1
|
Huang L, Xue R, Zhu M, Xu S, Luo Y, Qin C, Yang C, Jia L, Tang K, Tan Q. Triptolide's impact on ACER1 signaling: Inducing autophagy for triple-negative breast cancer suppression. Pathol Res Pract 2025; 266:155823. [PMID: 39879681 DOI: 10.1016/j.prp.2025.155823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 12/24/2024] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
Given the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (Her-2) in triple-negative breast cancer (TNBC) cells, the efficacy of targeted therapies is limited. In this study, we uncovered that triptolide (TP) effectively suppresses the migration and invasiveness of MDA-MB-231 cells by activating autophagic pathways. Western blotting analysis revealed that TP significantly reduced the expression levels of p62 protein, while simultaneously markedly increasing the expression levels of LC3B-II, BNIP3, BNIP3L, ATG5, and ULK1 proteins, strongly suggesting an enhancement of autophagic activity in the cells. Based on PCR array screening, we identified the ACER1 gene as exhibiting notable expression alterations post-TP treatment. Overexpression of ACER1 gene enhanced the TP-induced apoptosis in MDA-MB-231 cells and augmented the regulation of autophagy-related proteins p62 and LC3B-II, leading to an increase in autophagosome numbers and a marked reduction in cellular migration and invasiveness. Conversely, ACER1 gene knockdown reversed these effects. In vivo experiments demonstrated that TP effectively inhibits the growth of MDA-MB-231 xenograft tumors, concurrently upregulating ACER1 and LC3B-II expression in tumor tissues, while p62 protein levels were notably decreased. Hematoxylin and eosin (H&E) staining results indicated no evident toxicity in liver and kidney tissues of BALB/c mice at a TP dose of 0.4 mg/kg. This study, for the first time, elucidates a novel mechanism by which TP inhibits TNBC through an autophagic process mediated by ACER1.
Collapse
Affiliation(s)
- Lingyue Huang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Rui Xue
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Mingfei Zhu
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Siyuan Xu
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Yuqin Luo
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Chuling Qin
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Chang Yang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Lulu Jia
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Ke Tang
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China
| | - Qinyou Tan
- Clinical Pharmacy & Pharmacology Research Institute, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Affiliated Hospital of Guilin Medical University, Guilin 541001, China; Guangxi Health Commission Key Laboratory of Basic Research in Sphingolipid Metabolism Related Diseases, the Affiliated Hospital of Guilin Medical University, Guilin 541001, China; China-USA Lipids in Health and Disease Research Center, Guilin Medical University,Guilin 541001, China; Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
2
|
Zhang J, Xu G, Liu S, Yang M. Cadmium alters the cellular metabolome of human ovarian granulosa cells. Toxicol Appl Pharmacol 2025; 495:117187. [PMID: 39638002 DOI: 10.1016/j.taap.2024.117187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 11/14/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
Cadmium (Cd) is a toxic heavy metal that has been extensively implicated in disordered folliculogenesis, but the mechanisms underlying the ovarian toxicity of Cd remain to be explored fully. Granulosa cells are key players in ovarian follicular development and are the primary cells affected by Cd exposure-induced damage and dysfunction. In this study, we investigated how various levels of exposure of Cd (3 and 10 μM) to human granulosa cells (KGN cells) impacted the metabolism of the KGN cells utilizing a non-targeted metabolomics methodology. In vitro cell experiments revealed that Cd exposure dose-dependently diminished the viability of KGN cells. Metabolomics analysis revealed the presence of 296 (182 elevated and 114 reduced) and 397 (244 elevated and 153 reduced) differentially expressed metabolites after exposure to 3 and 10 μM, respectively. Cd exposure was found to significantly enrich nucleotide metabolism, sphingolipid metabolism, and ABC transporters in both groups. Although amino acid metabolic pathways exhibited significant enrichment across all groups, only glutathione, cysteine, and methionine metabolism were notably enriched in KGN cells exposed to 3 μM Cd, while glutathione and tryptophan metabolism were significantly enriched in the 10 μM Cd exposure cohort. The outcomes of this study provide mechanistic clues for elucidating Cd's cytotoxic impact on granulosa cells, and deepen our understanding of the ovarian toxicity of Cd.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Medical Cell Biology and Genetics, School of Basic Medical Sciences, Key Laboratory of Medical Electrophysiology, Ministry of Education (Collaborative Innovation Center for Prevention of Cardiovascular Diseases), Southwest Medical University, Luzhou, China
| | - Guofeng Xu
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shuang Liu
- Department of Reproductive Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Meng Yang
- Inflammation & Allergic Diseases Research Unit, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Department of Reproductive Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
3
|
Lee J, Mani A, Shin MJ, Krauss RM. Leveraging altered lipid metabolism in treating B cell malignancies. Prog Lipid Res 2024; 95:101288. [PMID: 38964473 PMCID: PMC11347096 DOI: 10.1016/j.plipres.2024.101288] [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: 02/19/2024] [Revised: 06/12/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024]
Abstract
B cell malignancies, comprising over 80 heterogeneous blood cancers, pose significant prognostic challenges due to intricate oncogenic signaling. Emerging evidence emphasizes the pivotal role of disrupted lipid metabolism in the development of these malignancies. Variations in lipid species, such as phospholipids, cholesterol, sphingolipids, and fatty acids, are widespread across B cell malignancies, contributing to uncontrolled cell proliferation and survival. Phospholipids play a crucial role in initial signaling cascades leading to B cell activation and malignant transformation through constitutive B cell receptor (BCR) signaling. Dysregulated cholesterol and sphingolipid homeostasis support lipid raft integrity, crucial for propagating oncogenic signals. Sphingolipids impact malignant B cell stemness, proliferation, and survival, while glycosphingolipids in lipid rafts modulate BCR activation. Additionally, cancer cells enhance fatty acid-related processes to meet heightened metabolic demands. In obese individuals, the obesity-derived lipids and adipokines surrounding adipocytes rewire lipid metabolism in malignant B cells, evading cytotoxic therapies. Genetic drivers such as MYC translocations also intrinsically alter lipid metabolism in malignant B cells. In summary, intrinsic and extrinsic factors converge to reprogram lipid metabolism, fostering aggressive phenotypes in B cell malignancies. Therefore, targeting altered lipid metabolism has translational potential for improving risk stratification and clinical management of diverse B cell malignancy subtypes.
Collapse
Affiliation(s)
- Jaewoong Lee
- School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea; Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul 02841, Republic of Korea; Center of Molecular and Cellular Oncology, Yale University, New Haven, CT 06511, USA.
| | - Arya Mani
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University, New Haven, CT 06511, USA; Department of Genetics, Yale University, New Haven, CT 06511, USA
| | - Min-Jeong Shin
- School of Biosystems and Biomedical Sciences, College of Health Science, Korea University, Seoul 02841, Republic of Korea; Department of Integrated Biomedical and Life Science, Korea University, Seoul 02841, Republic of Korea; Interdisciplinary Program in Precision Public Health, Korea University, Seoul 02841, Republic of Korea
| | - Ronald M Krauss
- Department of Pediatrics and Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
4
|
Jamil M, Cowart LA. Sphingolipids in mitochondria-from function to disease. Front Cell Dev Biol 2023; 11:1302472. [PMID: 38078003 PMCID: PMC10702779 DOI: 10.3389/fcell.2023.1302472] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 11/03/2023] [Indexed: 02/12/2024] Open
Abstract
Sphingolipids are not only structural components of cellular membranes but also play vital roles in cell signaling and modulation of cellular processes. Within mitochondria, sphingolipids exert diverse effects on mitochondrial dynamics, energy metabolism, oxidative stress, and cell death pathways. In this review, we summarize literature addressing the crucial role of sphingolipids in mitochondria, highlighting their impact on mitochondrial dynamics, cellular bioenergetics, and important cell processes including apoptosis and mitophagy.
Collapse
Affiliation(s)
- Maryam Jamil
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA, United States
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Lauren Ashley Cowart
- Department of Biochemistry and Molecular Biology, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
- Richmond Veteran’s Affairs Medical Center, Richmond, VA, United States
| |
Collapse
|
5
|
Velazquez FN, Stith JL, Zhang L, Allam AM, Haley J, Obeid LM, Snider AJ, Hannun YA. Targeting sphingosine kinase 1 in p53KO thymic lymphoma. FASEB J 2023; 37:e23247. [PMID: 37800872 PMCID: PMC11740227 DOI: 10.1096/fj.202301417r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/19/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023]
Abstract
Sphingosine kinase 1 (SK1) is a key sphingolipid enzyme that is upregulated in several types of cancer, including lymphoma which is a heterogenous group of malignancies. Treatment for lymphoma has improved significantly by the introduction of new therapies; however, subtypes with tumor protein P53 (p53) mutations or deletion have poor prognosis, making it critical to explore new therapeutic strategies in this context. SK1 has been proposed as a therapeutic target in different types of cancer; however, the effect of targeting SK1 in cancers with p53 deletion has not been evaluated yet. Previous work from our group suggests that loss of SK1 is a key event in mediating the tumor suppressive effect of p53. Employing both genetic and pharmacological approaches to inhibit SK1 function in Trp53KO mice, we show that targeting SK1 decreases tumor growth of established p53KO thymic lymphoma. Inducible deletion of Sphk1 or its pharmacological inhibition drive increased cell death in tumors which is accompanied by selective accumulation of sphingosine levels. These results demonstrate the relevance of SK1 in the growth and maintenance of lymphoma in the absence of p53 function, positioning this enzyme as a potential therapeutic target for the treatment of tumors that lack functional p53.
Collapse
Affiliation(s)
- Fabiola N. Velazquez
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Jeffrey L. Stith
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Leiqing Zhang
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Amira M. Allam
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - John Haley
- Biological Mass Spectrometry Center, Stony Brook Medicine, Stony Brook University, Stony Brook, New York, USA
| | - Lina M. Obeid
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| | - Ashley J. Snider
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
- School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, and University of Arizona Cancer Center, University of Arizona, Tucson, Arizona, USA
| | - Yusuf A. Hannun
- Department of Medicine, Stony Brook University, Stony Brook, New York, USA
- Cancer Center, Stony Brook University, Stony Brook, New York, USA
| |
Collapse
|
6
|
Carbone D, Gallo C, Nuzzo G, Barra G, Dell'Isola M, Affuso M, Follero O, Albiani F, Sansone C, Manzo E, d'Ippolito G, Fontana A. Marine natural product lepadin A as a novel inducer of immunogenic cell death via CD91-dependent pathway. NATURAL PRODUCTS AND BIOPROSPECTING 2023; 13:34. [PMID: 37779162 PMCID: PMC10542626 DOI: 10.1007/s13659-023-00401-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/17/2023] [Indexed: 10/03/2023]
Abstract
Immunogenic Cell Death (ICD) represents a mechanism of enhancing T cell-driven response against tumor cells. The process is enabled by release of damage-associated molecular patterns (DAMPs) and cytokines by dying cells. Based on molecular studies and clinical marker assessment, ICD can be a new target for cancer chemotherapy hitherto restricted to a few conventional anticancer drugs. In view of the development of small molecules in targeted cancer therapy, we reported the preliminary evidence on the role of the natural product lepadin A (1) as a novel ICD inducer. Here we describe the ICD mechanism of lepadin A (1) by proving the translocation of the protein calreticulin (CRT) to the plasma membrane of human A2058 melanoma cells. CRT exposure is an ICD marker in clinical studies and was associated with the activation of the intrinsic apoptotic pathway in A2058 cells with lepadin A (1). After the treatment, the tumour cells acquired the ability to activate dendritic cells (DCs) with cytokine release and costimulatory molecule expression that is consistent with a phenotypic profile committed to priming T lymphocytes via a CD91-dependent mechanism. The effect of lepadin A (1) was dose-dependent and comparable to the response of the chemotherapy drug doxorubicin (2), a well-established ICD inducer.
Collapse
Affiliation(s)
- Dalila Carbone
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Carmela Gallo
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy.
| | - Genoveffa Nuzzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Giusi Barra
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mario Dell'Isola
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Mario Affuso
- Department of Biology, University of Naples "Federico II", Via Cupa Nuova Cinthia 21, 80126, Naples, Italy
| | - Olimpia Follero
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Federica Albiani
- Department of Biology, University of Naples "Federico II", Via Cupa Nuova Cinthia 21, 80126, Naples, Italy
| | - Clementina Sansone
- Stazione Zoologica Anton Dohrn, Istituto Nazionale di Biologia, Ecologia e Biotecnologie Marine, University of Naples "Federico II", Villa Comunale, 80121, Naples, Italy
| | - Emiliano Manzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Giuliana d'Ippolito
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
| | - Angelo Fontana
- Institute of Biomolecular Chemistry, Consiglio Nazionale Delle Ricerche, Via Campi Flegrei 34, Pozzuoli, 80078, Naples, Italy
- Department of Biology, University of Naples "Federico II", Via Cupa Nuova Cinthia 21, 80126, Naples, Italy
| |
Collapse
|
7
|
Kieda J, Appak-Baskoy S, Jeyhani M, Navi M, Chan KWY, Tsai SSH. Microfluidically-generated Encapsulated Spheroids (μ-GELS): An All-Aqueous Droplet Microfluidics Platform for Multicellular Spheroids Generation. ACS Biomater Sci Eng 2023; 9:1043-1052. [PMID: 36626575 DOI: 10.1021/acsbiomaterials.2c00963] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Spheroids are three-dimensional clusters of cells that serve as in vitro tumor models to recapitulate in vivo morphology. A limitation of many existing on-chip platforms for spheroid formation is the use of cytotoxic organic solvents as the continuous phase in droplet generation processes. All-aqueous methods do not contain cytotoxic organic solvents but have so far been unable to achieve complete hydrogel gelation on chip. Here, we describe an enhanced droplet microfluidic platform that achieves on-chip gelation of all-aqueous hydrogel multicellular spheroids (MCSs). Specifically, we generate dextran-alginate droplets containing MCF-7 breast cancer cells, surrounded by polyethylene glycol, at a flow-focusing junction. Droplets then travel to a second flow-focusing junction where they interact with calcium chloride and gel on chip to form hydrogel MCSs. On-chip gelation of the MCSs is possible here because of an embedded capillary at the second junction that delays the droplet gelation, which prevents channel clogging problems that would otherwise exist. In drug-free experiments, we demonstrate that MCSs remain viable for 6 days. We also confirm the applicability of this system for cancer drug testing by observing that dose-dependent cell death is achievable using doxorubicin.
Collapse
Affiliation(s)
- Jennifer Kieda
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Sila Appak-Baskoy
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Chemistry and Biology, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Morteza Jeyhani
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| | - Maryam Navi
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Katherine W Y Chan
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada
| | - Scott S H Tsai
- Graduate Program in Biomedical Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada.,Keenan Research Centre for Biomedical Science, St. Michael's Hospital, TorontoM5B 2K3, Canada.,Institute for Biomedical Engineering, Science, and Technology (iBEST) - A partnership between Toronto Metropolitan University and St. Michael's Hospital, TorontoM5B 1W8, Canada.,Department of Mechanical and Industrial Engineering, Toronto Metropolitan University, TorontoM5B 2K3, Canada
| |
Collapse
|
8
|
Wang X, Zhang C, Madji R, Voros C, Mazères S, Bijani C, Deraeve C, Cuvillier O, Gornitzka H, Maddelein ML, Hemmert C. N-Heterocyclic Carbene-Iridium Complexes as Photosensitizers for In Vitro Photodynamic Therapy to Trigger Non-Apoptotic Cell Death in Cancer Cells. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020691. [PMID: 36677751 PMCID: PMC9861386 DOI: 10.3390/molecules28020691] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/13/2023]
Abstract
A series of seven novel iridium complexes were synthetized and characterized as potential photosensitizers for photodynamic therapy (PDT) applications. Among them, four complexes were evaluated in vitro for their anti-proliferative activity with and without irradiation on a panel of five cancer cell lines, namely PC-3 (prostate cancer), T24 (bladder cancer), MCF7 (breast cancer), A549 (lung cancer) and HeLa (cervix cancer), and two non-cancerous cell models (NIH-3T3 fibroblasts and MC3T3 osteoblasts). After irradiation at 458 nm, all tested complexes showed a strong selectivity against cancer cells, with a selectivity index (SI) ranging from 8 to 34 compared with non-cancerous cells. The cytotoxic effect of all these complexes was found to be independent of the anti-apoptotic protein Bcl-xL. The compound exhibiting the best selectivity, complex 4a, was selected for further investigations. Complex 4a was mainly localized in the mitochondria. We found that the loss of cell viability and the decrease in ATP and GSH content induced by complex 4a were independent of both Bcl-xL and caspase activation, leading to a non-apoptotic cell death. By counteracting the intrinsic or acquired resistance to apoptosis associated with cancer, complex 4a could be an interesting therapeutic alternative to be studied in preclinical models.
Collapse
Affiliation(s)
- Xing Wang
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Chen Zhang
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Ryma Madji
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Camille Voros
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Serge Mazères
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Christian Bijani
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Céline Deraeve
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
| | - Olivier Cuvillier
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Heinz Gornitzka
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Marie-Lise Maddelein
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Institut de Pharmacologie et de Biologie Structurale (IPBS), Université de Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| | - Catherine Hemmert
- Coordination Chemistry Laboratory of the National Centre for Scientific Research (LCC-CNRS), University of Toulouse, CNRS, Université Toulouse III–Paul Sabatier (UPS), 31077 Toulouse, France
- Correspondence: (O.C.); (H.G.); (M.-L.M.); (C.H.)
| |
Collapse
|
9
|
S-phase arrest and apoptosis in human breast adenocarcinoma MCF-7 cells via mitochondrial dependent pathway induced by tricyclohexylphosphine gold (I) n-mercaptobenzoate complexes. Life Sci 2022; 311:121161. [DOI: 10.1016/j.lfs.2022.121161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022]
|
10
|
Linzer RW, Guida DL, Aminov J, Snider JM, Khalife G, Buyukbayraktar AB, Alhaddad C, Resnick AE, Wang P, Pan CH, Allopenna JJ, Clarke CJ. Dihydroceramide desaturase 1 (DES1) promotes anchorage-independent survival downstream of HER2-driven glucose uptake and metabolism. FASEB J 2022; 36:e22558. [PMID: 36165222 PMCID: PMC9597949 DOI: 10.1096/fj.202200748r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 08/03/2022] [Accepted: 09/07/2022] [Indexed: 11/11/2022]
Abstract
Oncogenic reprogramming of cellular metabolism is a hallmark of many cancers, but our mechanistic understanding of how such dysregulation is linked to tumor behavior remains poor. In this study, we have identified dihydroceramide desaturase (DES1)-which catalyzes the last step in de novo sphingolipid synthesis-as necessary for the acquisition of anchorage-independent survival (AIS), a key cancer enabling biology, and establish DES1 as a downstream effector of HER2-driven glucose uptake and metabolism. We further show that DES1 is sufficient to drive AIS and in vitro tumorigenicity and that increased DES1 levels-found in a third of HER2+ breast cancers-are associated with worse survival outcomes. Taken together, our findings reveal a novel pro-tumor role for DES1 as a transducer of HER2-driven glucose metabolic signals and provide evidence that targeting DES1 is an effective approach for overcoming AIS. Results further suggest that DES1 may have utility as a biomarker of aggressive and metastasis-prone HER2+ breast cancer.
Collapse
Affiliation(s)
- Ryan W Linzer
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Danielle L Guida
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Jonathan Aminov
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Justin M Snider
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Gabrielle Khalife
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - A Burak Buyukbayraktar
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Charbel Alhaddad
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Andrew E Resnick
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Pule Wang
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Chun-Hao Pan
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Janet J Allopenna
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Christopher J Clarke
- Department of Medicine and the Cancer Center, Stony Brook University, Stony Brook, NY, USA
| |
Collapse
|
11
|
Porubský M, Řezníčková E, Křupková S, Kryštof V, Hlaváč J. Development of fluorescent dual-FRET probe for simultaneous detection of caspase-8 and caspase-9 activities and their relative quantification. Bioorg Chem 2022; 129:106151. [DOI: 10.1016/j.bioorg.2022.106151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 08/24/2022] [Accepted: 09/08/2022] [Indexed: 11/27/2022]
|
12
|
Park Y, Yoon HJ, Lee SE, Lee LP. Multifunctional Cellular Targeting, Molecular Delivery, and Imaging by Integrated Mesoporous-Silica with Optical Nanocrescent Antenna: MONA. ACS NANO 2022; 16:2013-2023. [PMID: 35041396 DOI: 10.1021/acsnano.1c07015] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Multifunctional nanoprobes have attracted significant attention in a wide range of disciplines such as nanomedicine, precision medicine, and cancer diagnosis and treatment. However, integrating multifunctional ability in a nanoscale structure to precisely target, image, and deliver with cellular spatial/temporal resolution is still challenging in cellulo applications. This is because the development of such high-precision resolution needs to be carried out without labeling, photobleaching, and structurally segregating live cells. In this study, we present an integrated nanostructure of a mesoporous-silica nanosphere with an optical nanocrescent antenna (MONA) for multifunctional cellular targeting, drug delivery, and molecular imaging with spatiotemporal resolution. MONA comprises a systematically constructed Au nanocrescent (AuNC) antenna as a nanosensor and optical switch on a mesoporous-silica nanosphere as a cargo to molecular delivery. MONA made of antiepithelial cell adhesion molecules (anti-EpCAM)-conjugated AuNC facilitates the specific targeting of breast cancer cells, resulting in a highly focused photothermal gradient that functions as a molecular emitter. This light-driven molecular, doxorubicin (DOX) delivery function allows rapid apoptosis of breast cancer cells. Since MONA permits the tracking of quantum biological electron-transfer processes, in addition to its role as an on-demand optical switch, it enables the monitoring of the dynamic behavior of cellular cytochrome c pivoting cell apoptosis in response to the DOX delivery. Owing to the integrated functions of molecular actuation and direct sensing at the precisely targeted spot afforded by MONA, we anticipate that this multifunctional optical nanoantenna structure will have an impact in the fields of nanomedicine, cancer theranostics, and basic life sciences.
Collapse
Affiliation(s)
- Younggeun Park
- Department of Bioengineering and Biomolecular Nanotechnology Center, Berkeley Sensor and Actuator Center and University of California, Berkeley, California 94720, United States
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Hyeun Joong Yoon
- Department of Biomedical Engineering, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Somin Eunice Lee
- Department of Electrical & Computer Engineering, Department of Biomedical Engineering, Applied Physics, Biointerfaces Institute, Macromolecular Science and Engineering, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Luke P Lee
- Department of Bioengineering and Biomolecular Nanotechnology Center, Berkeley Sensor and Actuator Center and University of California, Berkeley, California 94720, United States
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, California 94720, United States
- Harvard Institute of Medicine, Harvard Medical School, Department of Medicine, Brigham and Women's Hospital, Harvard University, Boston, Massachusetts 02115 United States
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| |
Collapse
|
13
|
Gomez-Brouchet A, Illac C, Ledoux A, Fortin PY, de Barros S, Vabre C, Despas F, Peries S, Casaroli C, Bouvier C, Aubert S, de Pinieux G, Larousserie F, Galmiche L, Talmont F, Pitson S, Maddelein ML, Cuvillier O. Sphingosine Kinase-1 Is Overexpressed and Correlates with Hypoxia in Osteosarcoma: Relationship with Clinicopathological Parameters. Cancers (Basel) 2022; 14:cancers14030499. [PMID: 35158767 PMCID: PMC8833796 DOI: 10.3390/cancers14030499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/14/2022] [Accepted: 01/16/2022] [Indexed: 11/16/2022] Open
Abstract
The Sphingosine kinase-1/Sphingosine 1-Phosphate (SphK1/S1P) signaling pathway is overexpressed in various cancers, and is instrumental for the adaptation to hypoxia in a number of solid tumor models, but no data are available in osteosarcoma. Here we report that SphK1 and the S1P1 receptor are involved in HIF-1α accumulation in hypoxic osteosarcoma cells. FTY720 (Fingolimod), which targets SphK1 and S1P1, prevented HIF-1α accumulation, and also inhibited cell proliferation in both normoxia and hypoxia unlike conventional chemotherapy. In human biopsies, a significant increase of SphK1 activity was observed in cancer compared with normal bones. In all sets of TMA samples (130 cases of osteosarcoma), immunohistochemical analysis showed the hypoxic marker GLUT-1, SphK1 and S1P1 were expressed in tumors. SphK1 correlated with the GLUT-1 suggesting that SphK1 is overexpressed and correlates with intratumoral hypoxia. No correlation was found between GLUT-1 or SphK1 and response to chemotherapy, but a statistical difference was found with increased S1P1 expression in patients with poor response in long bone osteosarcomas. Importantly, multivariate analyses showed that GLUT-1 was associated with an increased risk of death in flat bone, whereas SphK1 and S1P1 were associated with an increased risk of death in long bones.
Collapse
Affiliation(s)
- Anne Gomez-Brouchet
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Département d’Anatomie et Cytologie Pathologies, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France
- Cancer Biobank, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France;
- Correspondence: (A.G.-B.); (O.C.)
| | - Claire Illac
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Département d’Anatomie et Cytologie Pathologies, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France
| | - Adeline Ledoux
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Pierre-Yves Fortin
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Sandra de Barros
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Clémentine Vabre
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Fabien Despas
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Sophie Peries
- Service de Pharmacologie Clinique, Hôpitaux de Toulouse, 31300 Toulouse, France; (S.d.B.); (C.V.); (F.D.); (S.P.)
| | - Christelle Casaroli
- Cancer Biobank, Institut Universitaire du Cancer de Toulouse–Oncopôle (IUCT-O), 31059 Toulouse, France;
| | - Corinne Bouvier
- Department of Pathology, CHU la Timone, 13005 Marseille, France;
| | | | | | - Frédérique Larousserie
- Department of Pathology, AP-HP, Hôpital Cochin, Universiteé Paris Descartes, 75014 Paris, France;
| | - Louise Galmiche
- Centre Hospitalier Universitaire de Nantes Hôtel Dieu, 44000 Nantes, France;
| | - Franck Talmont
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Stuart Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia;
| | - Marie-Lise Maddelein
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
| | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France; (C.I.); (A.L.); (P.-Y.F.); (F.T.); (M.-L.M.)
- Université de Toulouse, UPS, 31400 Toulouse, France
- Correspondence: (A.G.-B.); (O.C.)
| |
Collapse
|
14
|
Nduwumwami AJ, Hengst JA, Yun JK. Sphingosine Kinase Inhibition Enhances Dimerization of Calreticulin at the Cell Surface in Mitoxantrone-Induced Immunogenic Cell Death. J Pharmacol Exp Ther 2021; 378:300-310. [PMID: 34158403 DOI: 10.1124/jpet.121.000629] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 06/16/2021] [Indexed: 11/22/2022] Open
Abstract
Agents that induce immunogenic cell death (ICD) alter the cellular localization of calreticulin (CRT), causing it to become cell surface-exposed within the plasma membrane lipid raft microdomain [cell surface-exposed CRT (ectoCRT)] where it serves as a damage associated-molecular pattern that elicits an antitumor immune response. We have identified the sphingolipid metabolic pathway as an integral component of the process of ectoCRT exposure. Inhibition of the sphingosine kinases (SphKs) enhances mitoxantrone-induced production of hallmarks of ICD, including ectoCRT production, with an absolute mean difference of 40 MFI (95% CI: 19-62; P = 0.0014) and 1.3-fold increase of ATP secretion with an absolute mean difference of 87 RLU (95% CI: 55-120; P < 0.0001). Mechanistically, sphingosine kinase inhibition increases mitoxantrone-induced accumulation of ceramide species, including C16:0 ceramide 2.8-fold with an absolute mean difference of 1.390 pmol/nmol Pi (95% CI: 0.798-1.983; P = 0.0023). We further examined the localization of ectoCRT to the lipid raft microdomain and demonstrate that ectoCRT forms disulfide-bridged dimers. Together, our findings suggest that ceramide accumulation impinges on the homeostatic function of the endoplasmic reticulum to induce ectoCRT exposure and that structural alterations of ectoCRT may underlie its immunogenicity. Our findings further suggest that inhibition of the SphKs may represent a means to enhance the therapeutic immunogenic efficacy of ICD-inducing agents while reducing overt toxicity/immunosuppressive effects by allowing for the modification of dosing regimens or directly lowering the dosages of ICD-inducing agents employed in therapeutic regimens. SIGNIFICANCE STATEMENT: This study demonstrates that inhibition of sphingosine kinase enhances the mitoxantrone-induced cell surface exposure of a dimeric form of the normally endoplasmic reticulum resident chaperone calreticulin as part of the process of a unique form of regulated cell death termed immunogenic cell death. Importantly, inhibition of sphingosine kinase may represent a means to enhance the therapeutic efficacy of immunogenic cell death-inducing agents, such as mitoxantrone, while reducing their overt toxicity and immunosuppressive effects, leading to better therapeutic outcomes for patients.
Collapse
Affiliation(s)
- Asvelt J Nduwumwami
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jeremy A Hengst
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| | - Jong K Yun
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
15
|
Alessenko AV, Blokhin VE, Shupik MA, Gutner UA, Lebedev AT, Maloshitskaya OA, Sokolov SA, Ugrumov MV. Changes in the Content of Sphingolipids in the Nigrostriatal Dopaminergic System in the Brain of Mice with a Neurotoxic Model of Parkinson’s Disease. NEUROCHEM J+ 2021. [DOI: 10.1134/s1819712421020021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
16
|
Acid ceramidase controls apoptosis and increases autophagy in human melanoma cells treated with doxorubicin. Sci Rep 2021; 11:11221. [PMID: 34045496 PMCID: PMC8159975 DOI: 10.1038/s41598-021-90219-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/04/2021] [Indexed: 02/04/2023] Open
Abstract
Acid ceramidase (AC) is a lysosomal hydrolase encoded by the ASAH1 gene, which cleaves ceramides into sphingosine and fatty acid. AC is expressed at high levels in most human melanoma cell lines and may confer resistance against chemotherapeutic agents. One such agent, doxorubicin, was shown to increase ceramide levels in melanoma cells. Ceramides contribute to the regulation of autophagy and apoptosis. Here we investigated the impact of AC ablation via CRISPR-Cas9 gene editing on the response of A375 melanoma cells to doxorubicin. We found that doxorubicin activates the autophagic response in wild-type A375 cells, which effectively resist apoptotic cell death. In striking contrast, doxorubicin fails to stimulate autophagy in A375 AC-null cells, which rapidly undergo apoptosis when exposed to the drug. The present work highlights changes that affect melanoma cells during incubation with doxorubicin, in A375 melanoma cells lacking AC. We found that the remarkable reduction in recovery rate after doxorubicin treatment is strictly associated with the impairment of autophagy, that forces the AC-inhibited cells into apoptotic path.
Collapse
|
17
|
Simon MV, Basu SK, Qaladize B, Grambergs R, Rotstein NP, Mandal N. Sphingolipids as critical players in retinal physiology and pathology. J Lipid Res 2021; 62:100037. [PMID: 32948663 PMCID: PMC7933806 DOI: 10.1194/jlr.tr120000972] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/04/2020] [Indexed: 12/24/2022] Open
Abstract
Sphingolipids have emerged as bioactive lipids involved in the regulation of many physiological and pathological processes. In the retina, they have been established to participate in numerous processes, such as neuronal survival and death, proliferation and migration of neuronal and vascular cells, inflammation, and neovascularization. Dysregulation of sphingolipids is therefore crucial in the onset and progression of retinal diseases. This review examines the involvement of sphingolipids in retinal physiology and diseases. Ceramide (Cer) has emerged as a common mediator of inflammation and death of neuronal and retinal pigment epithelium cells in animal models of retinopathies such as glaucoma, age-related macular degeneration (AMD), and retinitis pigmentosa. Sphingosine-1-phosphate (S1P) has opposite roles, preventing photoreceptor and ganglion cell degeneration but also promoting inflammation, fibrosis, and neovascularization in AMD, glaucoma, and pro-fibrotic disorders. Alterations in Cer, S1P, and ceramide 1-phosphate may also contribute to uveitis. Notably, use of inhibitors that either prevent Cer increase or modulate S1P signaling, such as Myriocin, desipramine, and Fingolimod (FTY720), preserves neuronal viability and retinal function. These findings underscore the relevance of alterations in the sphingolipid metabolic network in the etiology of multiple retinopathies and highlight the potential of modulating their metabolism for the design of novel therapeutic approaches.
Collapse
Affiliation(s)
- M Victoria Simon
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Sandip K Basu
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Bano Qaladize
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Richard Grambergs
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina.
| | - Nawajes Mandal
- Departments of Ophthalmology and Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
18
|
Lam BWS, Yam TYA, Chen CP, Lai MKP, Ong WY, Herr DR. The noncanonical chronicles: Emerging roles of sphingolipid structural variants. Cell Signal 2020; 79:109890. [PMID: 33359087 DOI: 10.1016/j.cellsig.2020.109890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/08/2023]
Abstract
Sphingolipids (SPs) are structurally diverse and represent one of the most quantitatively abundant classes of lipids in mammalian cells. In addition to their structural roles, many SP species are known to be bioactive mediators of essential cellular processes. Historically, studies have focused on SP species that contain the canonical 18‑carbon, mono-unsaturated sphingoid backbone. However, increasingly sensitive analytical technologies, driven by advances in mass spectrometry, have facilitated the identification of previously under-appreciated, molecularly distinct SP species. Many of these less abundant species contain noncanonical backbones. Interestingly, a growing number of studies have identified clinical associations between these noncanonical SPs and disease, suggesting that there is functional significance to the alteration of SP backbone structure. For example, associations have been found between SP chain length and cardiovascular disease, pain, diabetes, and dementia. This review will provide an overview of the processes that are known to regulate noncanonical SP accumulation, describe the clinical correlations reported for these molecules, and review the experimental evidence for the potential functional implications of their dysregulation. It is likely that further scrutiny of noncanonical SPs may provide new insight into pathophysiological processes, serve as useful biomarkers for disease, and lead to the design of novel therapeutic strategies.
Collapse
Affiliation(s)
- Brenda Wan Shing Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ting Yu Amelia Yam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory Aging and Cognition Centre, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Memory Aging and Cognition Centre, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Biology, San Diego State University, San Diego, CA, USA; American University of Health Sciences, Long Beach, CA, USA.
| |
Collapse
|
19
|
Alkaline ceramidase family: The first two decades. Cell Signal 2020; 78:109860. [PMID: 33271224 DOI: 10.1016/j.cellsig.2020.109860] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 11/21/2022]
Abstract
Ceramidases are a group of enzymes that catalyze the hydrolysis of ceramide, dihydroceramide, and phytoceramide into sphingosine (SPH), dihydrosphingosine (DHS), and phytosphingosine (PHS), respectively, along with a free fatty acid. Ceramidases are classified into the acid, neutral, and alkaline ceramidase subtypes according to the pH optima for their catalytic activity. YPC1 and YDC1 were the first alkaline ceramidase genes to be identified and cloned from the yeast Saccharomyces cerevisiae two decades ago. Subsequently, alkaline ceramidase genes were identified from other species, including one Drosophila melanogaster ACER gene (Dacer), one Arabidopsis thaliana ACER gene (AtACER), three Mus musculus ACER genes (Acer1, Acer2, and Acer3), and three Homo sapiens ACER genes (ACER1, ACER2, and ACER3). The protein products of these genes constitute a large protein family, termed the alkaline ceramidase (ACER) family. All the biochemically characterized members of the ACER family are integral membrane proteins with seven transmembrane segments in the Golgi complex or endoplasmic reticulum, and they each have unique substrate specificity. An increasing number of studies suggest that the ACER family has diverse roles in regulating sphingolipid metabolism and biological processes. Here we discuss the discovery of the ACER family, the biochemical properties, structures, and catalytic mechanisms of its members, and its role in regulating sphingolipid metabolism and biological processes in yeast, insects, plants, and mammals.
Collapse
|
20
|
Wang X, Yang Y, Cai WQ, Lu Y. The Relationship of Sphingosine Kinase 1 With Pyroptosis Provides a New Strategy for Tumor Therapy. Front Immunol 2020; 11:574990. [PMID: 33123153 PMCID: PMC7566665 DOI: 10.3389/fimmu.2020.574990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Sphingosine kinase 1 (SPHK1) is a crucial molecule that catalyzes sphingosine to synthesize sphingosine-1-phosphate (S1P), facilitating cell survival signaling. Pyroptosis is a perplexing inflammatory mode of cell death primarily triggered by caspase-1, evoked by the NLRP3 inflammasome. Sphingosine is identified as a danger-associated molecular pattern (DAMP), which activates the NLRP3 inflammasome assembly and induces the pyroptosis. It has been demonstrated that macrophages play a pro-tumorigenic role and are closely associated with tumor progression. Attenuation of SPHK1 activity contributes significantly to macrophage pyroptosis and tumor inhibition. Calcium and integrin-binding protein 1 (CIB1) plays an important role in the translocation of SPHK1 from the cytoplasm to the plasma membrane, whereas CIB2 blocks the subcellular trafficking of SPHK1. Therefore, knockout of CIB1 or over-expression of CIB2 will result in sphingosine accumulation and contribute significantly to cancer treatment by several approaches. First, it directly provokes cancer cell apoptosis or triggers robust anti-tumor immunity by pyroptosis-induced inflammation. Second, it could restrain SPHK1 translocation from the cytoplasm to the plasma membrane and further pyroptosis, which not only drive M2 macrophages death but also facilitate tumor microenvironment inflammation as well as the further release of sphingosine from damaged macrophages. The perspective might provide novel insight into the association between SPHK1 and pyroptosis and suggest the potential target for cancer therapy.
Collapse
Affiliation(s)
- Xianwang Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Yue Yang
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Wen-Qi Cai
- Department of Biochemistry and Molecular Biology, Health Science Center, Yangtze University, Jingzhou, China
| | - Yingying Lu
- The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
21
|
Park J, Lee NG, Oh M, Song J, Kim W, Kwon MG, Kim SG, Han BS, Bae KH, Lee DG, Lee SH, Park JG, Kim JH, Lee J, Min JK. Selective elimination of human pluripotent stem cells by Anti-Dsg2 antibody-doxorubicin conjugates. Biomaterials 2020; 259:120265. [PMID: 32827795 DOI: 10.1016/j.biomaterials.2020.120265] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/26/2020] [Indexed: 01/19/2023]
Abstract
The self-renewal properties of human pluripotent stem cells (hPSCs) contribute to their efficacy in tissue regeneration applications yet increase the likelihood of teratoma formation, thereby limiting their clinical utility. To address this issue, we developed a tool to specifically target and neutralize undifferentiated hPSCs, thereby minimizing tumorigenicity risk without negatively affecting regenerated and somatic tissues. Specifically, we conjugated a monoclonal antibody (K6-1) previously generated in our laboratory against desmoglein 2 (Dsg2), which is highly differentially expressed in undifferentiated hPSCs versus somatic tissues, to the chemotherapeutic agent doxorubicin (DOX). The K6-1-DOX conjugates were selectively targeted and incorporated into Dsg2-positive hPSCs, leading to pH-dependent endosomal release and nuclear localization of DOX with subsequent cytotoxicity via an apoptotic caspase cascade. Conversely, Dsg2-negative fibroblasts showed minimal conjugate uptake or cytotoxicity, suggesting that K6-1-DOX treatment would yield few side effects owing to off-target effects. Selective removal of undifferentiated stem cells was also supported by in vivo studies using a mouse xenograft model, wherein hIgG-DOX- but not K6-1-DOX-pretreated-hPSC injection led to teratoma development. Together, these results validated the ability of the Dsg2-targeted antibody-anticancer drug conjugate to facilitate the safety of stem cell therapies.
Collapse
Affiliation(s)
- Jongjin Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Na Geum Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Mihee Oh
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jinhoi Song
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Wooil Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Min-Gi Kwon
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Seul Gi Kim
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Baek Soo Han
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Kwang-Hee Bae
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Dong Gwang Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Sang-Hyun Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jong-Gil Park
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea
| | - Jae Ho Kim
- Department of Physiology, Pusan National University Yangsan Hospital, Yangsan, 50612, Republic of Korea
| | - Jangwook Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea.
| | - Jeong-Ki Min
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, 34141, Republic of Korea; Department of Biomolecular Science, KRIBB School of Bioscience, Korea University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon, 34113, Republic of Korea.
| |
Collapse
|
22
|
Rothemund M, Bär A, Klatt F, Weidler S, Köhler L, Unverzagt C, Kuhn CD, Schobert R. N-Metallocenoylsphingosines as targeted ceramidase inhibitors: Syntheses and antitumoral effects. Bioorg Chem 2020; 97:103703. [PMID: 32143017 DOI: 10.1016/j.bioorg.2020.103703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 12/14/2022]
Abstract
Three N-metallocenoylsphingosines with variance in the central metal (Fe, Co, Ru), the charge (neutral or cationic), and the arene ligands (Cp2, Cp*Ph) were synthesized from serine and metallocene carboxylic acids as substrate-analogous inhibitors of human acid ceramidase (AC). Their inhibitory potential was examined using the recombinant full length ASAH1 enzyme, expressed and secreted from High Five insect cells, and the fluorescent substrate Rbm14-12. All complexes inhibited AC, most strongly so ruthenium(II) complex 13a. Some antitumoral effects of the complexes, such as the interference with the microtubular and F-actin cytoskeleton of cancer cells, were correlated to their AC-inhibition, whereas others, e.g. their cytotoxicity and their induction of caspase-3/-7 activity in cancer cells, were not. All complexes accumulated preferentially in the lysosomes of cancer cells like their target AC, arrested the cells in G1 phase of the cell cycle, and displayed cytotoxicity with mostly single-digit micromolar IC50 values while inducing cancer cell apoptosis.
Collapse
Affiliation(s)
- Matthias Rothemund
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440 Bayreuth, Germany
| | - Alexander Bär
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440 Bayreuth, Germany
| | - Felix Klatt
- Gene Regulation by Non-Coding RNA, Elite Network of Bavaria and University of Bayreuth, Universitaetsstr. 30, 95447 Bayreuth, Germany
| | - Sascha Weidler
- Bioorganic Chemistry, University of Bayreuth, Universitaetsstr. 30, 95447 Bayreuth, Germany
| | - Leonhard Köhler
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440 Bayreuth, Germany
| | - Carlo Unverzagt
- Bioorganic Chemistry, University of Bayreuth, Universitaetsstr. 30, 95447 Bayreuth, Germany
| | - Claus-D Kuhn
- Gene Regulation by Non-Coding RNA, Elite Network of Bavaria and University of Bayreuth, Universitaetsstr. 30, 95447 Bayreuth, Germany
| | - Rainer Schobert
- Department of Chemistry, University Bayreuth, Universitaetsstrasse 30, 95440 Bayreuth, Germany.
| |
Collapse
|
23
|
Gutner UA, Shupik MA, Maloshitskaya OA, Sokolov SA, Rezvykh AP, Funikov SY, Lebedev AT, Ustyugov AA, Alessenko AV. Changes in the Metabolism of Sphingoid Bases in the Brain and Spinal Cord of Transgenic FUS(1-359) Mice, a Model of Amyotrophic Lateral Sclerosis. BIOCHEMISTRY (MOSCOW) 2019; 84:1166-1176. [PMID: 31694512 DOI: 10.1134/s0006297919100055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The aim of this study was to evaluate changes in the content of sphingoid bases - sphingosine (SPH), sphinganine, and sphingosine-1-phosphate (SPH-1-P) - and in expression of genes encoding enzymes involved in their metabolism in the brain structures (hippocampus, cortex, and cerebellum) and spinal cord of transgenic FUS(1-359) mice. FUS(1-359) mice are characterized by motor impairments and can be used as a model of amyotrophic lateral sclerosis (ALS). Lipids from the mouse brain structures and spinal cord after 2, 3, and 4 months of disease development were analyzed by chromatography/mass spectrometry, while changes in the expression of the SPHK1, SPHK2, SGPP2, SGPL1, ASAH1, and ASAH2 genes were assayed using RNA sequencing. The levels of SPH and sphinganine (i.e., sphingoid bases with pronounced pro-apoptotic properties) were dramatically increased in the spinal cord at the terminal stage of the disease. The ratio of the anti-apoptotic SPH-1-P to SPH and sphinganine sharply reduced, indicating massive apoptosis of spinal cord cells. Significant changes in the content of SPH and SPH-1-P and in the expression of genes related to their metabolism were found at the terminal ALS stage in the spinal cord. Expression of the SGPL gene (SPH-1-P lyase) was strongly activated, while expression of the SGPP2 (SPH-1-P phosphatase) gene was reduced. Elucidation of mechanisms for the regulation of sphingolipid metabolism in ALS will help to identify molecular targets for the new-generation drugs.
Collapse
Affiliation(s)
- U A Gutner
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - M A Shupik
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia
| | - O A Maloshitskaya
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119999, Russia
| | - S A Sokolov
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119999, Russia
| | - A P Rezvykh
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - S Yu Funikov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, 119991, Russia
| | - A T Lebedev
- Lomonosov Moscow State University, Faculty of Chemistry, Moscow, 119999, Russia
| | - A A Ustyugov
- Institute of Physiologically Active Compounds, Russian Academy of Sciences, Chernogolovka, Moscow Region, 142432, Russia
| | - A V Alessenko
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, 119991, Russia.
| |
Collapse
|
24
|
Suhrland C, Truman J, Obeid LM, Sitharaman B. Delivery of long chain C16and C24ceramide in HeLa cells using oxidized graphene nanoribbons. J Biomed Mater Res B Appl Biomater 2019; 108:1141-1156. [DOI: 10.1002/jbm.b.34465] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 06/24/2019] [Accepted: 07/13/2019] [Indexed: 01/15/2023]
Affiliation(s)
- Cassandra Suhrland
- Department of Biomedical EngineeringStony Brook University Stony Brook New York
| | - Jean‐Philip Truman
- Department of Medicine and the Stony Brook Cancer Center, Health Science CenterStony Brook University Stony Brook New York
| | - Lina M. Obeid
- Department of Medicine and the Stony Brook Cancer Center, Health Science CenterStony Brook University Stony Brook New York
| | - Balaji Sitharaman
- Department of Biomedical EngineeringStony Brook University Stony Brook New York
| |
Collapse
|
25
|
Simón MV, Prado Spalm FH, Vera MS, Rotstein NP. Sphingolipids as Emerging Mediators in Retina Degeneration. Front Cell Neurosci 2019; 13:246. [PMID: 31244608 PMCID: PMC6581011 DOI: 10.3389/fncel.2019.00246] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/17/2019] [Indexed: 12/12/2022] Open
Abstract
The sphingolipids ceramide (Cer), sphingosine-1-phosphate (S1P), sphingosine (Sph), and ceramide-1-phosphate (C1P) are key signaling molecules that regulate major cellular functions. Their roles in the retina have gained increasing attention during the last decade since they emerge as mediators of proliferation, survival, migration, neovascularization, inflammation and death in retina cells. As exacerbation of these processes is central to retina degenerative diseases, they appear as crucial players in their progression. This review analyzes the functions of these sphingolipids in retina cell types and their possible pathological roles. Cer appears as a key arbitrator in diverse retinal pathologies; it promotes inflammation in endothelial and retina pigment epithelium (RPE) cells and its increase is a common feature in photoreceptor death in vitro and in animal models of retina degeneration; noteworthy, inhibiting Cer synthesis preserves photoreceptor viability and functionality. In turn, S1P acts as a double edge sword in the retina. It is essential for retina development, promoting the survival of photoreceptors and ganglion cells and regulating proliferation and differentiation of photoreceptor progenitors. However, S1P has also deleterious effects, stimulating migration of Müller glial cells, angiogenesis and fibrosis, contributing to the inflammatory scenario of proliferative retinopathies and age related macular degeneration (AMD). C1P, as S1P, promotes photoreceptor survival and differentiation. Collectively, the expanding role for these sphingolipids in the regulation of critical processes in retina cell types and in their dysregulation in retina degenerations makes them attractive targets for treating these diseases.
Collapse
Affiliation(s)
- M Victoria Simón
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Facundo H Prado Spalm
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Marcela S Vera
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| | - Nora P Rotstein
- Instituto de Investigaciones Bioquímicas de Bahía Blanca (INIBIBB), Departamento De Biología, Bioquímica y Farmacia, Universidad Nacional del Sur (UNS), Argentine National Research Council (CONICET), Bahía Blanca, Argentina
| |
Collapse
|
26
|
Suhrland C, Truman JP, Obeid LM, Sitharaman B. Oxidized graphene nanoparticles as a delivery system for the pro-apoptotic sphingolipid C 6 ceramide. J Biomed Mater Res A 2018; 107:25-37. [PMID: 30422374 DOI: 10.1002/jbm.a.36474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/16/2018] [Accepted: 05/24/2018] [Indexed: 01/22/2023]
Abstract
Sphingolipids such as ceramide have attracted much attention as possible anticancer agents due to their potent pro-apoptotic effects. However, due to their extreme hydrophobicity, there is currently no clinically approved delivery method for in vivo use as a therapeutic agent. To this end, we have developed a novel method for loading the short-chain C6 ceramide onto oxidized graphene nanoribbons (O-GNRs) and graphene nanoplatelets (GNPs). Mass spectrometry revealed loading efficiencies of 57% and 51.5% for C6 ceramide onto O-GNRs and GNPs, respectively. The PrestoBlue viability assay revealed that 100 µg/mL of C6 ceramide-loaded O-GNRs and C6 ceramide-loaded GNPs reduced HeLa cell viability by approximately 93% and approximately 76%, respectively, compared to untreated HeLa cells, while equal concentrations of these nanoparticles without C6 ceramide did not significantly reduce HeLa cell viability. We confirmed that this cytotoxicity was apoptotic in nature via capase-3 activity and Hoechst staining. Using live-cell confocal imaging with the fluorescent NBD-ceramide loaded on O-GNRs, we observed robust uptake into HeLa cells within 30 min while NBD-ceramide on its own was uptaken much more rapidly. Transmission electron microscopy confirmed that C6 ceramide-loaded O-GNRs were actually entering cells. Taken together, these data show that O-GNRs are a promising delivery agent for ceramide. To our knowledge, this study is the first to use such a loading method. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 25-37, 2019.
Collapse
Affiliation(s)
- Cassandra Suhrland
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| | - Jean-Philip Truman
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, New York, New York
| | - Lina M Obeid
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, New York, New York
| | - Balaji Sitharaman
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, New York
| |
Collapse
|
27
|
Zhang C, Maddelein ML, Wai-Yin Sun R, Gornitzka H, Cuvillier O, Hemmert C. Pharmacomodulation on Gold-NHC complexes for anticancer applications - is lipophilicity the key point? Eur J Med Chem 2018; 157:320-332. [PMID: 30099254 DOI: 10.1016/j.ejmech.2018.07.070] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 07/24/2018] [Accepted: 07/29/2018] [Indexed: 12/20/2022]
Abstract
A series of four new mononuclear cationic gold(I) complexes containing nitrogen functionalized N-heterocyclic carbenes (NHCs) was synthesized and fully characterized by spectroscopic methods. The X-ray structures of three complexes are presented. These lipophilic gold(I) complexes originate from a pharmacomodulation of previously described gold(I)-NHC complexes, by replacing an aliphatic spacer with an aromatic one. The Log P values of the resulting complexes increased by 0.7-1.5, depending on the substituents in comparison to the aliphatic-linker systems. The new series of complexes has been investigated in vitro for their anti-cancer activities in PC-3 (prostate cancer) and T24 (bladder cancer) cell lines and in the non-cancerous MC3T3 (osteoblast) cell line. All tested complexes show high activities against the cancer cell lines with GI50 values lower than 500 nM. One complex (11) has been selected for further investigations. It has been tested in vitro in six cancer cell lines from different origins (prostate, bladder, lung, bone, liver and breast) and two non-cancerous cell lines (osteoblasts, fibroblasts). Moreover, cellular uptake measurements were indicative of a good bioavailability. By various biochemical assays, this complex was found to effectively inhibit the thioredoxin reductase (TrxR) and its cytotoxicity towards prostate PC-3, bladder T24 and liver HepG2 cells was found to be ROS-dependent.
Collapse
Affiliation(s)
- Chen Zhang
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Marie-Lise Maddelein
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France
| | - Raymond Wai-Yin Sun
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, PR China
| | - Heinz Gornitzka
- LCC-CNRS, Université de Toulouse, CNRS, UPS, Toulouse, France.
| | - Olivier Cuvillier
- Institut de Pharmacologie et de Biologie Structurale, Université de Toulouse, CNRS, UPS, Toulouse, France.
| | | |
Collapse
|
28
|
Li W, Wang J, Hu H, Li Q, Liu Y, Wang K. Functional polysaccharide Lentinan suppresses human breast cancer growth via inducing autophagy and caspase-7-mediated apoptosis. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.03.024] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
29
|
Yi JK, Xu R, Jeong E, Mileva I, Truman JP, Lin CL, Wang K, Snider J, Wen S, Obeid LM, Hannun YA, Mao C. Aging-related elevation of sphingoid bases shortens yeast chronological life span by compromising mitochondrial function. Oncotarget 2018; 7:21124-44. [PMID: 27008706 PMCID: PMC5008273 DOI: 10.18632/oncotarget.8195] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 01/08/2023] Open
Abstract
Sphingoid bases (SBs) as bioactive sphingolipids, have been implicated in aging in yeast. However, we know neither how SBs are regulated during yeast aging nor how they, in turn, regulate it. Herein, we demonstrate that the yeast alkaline ceramidases (YPC1 and YDC1) and SB kinases (LCB4 and LCB5) cooperate in regulating SBs during the aging process and that SBs shortens chronological life span (CLS) by compromising mitochondrial functions. With a lipidomics approach, we found that SBs were increased in a time-dependent manner during yeast aging. We also demonstrated that among the enzymes known for being responsible for the metabolism of SBs, YPC1 was upregulated whereas LCB4/5 were downregulated in the course of aging. This inverse regulation of YPC1 and LCB4/5 led to the aging-related upregulation of SBs in yeast and a reduction in CLS. With the proteomics-based approach (SILAC), we revealed that increased SBs altered the levels of proteins related to mitochondria. Further mechanistic studies demonstrated that increased SBs inhibited mitochondrial fusion and caused fragmentation, resulting in decreases in mtDNA copy numbers, ATP levels, mitochondrial membrane potentials, and oxygen consumption. Taken together, these results suggest that increased SBs mediate the aging process by impairing mitochondrial structural integrity and functions.
Collapse
Affiliation(s)
- Jae Kyo Yi
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Ruijuan Xu
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Eunmi Jeong
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Izolda Mileva
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | | | - Chih-Li Lin
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Kai Wang
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Justin Snider
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Sally Wen
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA.,Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Cungui Mao
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| |
Collapse
|
30
|
Lin CL, Xu R, Yi JK, Li F, Chen J, Jones EC, Slutsky JB, Huang L, Rigas B, Cao J, Zhong X, Snider AJ, Obeid LM, Hannun YA, Mao C. Alkaline Ceramidase 1 Protects Mice from Premature Hair Loss by Maintaining the Homeostasis of Hair Follicle Stem Cells. Stem Cell Reports 2017; 9:1488-1500. [PMID: 29056331 PMCID: PMC5829345 DOI: 10.1016/j.stemcr.2017.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/31/2022] Open
Abstract
Ceramides and their metabolites are important for the homeostasis of the epidermis, but much remains unknown about the roles of specific pathways of ceramide metabolism in skin biology. With a mouse model deficient in the alkaline ceramidase (Acer1) gene, we demonstrate that ACER1 plays a key role in the homeostasis of the epidermis and its appendages by controlling the metabolism of ceramides. Loss of Acer1 elevated the levels of various ceramides and sphingoid bases in the skin and caused progressive hair loss in mice. Mechanistic studies revealed that loss of Acer1 widened follicular infundibulum and caused progressive loss of hair follicle stem cells (HFSCs) due to reduced survival and stemness. These results suggest that ACER1 plays a key role in maintaining the homeostasis of HFSCs, and thereby the hair follicle structure and function, by regulating the metabolism of ceramides in the epidermis. Acer1 is a skin-specific ceramidase that controls the catabolism of ceramides Acer1 plays a key role in the homeostasis of the epidermis and its appendages Acer1−/− mice suffer from progressive alopecia Loss of Acer1 progressively depletes the population of hair follicle stem cells
Collapse
Affiliation(s)
- Chih-Li Lin
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Ruijuan Xu
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA
| | - Jae Kyo Yi
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Fang Li
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA
| | - Jiang Chen
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Evan C Jones
- Department of Dermatology, Stony Brook University, Stony Brook, NY, USA
| | - Jordan B Slutsky
- Department of Dermatology, Stony Brook University, Stony Brook, NY, USA
| | - Liqun Huang
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Basil Rigas
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Jian Cao
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Xiaoming Zhong
- Industrial Technology Research Institute, Zhejiang University, Hangzhou, Zhejiang Province, China
| | - Ashley J Snider
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA
| | - Cungui Mao
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, HSC T15-023, Stony Brook, NY 11794, USA; Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA; Department of Dermatology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
31
|
Jabłońska-Trypuć A, Świderski G, Krętowski R, Lewandowski W. Newly Synthesized Doxorubicin Complexes with Selected Metals-Synthesis, Structure and Anti-Breast Cancer Activity. Molecules 2017; 22:molecules22071106. [PMID: 28677642 PMCID: PMC6152354 DOI: 10.3390/molecules22071106] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 06/30/2017] [Accepted: 07/01/2017] [Indexed: 12/31/2022] Open
Abstract
Doxorubicin (DOX) is very effective chemotherapeutic agent, however it has several major drawbacks. Therefore the motivation for developing novel drug complexes as anticancer agents with different mechanism of action has arisen. The aim of the present study was to evaluate the influence of newly synthesized DOX complexes with selected metals (Mg, Mn, Co, Ni, Fe, Cu, Zn) on apoptosis, cell cycle, viability, proliferation and cytotoxicity in the breast cancer cell line MCF-7. Complexation of DOX with metals has likewise been the subject of our research. The current work showed that the tested bivalent metals at a given pH condition formed metal:DOX complexes in a ratio of 2:1, while iron complexes with DOX in a ratio of 3:1. The studies also showed that selected metal-DOX complexes (Mg-DOX, Mn-DOX, Ni-DOX) at 0.5 µM concentration significantly decreased cell viability and proliferation, however they increased caspase 7 activity. Results also indicated that studied metal-DOX complexes showed high cytotoxicity in MCF-7 cells. Therefore they were chosen for cell cycle check-points and apoptosis/necrosis analysis studied by flow cytometry. Obtained results suggest that doxorubicin complexed by specified metals can be considered as a potential anti-breast cancer agent, which is characterized by a higher efficacy than a parent drug.
Collapse
Affiliation(s)
- Agata Jabłońska-Trypuć
- Division of Sanitary Biology and Biotechnology, Faculty of Civil Engineering and Environmental Engineering, Białystok University of Technology, Wiejska 45E Street, Białystok 15-351, Poland.
| | - Grzegorz Świderski
- Division of Chemistry, Bialystok University of Technology, Białystok 15-351, Poland.
| | - Rafał Krętowski
- Department of Pharmaceutical Biochemistry, Medical University of Białystok, Białystok 15-222, Poland.
| | | |
Collapse
|
32
|
Cingolani F, Simbari F, Abad JL, Casasampere M, Fabrias G, Futerman AH, Casas J. Jaspine B induces nonapoptotic cell death in gastric cancer cells independently of its inhibition of ceramide synthase. J Lipid Res 2017; 58:1500-1513. [PMID: 28572516 DOI: 10.1194/jlr.m072611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Sphingolipids (SLs) have been extensively investigated in biomedical research due to their role as bioactive molecules in cells. Here, we describe the effect of a SL analog, jaspine B (JB), a cyclic anhydrophytosphingosine found in marine sponges, on the gastric cancer cell line, HGC-27. JB induced alterations in the sphingolipidome, mainly the accumulation of dihydrosphingosine, sphingosine, and their phosphorylated forms due to inhibition of ceramide synthases. Moreover, JB provoked atypical cell death in HGC-27 cells, characterized by the formation of cytoplasmic vacuoles in a time and dose-dependent manner. Vacuoles appeared to originate from macropinocytosis and triggered cytoplasmic disruption. The pan-caspase inhibitor, z-VAD, did not alter either cytotoxicity or vacuole formation, suggesting that JB activates a caspase-independent cell death mechanism. The autophagy inhibitor, wortmannin, did not decrease JB-stimulated LC3-II accumulation. In addition, cell vacuolation induced by JB was characterized by single-membrane vacuoles, which are different from double-membrane autophagosomes. These findings suggest that JB-induced cell vacuolation is not related to autophagy and it is also independent of its action on SL metabolism.
Collapse
Affiliation(s)
- Francesca Cingolani
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.
| | - Fabio Simbari
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Jose Luis Abad
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Mireia Casasampere
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Gemma Fabrias
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain
| | - Anthony H Futerman
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot, Israel
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Barcelona, Spain.
| |
Collapse
|
33
|
Wang Y, Zhang C, Jin Y, Wang, He Q, Liu Z, Ai Q, Lei Y, Li Y, Song F, Bu Y. Alkaline ceramidase 2 is a novel direct target of p53 and induces autophagy and apoptosis through ROS generation. Sci Rep 2017; 7:44573. [PMID: 28294157 PMCID: PMC5353723 DOI: 10.1038/srep44573] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 02/09/2017] [Indexed: 12/22/2022] Open
Abstract
ACER2 is a critical sphingolipid metabolizing enzyme, and has been shown to be remarkably up-regulated following various stimuli such as DNA damage. However, the transcriptional regulatory mechanism of ACER2 gene and its potential role in the regulation of autophagy remain unknown. In this study, we have for the first time identified the human ACER2 gene promoter, and found that human ACER2 transcription is directly regulated by p53 and ACER2 is implicated in the induction of autophagy as well as apoptosis. A series of luciferase reporter assay demonstrated that ACER2 major promoter is located within its first intron where the consensus p53-binding sites exist. Consistently, forced expression of p53 significantly stimulated ACER2 transcription. Notably, p53-mediated autophagy and apoptosis were markedly enhanced by ACER2. Depletion of the essential autophagy gene ATG5 revealed that ACER2-induced autophagy facilitates its effect on apoptosis. Further studies clearly showed that ACER2-mediated autophagy and apoptosis are accompanied by ROS generation. In summary, our present study strongly suggests that ACER2 plays a pivotal role in p53-induced autophagy and apoptosis, and thus might serve as a novel and attractive molecular target for cancer treatment.
Collapse
Affiliation(s)
- Yitao Wang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Chunxue Zhang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yuelei Jin
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Wang
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Qing He
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Zhu Liu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Qing Ai
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Yi Li
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Fangzhou Song
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| | - Youquan Bu
- Department of Biochemistry and Molecular Biology, Chongqing Medical University, Chongqing 400016, China
- Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, China
| |
Collapse
|
34
|
Xu R, Wang K, Mileva I, Hannun YA, Obeid LM, Mao C. Alkaline ceramidase 2 and its bioactive product sphingosine are novel regulators of the DNA damage response. Oncotarget 2017; 7:18440-57. [PMID: 26943039 PMCID: PMC4951300 DOI: 10.18632/oncotarget.7825] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 01/29/2016] [Indexed: 12/17/2022] Open
Abstract
Human cells respond to DNA damage by elevating sphingosine, a bioactive sphingolipid that induces programmed cell death (PCD) in response to various forms of stress, but its regulation and role in the DNA damage response remain obscure. Herein we demonstrate that DNA damage increases sphingosine levels in tumor cells by upregulating alkaline ceramidase 2 (ACER2) and that the upregulation of the ACER2/sphingosine pathway induces PCD in response to DNA damage by increasing the production of reactive oxygen species (ROS). Treatment with the DNA damaging agent doxorubicin increased both ACER2 expression and sphingosine levels in HCT116 cells in a dose-dependent manner. ACER2 overexpression increased sphingosine in HeLa cells whereas knocking down ACER2 inhibited the doxorubicin-induced increase in sphingosine in HCT116 cells, suggesting that DNA damage elevates sphingosine by upregulating ACER2. Knocking down ACER2 inhibited an increase in the apoptotic and necrotic cell population and the cleavage of poly ADP ribose polymerase (PARP) in HCT116 cells in response to doxorubicin as well as doxorubicin-induced release of lactate dehydrogenase (LDH) from these cells. Similar to treatment with doxorubicin, ACER2 overexpression induced an increase in the apoptotic and necrotic cell population and PARP cleavage in HeLa cells and LDH release from cells, suggesting that ACER2 upregulation mediates PCD in response to DNA damage through sphingosine. Mechanistic studies demonstrated that the upregulation of the ACER2/sphingosine pathway induces PCD by increasing ROS levels. Taken together, these results suggest that the ACER2/sphingosine pathway mediates PCD in response to DNA damage through ROS production.
Collapse
Affiliation(s)
- Ruijuan Xu
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Stony Brook Cancer Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Kai Wang
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Stony Brook Cancer Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Izolda Mileva
- Lipidomics Core Facility, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Yusuf A Hannun
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Stony Brook Cancer Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Stony Brook Cancer Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Ralph H. Johnson Veterans Administration Hospital, Stony Brook, NY 11794, USA
| | - Cungui Mao
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY 11794, USA.,Stony Brook Cancer Center, State University of New York at Stony Brook, Stony Brook, NY 11794, USA
| |
Collapse
|
35
|
Hernández-Corbacho MJ, Salama MF, Canals D, Senkal CE, Obeid LM. Sphingolipids in mitochondria. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:56-68. [PMID: 27697478 DOI: 10.1016/j.bbalip.2016.09.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Revised: 08/01/2016] [Accepted: 09/24/2016] [Indexed: 01/16/2023]
Abstract
Sphingolipids are bioactive lipids found in cell membranes that exert a critical role in signal transduction. In recent years, it has become apparent that sphingolipids participate in growth, senescence, differentiation and apoptosis. The anabolism and catabolism of sphingolipids occur in discrete subcellular locations and consist of a strictly regulated and interconnected network, with ceramide as the central hub. Altered sphingolipid metabolism is linked to several human diseases. Hence, an advanced knowledge of how and where sphingolipids are metabolized is of paramount importance in order to understand the role of sphingolipids in cellular functions. In this review, we provide an overview of sphingolipid metabolism. We focus on the distinct pathways of ceramide synthesis, highlighting the mitochondrial ceramide generation, transport of ceramide to mitochondria and its role in the regulation of mitochondrial-mediated apoptosis, mitophagy and implications to disease. We will discuss unanswered questions and exciting future directions. This article is part of a Special Issue entitled: Lipids of Mitochondria edited by Guenther Daum.
Collapse
Affiliation(s)
- María José Hernández-Corbacho
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Mohamed F Salama
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA; Department of Biochemistry, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Daniel Canals
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Can E Senkal
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and the Department of Medicine, Stony Brook University, Health Sciences Center, Stony Brook, NY 11794, USA; The Northport VA Medical Center, Northport, NY 11768, USA.
| |
Collapse
|
36
|
Pisklakova A, McKenzie B, Zemp F, Lun X, Kenchappa RS, Etame AB, Rahman MM, Reilly K, Pilon-Thomas S, McFadden G, Kurz E, Forsyth PA. M011L-deficient oncolytic myxoma virus induces apoptosis in brain tumor-initiating cells and enhances survival in a novel immunocompetent mouse model of glioblastoma. Neuro Oncol 2016; 18:1088-1098. [PMID: 26962017 DOI: 10.1093/neuonc/now006] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Myxoma virus (MYXV) is a promising oncolytic agent and is highly effective against immortalized glioma cells but less effective against brain tumor initiating cells (BTICs), which are believed to mediate glioma development/recurrence. MYXV encodes various proteins to attenuate host cell apoptosis, including an antiapoptotic Bcl-2 homologue known as M011L. Such proteins may limit the ability of MYXV to kill BTICs, which have heightened resistance to apoptosis. We hypothesized that infecting BTICs with an M011L-deficient MYXV construct would overcome BTIC resistance to MYXV. METHODS We used patient-derived BTICs to evaluate the efficacy of M011L knockout virus (vMyx-M011L-KO) versus wild-type MYXV (vMyx-WT) and characterized the mechanism of virus-induced cell death in vitro. To extend our findings in a novel immunocompetent animal model, we derived, cultured, and characterized a C57Bl/6J murine BTIC (mBTIC0309) from a spontaneous murine glioma and evaluated vMyx-M011L-KO efficacy with and without temozolomide (TMZ) in mBTIC0309-bearing mice. RESULTS We demonstrated that vMyx-M011L-KO induces apoptosis in BTICs, dramatically increasing sensitivity to the virus. vMyx-WT failed to induce apoptosis as M011L protein prevented Bax activation and cytochrome c release. In vivo, intracranial implantation of mBTIC0309 generated tumors that closely recapitulated the pathological and molecular profile of human gliomas. Treatment of tumor-bearing mice with vMyx-M011L-KO significantly prolonged survival in immunocompetent-but not immunodeficient-mouse models, an effect that is significantly enhanced in combination with TMZ. CONCLUSIONS Our data suggest that vMyx-M011L-KO is an effective, well-tolerated, proapoptotic oncolytic virus and a strong candidate for clinical translation.
Collapse
Affiliation(s)
- Alexandra Pisklakova
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Brienne McKenzie
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Franz Zemp
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Xueqing Lun
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Rajappa S Kenchappa
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Arnold B Etame
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Masmudur M Rahman
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Karlyne Reilly
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Shari Pilon-Thomas
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Grant McFadden
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Ebba Kurz
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| | - Peter A Forsyth
- Department of Neuro-Oncology and Tumor Biology, Moffitt Cancer Center, Tampa, Florida (A.P., R.S.K., A.B.E., P.A.F.); Southern Alberta Cancer Research Institute, University of Calgary, Calgary, Alberta, Canada (B.M., F.Z., X.L., E.K., P.A.F.); Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, Florida (M.M.R., G.M.); Center for Cancer Research, National Cancer Institute, Bethesda, Maryland (K.R.); Department of Immunology, Moffitt Cancer Center, Tampa, Florida (S.P.-T.)
| |
Collapse
|
37
|
Teixeira V, Costa V. Unraveling the role of the Target of Rapamycin signaling in sphingolipid metabolism. Prog Lipid Res 2015; 61:109-33. [PMID: 26703187 DOI: 10.1016/j.plipres.2015.11.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 11/04/2015] [Accepted: 11/09/2015] [Indexed: 02/06/2023]
Abstract
Sphingolipids are important bioactive molecules that regulate basic aspects of cellular metabolism and physiology, including cell growth, adhesion, migration, senescence, apoptosis, endocytosis, and autophagy in yeast and higher eukaryotes. Since they have the ability to modulate the activation of several proteins and signaling pathways, variations in the relative levels of different sphingolipid species result in important changes in overall cellular functions and fate. Sphingolipid metabolism and their route of synthesis are highly conserved from yeast to mammalian cells. Studies using the budding yeast Saccharomyces cerevisiae have served in many ways to foster our understanding of sphingolipid dynamics and their role in the regulation of cellular processes. In the past decade, studies in S. cerevisiae have unraveled a functional association between the Target of Rapamycin (TOR) pathway and sphingolipids, showing that both TOR Complex 1 (TORC1) and TOR Complex 2 (TORC2) branches control temporal and spatial aspects of sphingolipid metabolism in response to physiological and environmental cues. In this review, we report recent findings in this emerging and exciting link between the TOR pathway and sphingolipids and implications in human health and disease.
Collapse
Affiliation(s)
- Vitor Teixeira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal; ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Departamento de Biologia Molecular, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal
| | - Vítor Costa
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; IBMC, Instituto de Biologia Molecular e Celular, Porto, Portugal; ICBAS, Instituto de Ciências Biomédicas Abel Salazar, Departamento de Biologia Molecular, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal.
| |
Collapse
|
38
|
Hair Cell Loss Induced by Sphingosine and a Sphingosine Kinase Inhibitor in the Rat Cochlea. Neurotox Res 2015; 29:35-46. [DOI: 10.1007/s12640-015-9563-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/27/2022]
|
39
|
Cingolani F, Futerman AH, Casas J. Ceramide synthases in biomedical research. Chem Phys Lipids 2015; 197:25-32. [PMID: 26248326 DOI: 10.1016/j.chemphyslip.2015.07.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/30/2015] [Accepted: 07/31/2015] [Indexed: 01/05/2023]
Abstract
Sphingolipid metabolism consists of multiple metabolic pathways that converge upon ceramide, one of the key molecules among sphingolipids (SLs). In mammals, ceramide synthesis occurs via N-acylation of sphingoid backbones, dihydrosphingosine (dhSo) or sphingosine (So). The reaction is catalyzed by ceramide synthases (CerS), a family of enzymes with six different isoforms, with each one showing specificity towards a restricted group of acyl-CoAs, thus producing ceramides (Cer) and dihydroceramides (dhCer) with different fatty acid chain lengths. A large body of evidence documents the role of both So and dhSo as bioactive molecules, as well as the involvement of dhCer and Cer in physiological and pathological processes. In particular, the fatty acid composition of Cer has different effects in cell biology and in the onset and progression of different diseases. Therefore, modulation of CerS activity represents an attractive target in biomedical research and in finding new treatment modalities. In this review, we discuss functional, structural and biochemical features of CerS and examine CerS inhibitors that are currently available.
Collapse
Affiliation(s)
- Francesca Cingolani
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034 Barcelona, Spain
| | - Anthony H Futerman
- Department of Biological Chemistry, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034 Barcelona, Spain.
| |
Collapse
|
40
|
Ruan Y, Hu K, Chen H. Autophagy inhibition enhances isorhamnetin‑induced mitochondria‑dependent apoptosis in non‑small cell lung cancer cells. Mol Med Rep 2015; 12:5796-806. [PMID: 26238746 PMCID: PMC4581743 DOI: 10.3892/mmr.2015.4148] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 06/22/2015] [Indexed: 12/26/2022] Open
Abstract
Isorhamnetin (ISO) is a flavonoid from plants of the Polygonaceae family and is also an immediate metabolite of quercetin in mammals. To date, the anti-tumor effects of ISO and the underlying mechanisms have not been elucidated in lung cancer cells. The present study investigated the inhibitory effects of ISO on the growth of human lung cancer A549 cells. Treatment of the lung cancer cells with ISO significantly suppressed cell proliferation and colony formation. ISO treatment also resulted in a significant increase in apoptotic cell death of A549 cells in a time- and dose-dependent manner. Further investigation showed that the apoptosis proceeded via the mitochondria-dependent pathway as indicated by alteration of the mitochondrial membrane potential, the release of cytochrome C and caspase activation. Of note, treatment with ISO also induced the formation of autophagosomes and light chain 3-II protein in A549 cells. Furthermore, co-treatment with autophagy inhibitors 3-methyladenine and hydroxychloroquine significantly inhibited the ISO-induced autophagy and enhanced the ISO-induced apoptotic cell death in vitro as well as in vivo. Thus, the results of the present study suggested that ISO is a potential anti-lung cancer agent. In addition, the results indicated that the inhibition of autophagy may be a useful strategy for enhancing the chemotherapeutic effect of ISO on lung cancer cells.
Collapse
Affiliation(s)
- Yushu Ruan
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ke Hu
- Division of Respiratory Disease, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Hongbo Chen
- The Shenzhen Key Lab of Gene and Antibody Therapy, Division of Life and Health Sciences, Shenzhen Graduate School of Tsinghua University, Shenzhen, Guangdong 518055, P.R. China
| |
Collapse
|
41
|
de Queiroz EAIF, Akamine EH, de Carvalho MHC, Sampaio SC, Fortes ZB. Metformin reduces the Walker-256 tumor development in obese-MSG rats via AMPK and FOXO3a. Life Sci 2014; 121:78-87. [PMID: 25497710 DOI: 10.1016/j.lfs.2014.11.028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/11/2014] [Accepted: 11/24/2014] [Indexed: 11/24/2022]
Abstract
AIMS Studies have associated obesity with a wide variety of cancers. Metformin, an anti-diabetic drug, has recently received attention as a potentially useful therapeutic agent for treating cancer. Therefore, the objective of this study was to analyze the mechanisms involved in the increase in tumor development and the reduction of it by metformin in obesity using an experimental breast tumor model. MATERIAL AND METHODS Newborn male Wistar rats were subcutaneously injected with 400mg/kg monosodium glutamate (MSG) (obese) or saline (control) at 2, 3, 4, 5 and 6 days of age. After 16 weeks, 1 × 10(7) Walker-256 tumor cells were subcutaneously injected in the right flank of the rats and concomitantly the treatment with metformin 300 mg/kg/15 days, via gavage, started. The rats were divided into 4 groups: control tumor (CT), control tumor metformin (CTM), obese-MSG tumor (OT) and obese-MSG tumor metformin (OTM). On the 18th week the tumor development and metformin effect were analyzed. KEY FINDINGS Tumor development was higher in OT rats compared with CT rats. Activation of insulin-IR-ERK1/2 pathway and an anti-apoptotic effect might be the mechanisms involved in the higher development of tumor in obesity. The effect of metformin reducing the tumor development in obese rats might involve increased mRNA expression of pRb and p27, increased activity of AMPK and FOXO3a and decreased expression of p-ERK1/2 (Thr202/Tyr204) in Walker-256 tumor. SIGNIFICANCE Our data allow us to suggest that metformin, reducing the stimulatory effect of obesity on tumor development, has a potential role in the management of cancers.
Collapse
Affiliation(s)
- Eveline A I F de Queiroz
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, Brazil.
| | - Eliana H Akamine
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Maria Helena C de Carvalho
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Sandra C Sampaio
- Laboratory of Pathophysiology, Butantan Institute, São Paulo, Brazil
| | - Zuleica B Fortes
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|
42
|
Lamch L, Bazylińska U, Kulbacka J, Pietkiewicz J, Bieżuńska-Kusiak K, Wilk KA. Polymeric micelles for enhanced Photofrin II ® delivery, cytotoxicity and pro-apoptotic activity in human breast and ovarian cancer cells. Photodiagnosis Photodyn Ther 2014; 11:570-85. [PMID: 25449154 DOI: 10.1016/j.pdpdt.2014.10.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 10/13/2014] [Accepted: 10/14/2014] [Indexed: 11/26/2022]
Abstract
BACKGROUND Searching for photodynamic therapy (PDT) - effective nanocarriers which enable a photosensitizer to be selectively delivered to tumor cells with enhanced bioavailability and diminished dark cytotoxicity is of current interest. The main objective of this study is to evaluate newly designed mixed polymeric micelles based on Pluronics P123 and F127 for the improved delivery of Photofrin II(®) (Ph II(®)) to circumvent unfavorable effects overcoming multidrug resistance (MDR) in tumor cells - in breast MCF-7/WT (caspase-3 deficient) and ovarian SKOV-3 (resistant to chemotherapy). METHODS Ph II(®)-loaded micelles were obtained and analyzed for size and morphology, solubilization efficiency, physical stability and in vitro drug release. Intracellular uptake, reactive oxygen species (ROS) generation, mitochondrial oxidoreductive potential and proapoptotic activity (TUNEL assay) studies were evaluated in the examined cancer cells. The preliminary biocompatibility characteristics of all nanocarriers was determined by assessment of their hemolytic activity in human erythrocytes and dark toxicity in cancer cells. RESULTS Dynamic light scattering (DLS) and atomic force microscopy (AFM) confirmed that almost monodisperse, sphere-shaped and nanosized (DH<20 nm) carriers were developed. Biological studies after photodynamic reaction (PDR) with encapsulated Ph II(®) revealed increased ROS level, malondialdehyde (MDA) concentration and protein damage in SKOV-3 and MCF-7/WT cells in comparison to treatment with free Ph II(®). Numerous apoptotic cells were detected after nano-therapy in both cell lines, with observed significant morphological disorders in ovarian cancer cells. In the case of encapsulated Ph II(®) only negligible disruption of human erythrocytes and cancer cells was observed. CONCLUSIONS The obtained biocompatible long-lasting nanocarriers significantly enhance the Photofrin II(®) photodynamic effect and apoptosis in both SKOV-3 and MCF-7/WT cell lines.
Collapse
Affiliation(s)
- Lukasz Lamch
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Urszula Bazylińska
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| | - Julita Kulbacka
- Department of Medical Biochemistry, Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland.
| | - Jadwiga Pietkiewicz
- Department of Medical Biochemistry, Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland
| | | | - Kazimiera A Wilk
- Department of Organic and Pharmaceutical Technology, Faculty of Chemistry, Wroclaw University of Technology, Wybrzeże Wyspiańskiego 27, 50-370 Wrocław, Poland
| |
Collapse
|
43
|
Lysosomotropic agents: impact on lysosomal membrane permeabilization and cell death. Biochem Soc Trans 2014; 42:1460-4. [DOI: 10.1042/bst20140145] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lysosomes are acidic organelles essential for degradation, signalling and cell homoeostasis. In addition, they play a key role in cell death. Permeabilization of the lysosomal membrane and release of hydrolytic enzymes to the cytosol accompanies apoptosis signalling in several systems. The regulatory mechanism of lysosomal stability is, however, poorly understood. Lipophilic or amphiphilic compounds with a basic moiety will become protonated and trapped within lysosomes, and such lysosomotropic behaviour is also found in many pharmacological drugs. The natural sphingolipid sphingosine exhibits lysosomotropic detergent ability and is an endogenous candidate for controlling lysosomal membrane permeabilization. The lysosomotropic properties of certain detergents might be of use in lysosome-targeting anticancer drugs and drug delivery system in the future. The present review summarizes the current knowledge on the targeting and permeabilizing properties of lysosomotropic detergents from a cellular and physicochemical perspective.
Collapse
|
44
|
Boselli L, Ader I, Carraz M, Hemmert C, Cuvillier O, Gornitzka H. Synthesis, structures, and selective toxicity to cancer cells of gold(I) complexes involving N-heterocyclic carbene ligands. Eur J Med Chem 2014; 85:87-94. [PMID: 25078312 DOI: 10.1016/j.ejmech.2014.07.086] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 07/21/2014] [Accepted: 07/24/2014] [Indexed: 01/10/2023]
Abstract
New gold(I) complexes containing two 1-[2-(diethylamino)ethyl]imidazolydene ligands have been synthesized and characterized. The X-ray structures of two key compounds are presented. All complexes have been tested for their antiproliferative activities in prostate cancer cell line PC-3. Lipophilicity (Log P) has been determined for these complexes. The most active complex has been tested for the cytotoxic activities in five human cancer cell lines and primary endothelial cells. The most active complex demonstrated a potent selectivity for cancer cells.
Collapse
Affiliation(s)
- Luca Boselli
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, F-31077 Toulouse cedex 4, France; Université de Toulouse, UPS, INP, LCC, F-31077 Toulouse, France
| | - Isabelle Ader
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France
| | - Maëlle Carraz
- Université de Toulouse, UPS, UMR 152 Pharma-DEV, Université Toulouse 3, Faculté des sciences pharmaceutiques, 35 Chemin des maraîchers, F-31062 Toulouse cedex 9, France; Institut de Recherche pour le Développement, IRD, UMR 152 Pharma-DEV, F-31062 Toulouse cedex 9, France
| | - Catherine Hemmert
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, F-31077 Toulouse cedex 4, France; Université de Toulouse, UPS, INP, LCC, F-31077 Toulouse, France.
| | - Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, UPS, IPBS, Toulouse, France.
| | - Heinz Gornitzka
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, BP 44099, F-31077 Toulouse cedex 4, France; Université de Toulouse, UPS, INP, LCC, F-31077 Toulouse, France.
| |
Collapse
|
45
|
Queiroz EAIF, Puukila S, Eichler R, Sampaio SC, Forsyth HL, Lees SJ, Barbosa AM, Dekker RFH, Fortes ZB, Khaper N. Metformin induces apoptosis and cell cycle arrest mediated by oxidative stress, AMPK and FOXO3a in MCF-7 breast cancer cells. PLoS One 2014; 9:e98207. [PMID: 24858012 PMCID: PMC4032293 DOI: 10.1371/journal.pone.0098207] [Citation(s) in RCA: 205] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 04/30/2014] [Indexed: 12/23/2022] Open
Abstract
Recent studies have demonstrated that the anti-diabetic drug, metformin, can exhibit direct antitumoral effects, or can indirectly decrease tumor proliferation by improving insulin sensitivity. Despite these recent advances, the underlying molecular mechanisms involved in decreasing tumor formation are not well understood. In this study, we examined the antiproliferative role and mechanism of action of metformin in MCF-7 cancer cells treated with 10 mM of metformin for 24, 48, and 72 hours. Using BrdU and the MTT assay, it was found that metformin demonstrated an antiproliferative effect in MCF-7 cells that occurred in a time- and concentration- dependent manner. Flow cytometry was used to analyze markers of cell cycle, apoptosis, necrosis and oxidative stress. Exposure to metformin induced cell cycle arrest in G0-G1 phase and increased cell apoptosis and necrosis, which were associated with increased oxidative stress. Gene and protein expression were determined in MCF-7 cells by real time RT-PCR and western blotting, respectively. In MCF-7 cells metformin decreased the activation of IRβ, Akt and ERK1/2, increased p-AMPK, FOXO3a, p27, Bax and cleaved caspase-3, and decreased phosphorylation of p70S6K and Bcl-2 protein expression. Co-treatment with metformin and H2O2 increased oxidative stress which was associated with reduced cell number. In the presence of metformin, treating with SOD and catalase improved cell viability. Treatment with metformin resulted in an increase in p-p38 MAPK, catalase, MnSOD and Cu/Zn SOD protein expression. These results show that metformin has an antiproliferative effect associated with cell cycle arrest and apoptosis, which is mediated by oxidative stress, as well as AMPK and FOXO3a activation. Our study further reinforces the potential benefit of metformin in cancer treatment and provides novel mechanistic insight into its antiproliferative role.
Collapse
Affiliation(s)
- Eveline A. I. F. Queiroz
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Paraná, Brazil
| | - Stephanie Puukila
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
| | - Rosangela Eichler
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Sandra C. Sampaio
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Heidi L. Forsyth
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
| | - Simon J. Lees
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
| | - Aneli M. Barbosa
- Biorefining Research Institute, Lakehead University, Thunder Bay, Ontario, Canada
| | - Robert F. H. Dekker
- Biorefining Research Institute, Lakehead University, Thunder Bay, Ontario, Canada
| | - Zuleica B. Fortes
- Pharmacology Department, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
- * E-mail: (ZBF); (NK)
| | - Neelam Khaper
- Northern Ontario School of Medicine, Lakehead University, Thunder Bay, Ontario, Canada
- Biology Department, Lakehead University, Thunder Bay, Ontario, Canada
- * E-mail: (ZBF); (NK)
| |
Collapse
|
46
|
Calderon MR, Verway M, Benslama RO, Birlea M, Bouttier M, Dimitrov V, Mader S, White JH. Ligand-dependent corepressor contributes to transcriptional repression by C2H2 zinc-finger transcription factor ZBRK1 through association with KRAB-associated protein-1. Nucleic Acids Res 2014; 42:7012-27. [PMID: 24829459 PMCID: PMC4066800 DOI: 10.1093/nar/gku413] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
We identified a novel interaction between ligand-dependent corepressor (LCoR) and the corepressor KRAB-associated protein-1 (KAP-1). The two form a complex with C2H2 zinc-finger transcription factor ZBRK1 on an intronic binding site in the growth arrest and DNA-damage-inducible α (GADD45A) gene and a novel site in the fibroblast growth factor 2 (FGF2) gene. Chromatin at both sites is enriched for histone methyltransferase SETDB1 and histone 3 lysine 9 trimethylation, a repressive epigenetic mark. Depletion of ZBRK1, KAP-1 or LCoR led to elevated GADD45A and FGF2 expression in malignant and non-malignant breast epithelial cells, and caused apoptotic death. Loss of viability could be rescued by simultaneous knockdowns of FGF2 and transcriptional coregulators or by blocking FGF2 function. FGF2 was not concurrently expressed with any of the transcriptional coregulators in breast malignancies, suggesting an inverse correlation between their expression patterns. We propose that ZBRK1, KAP-1 and LCoR form a transcriptional complex that silences gene expression, in particular FGF2, which maintains breast cell viability. Given the broad expression patterns of both LCoR and KAP-1 during development and in the adult, this complex may have several regulatory functions that extend beyond cell survival, mediated by interactions with ZBRK1 or other C2H2 zinc-finger proteins.
Collapse
Affiliation(s)
- Mario R Calderon
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Mark Verway
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Radia Ouelaa Benslama
- Department of Biochemistry, Université de Montréal, Montreal, QC, Canada Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montreal, QC, Canada
| | - Mirela Birlea
- Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montreal, QC, Canada
| | | | - Vassil Dimitrov
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Sylvie Mader
- Department of Biochemistry, Université de Montréal, Montreal, QC, Canada Institut de Recherche en Immunologie et Cancérologie (IRIC), Université de Montréal, Montreal, QC, Canada
| | - John H White
- Department of Physiology, McGill University, Montreal, QC, Canada Department of Medicine, McGill University, Montreal, QC, Canada
| |
Collapse
|
47
|
Abstract
The signaling pathways activated by the steroid hormone oestrogen include a variety of cytoplasmic second messengers linked to a multitude of tissue-specific effects. In the last decade, sphingolipids and their membrane receptors were added to the list of oestrogen-activated mediators. Oestrogen triggers the sphingolipid signalling cascade in various tissues including breast cancer. Extensive research has shown that sphingolipids are the key regulatory molecules in growth factor networks. Sphingolipids can control the rate of cell proliferation and the differentiation outcome during malignant transformation. In this study, we summarise novel experimental evidences linking sphingolipids to oestrogen-activated effects, highlight the role of sphingolipids in cancer cells and discuss new avenues for future research at the intersection between oestrogen and sphingolipid signalling.
Collapse
Affiliation(s)
- O Sukocheva
- Division of Surgery, Flinders University of South Australia, Bedford Park, South Australia 5042, Australia Children's Cancer Institute Australia, University of New South Wales, Sydney, New South Wales, Australia
| | | |
Collapse
|
48
|
Caspase-3 and RasGAP: a stress-sensing survival/demise switch. Trends Cell Biol 2013; 24:83-9. [PMID: 24007977 DOI: 10.1016/j.tcb.2013.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
The final decision on cell fate, survival versus cell death, relies on complex and tightly regulated checkpoint mechanisms. The caspase-3 protease is a predominant player in the execution of apoptosis. However, recent progress has shown that this protease paradoxically can also protect cells from death. Here, we discuss the underappreciated, protective, and prosurvival role of caspase-3 and detail the evidence showing that caspase-3, through differential processing of p120 Ras GTPase-activating protein (RasGAP), can modulate a given set of proteins to generate, depending on the intensity of the input signals, opposite outcomes (survival vs death).
Collapse
|
49
|
Cuvillier O, Ader I, Bouquerel P, Brizuela L, Gstalder C, Malavaud B. Hypoxia, therapeutic resistance, and sphingosine 1-phosphate. Adv Cancer Res 2013; 117:117-41. [PMID: 23290779 DOI: 10.1016/b978-0-12-394274-6.00005-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Hypoxia, defined as a poor oxygenation, has been long recognized as a hallmark of solid tumors and a negative prognostic factor for response to therapeutics and survival of patients. Cancer cells have evolved biochemical mechanisms that allow them to react and adapt to hypoxia. At the cellular level, this adaptation is under the control of two related transcription factors, HIF-1 and HIF-2 (hypoxia-inducible factor), that respond rapidly to decreased oxygen levels to activate the expression of a broad range of genes promoting neoangiogenesis, glycolysis, metastasis, increased tumor growth, and resistance to treatments. Recent studies have identified the sphingosine kinase 1/sphingosine 1-phosphate (SphK1/S1P) signaling pathway-which elicits various cellular processes including cell proliferation, cell survival, or angiogenesis-as a new regulator of HIF-1 or HIF-2 activity. In this review, we will focus on how the inhibition/neutralization of the SphK1/S1P signaling could be exploited for cancer therapy.
Collapse
Affiliation(s)
- Olivier Cuvillier
- CNRS, Institut de Pharmacologie et de Biologie Structurale, Toulouse, France; Université de Toulouse, Toulouse, France.
| | | | | | | | | | | |
Collapse
|
50
|
Young MM, Kester M, Wang HG. Sphingolipids: regulators of crosstalk between apoptosis and autophagy. J Lipid Res 2012; 54:5-19. [PMID: 23152582 DOI: 10.1194/jlr.r031278] [Citation(s) in RCA: 283] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Apoptosis and autophagy are two evolutionarily conserved processes that maintain homeostasis during stress. Although the two pathways utilize fundamentally distinct machinery, apoptosis and autophagy are highly interconnected and share many key regulators. The crosstalk between apoptosis and autophagy is complex, as autophagy can function to promote cell survival or cell death under various cellular conditions. The molecular mechanisms of crosstalk are beginning to be elucidated and have critical implications for the treatment of various diseases, such as cancer. Sphingolipids are a class of bioactive lipids that mediate many key cellular processes, including apoptosis and autophagy. By targeting several of the shared regulators, sphingolipid metabolites differentially regulate the induction of apoptosis and autophagy. Importantly, individual sphingolipid species appear to "switch" autophagy toward cell survival (e.g., sphingosine-1-phosphate) or cell death (e.g., ceramide, gangliosides). This review assesses the current understanding of sphingolipid-induced apoptosis and autophagy to address how sphingolipids mediate the "switch" between the cell survival and cell death. As sphingolipid metabolism is frequently dysregulated in cancer, sphingolipid-modulating agents, or sphingomimetics, have emerged as a novel chemotherapeutic strategy. Ultimately, a greater understanding of sphingolipid-mediated crosstalk between apoptosis and autophagy may be critical for enhancing the chemotherapeutic efficacy of these agents.
Collapse
Affiliation(s)
- Megan M Young
- Department of Pharmacology and Penn State Hershey Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | | | | |
Collapse
|