California 92617
| |
Collapse
|
50
|
Recognition Code of ZNF191(243-368) and Its Interaction with DNA. Bioinorg Chem Appl 2015; 2015:416751. [PMID: 26457075 PMCID: PMC4592708 DOI: 10.1155/2015/416751] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 09/02/2015] [Indexed: 02/05/2023] Open
Abstract
ZNF191(243-368) is the C-terminal region of ZNF191 which contains a putative DNA-binding domain of four Cys2His2 zinc finger motifs. In this study, an expression vector of a fusion protein of ZNF191(243-368) with glutathione-S-transferase (GST) was constructed and transformed into Escherichia coli BL21. The fusion protein GST-ZNF191(243-368) was expressed using this vector to investigate the protein-DNA binding reaction through an affinity selection strategy on the basis of the binding quality of the zinc finger domain. Results showed that ZNF191(243-368) can selectively bind with sequences and react with genes which contain an AGGG core. However, the recognition mechanism of Cys2His2 zinc finger proteins to DNA warrants further investigation.
Collapse
|