1
|
Rivero P, Ivanova V, Barril X, Casampere M, Casas J, Fabriàs G, Díaz Y, Matheu MI. Targeting dihydroceramide desaturase 1 (Des1): Syntheses of ceramide analogues with a rigid scaffold, inhibitory assays, and AlphaFold2-assisted structural insights reveal cyclopropenone PR280 as a potent inhibitor. Bioorg Chem 2024; 145:107233. [PMID: 38422591 DOI: 10.1016/j.bioorg.2024.107233] [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: 10/09/2023] [Revised: 01/04/2024] [Accepted: 02/19/2024] [Indexed: 03/02/2024]
Abstract
Dihydroceramide desaturase 1 (Des1) catalyzes the formation of a CC double bond in dihydroceramide to furnish ceramide. Inhibition of Des1 is related to cell cycle arrest and programmed cell death. The lack of the Des1 crystalline structure, as well as that of a close homologue, hampers the detailed understanding of its inhibition mechanism and difficults the design of new inhibitors, thus making Des1 a strategic target. Based on previous structure-activity studies, different ceramides containing rigid scaffolds were designed. The synthesis and evaluation of these compounds as Des1 inhibitors allowed the identification of PR280 as a better Des 1 inhibitor in vitro (IC50 = 700 nM) than GT11 and XM462, the current reference inhibitors. This cyclopropenone ceramide was obtained in a 6-step synthesis with a 24 % overall yield. The highly confident 3D structure of Des1, recently predicted by AlphaFold2, served as the basis for conducting docking studies of known Des1 inhibitors and the ceramide derivatives synthesized by us in this study. For this purpose, a complete holoprotein structure was previously constructed. This study has allowed a better knowledge of key ligand-enzyme interactions for Des1 inhibitory activity. Furthermore, it sheds some light on the inhibition mechanism of GT11.
Collapse
Affiliation(s)
- Pablo Rivero
- Universitat Rovira i Virgili, Departament de Química Analítica i Química Orgànica, Faculty of Chemistry, C/Marcel.lí Domingo 1, Tarragona 43007, Spain
| | - Varbina Ivanova
- Universitat de Barcelona, Department of Physical Chemistry, Faculty of Pharmacy, Av. Joan XXIII s/n, Barcelona 08028, Spain
| | - Xavier Barril
- Universitat de Barcelona, Department of Physical Chemistry, Faculty of Pharmacy, Av. Joan XXIII s/n, Barcelona 08028, Spain
| | - Mireia Casampere
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, C/Jordi Girona 18-26, Barcelona 08034, Spain
| | - Josefina Casas
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, C/Jordi Girona 18-26, Barcelona 08034, Spain
| | - Gemma Fabriàs
- Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Department of Biological Chemistry, C/Jordi Girona 18-26, Barcelona 08034, Spain
| | - Yolanda Díaz
- Universitat Rovira i Virgili, Departament de Química Analítica i Química Orgànica, Faculty of Chemistry, C/Marcel.lí Domingo 1, Tarragona 43007, Spain.
| | - M Isabel Matheu
- Universitat Rovira i Virgili, Departament de Química Analítica i Química Orgànica, Faculty of Chemistry, C/Marcel.lí Domingo 1, Tarragona 43007, Spain.
| |
Collapse
|
2
|
Rezaul Islam M, Rauf A, Akash S, Kumer A, Hussain MS, Akter S, Gupta JK, Thameemul Ansari L, Mahfoj Islam Raj MM, Bin Emran T, Aljohani AS, Abdulmonem WA, Thiruvengadam R, Thiruvengadam M. Recent perspective on the potential role of phytocompounds in the prevention of gastric cancer. Process Biochem 2023; 135:83-101. [DOI: 10.1016/j.procbio.2023.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
|
3
|
Afrin F, Mateen S, Oman J, Lai JCK, Barrott JJ, Pashikanti S. Natural Products and Small Molecules Targeting Cellular Ceramide Metabolism to Enhance Apoptosis in Cancer Cells. Cancers (Basel) 2023; 15:4645. [PMID: 37760612 PMCID: PMC10527029 DOI: 10.3390/cancers15184645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/08/2023] [Accepted: 09/11/2023] [Indexed: 09/29/2023] Open
Abstract
Molecular targeting strategies have been used for years in order to control cancer progression and are often based on targeting various enzymes involved in metabolic pathways. Keeping this in mind, it is essential to determine the role of each enzyme in a particular metabolic pathway. In this review, we provide in-depth information on various enzymes such as ceramidase, sphingosine kinase, sphingomyelin synthase, dihydroceramide desaturase, and ceramide synthase which are associated with various types of cancers. We also discuss the physicochemical properties of well-studied inhibitors with natural product origins and their related structures in terms of these enzymes. Targeting ceramide metabolism exhibited promising mono- and combination therapies at preclinical stages in preventing cancer progression and cemented the significance of sphingolipid metabolism in cancer treatments. Targeting ceramide-metabolizing enzymes will help medicinal chemists design potent and selective small molecules for treating cancer progression at various levels.
Collapse
Affiliation(s)
- Farjana Afrin
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Sameena Mateen
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Jordan Oman
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - James C. K. Lai
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| | - Jared J. Barrott
- Cell Biology and Physiology, College of Life Sciences, Brigham Young University, Provo, UT 84602, USA;
| | - Srinath Pashikanti
- Biomedical and Pharmaceutical Sciences, Kasiska Division of Health Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (F.A.); (S.M.); (J.O.); (J.C.K.L.)
| |
Collapse
|
4
|
Pharmacological Elevation of Cellular Dihydrosphingomyelin Provides a Novel Antiviral Strategy against West Nile Virus Infection. Antimicrob Agents Chemother 2023; 67:e0168722. [PMID: 36920206 PMCID: PMC10112131 DOI: 10.1128/aac.01687-22] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
The flavivirus life cycle is strictly dependent on cellular lipid metabolism. Polyphenols like gallic acid and its derivatives are promising lead compounds for new therapeutic agents as they can exert multiple pharmacological activities, including the alteration of lipid metabolism. The evaluation of our collection of polyphenols against West Nile virus (WNV), a representative medically relevant flavivirus, led to the identification of N,N'-(dodecane-1,12-diyl)bis(3,4,5-trihydroxybenzamide) and its 2,3,4-trihydroxybenzamide regioisomer as selective antivirals with low cytotoxicity and high antiviral activity (half-maximal effective concentrations [EC50s] of 2.2 and 0.24 μM, respectively, in Vero cells; EC50s of 2.2 and 1.9 μM, respectively, in SH-SY5Y cells). These polyphenols also inhibited the multiplication of other flaviviruses, namely, Usutu, dengue, and Zika viruses, exhibiting lower antiviral or negligible antiviral activity against other RNA viruses. The mechanism underlying their antiviral activity against WNV involved the alteration of sphingolipid metabolism. These compounds inhibited ceramide desaturase (Des1), promoting the accumulation of dihydrosphingomyelin (dhSM), a minor component of cellular sphingolipids with important roles in membrane properties. The addition of exogenous dhSM or Des1 blockage by using the reference inhibitor GT-11 {N-[(1R,2S)-2-hydroxy-1-hydroxymethyl-2-(2-tridecyl-1-cyclopropenyl)ethyl]octanamide} confirmed the involvement of this pathway in WNV infection. These results unveil the potential of novel antiviral strategies based on the modulation of the cellular levels of dhSM and Des1 activity for the control of flavivirus infection.
Collapse
|
5
|
Chowdhury MR, Jin HK, Bae JS. Diverse Roles of Ceramide in the Progression and Pathogenesis of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10081956. [PMID: 36009503 PMCID: PMC9406151 DOI: 10.3390/biomedicines10081956] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/31/2022] [Accepted: 08/02/2022] [Indexed: 11/26/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common neurodegenerative disorder, and is associated with several pathophysiological features, including cellular dysfunction, failure of neurotransmission, cognitive impairment, cell death, and other clinical consequences. Advanced research on the pathogenesis of AD has elucidated a mechanistic framework and revealed many therapeutic possibilities. Among the mechanisms, sphingolipids are mentioned as distinctive mediators to be associated with the pathology of AD. Reportedly, alteration in the metabolism of sphingolipids and their metabolites result in the dysfunction of mitochondria, autophagy, amyloid beta regulation, and neuronal homeostasis, which exacerbates AD progression. Considering the importance of sphingolipids, in this review, we discuss the role of ceramide, a bioactive sphingolipid metabolite, in the progression and pathogenesis of AD. Herein, we describe the ceramide synthesis pathway and its involvement in the dysregulation of homeostasis, which eventually leads to AD. Furthermore, this review references different therapeutics proposed to modulate the ceramide pathway to maintain ceramide levels and prevent the disease progression.
Collapse
Affiliation(s)
- Md Riad Chowdhury
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Hee Kyung Jin
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Laboratory Animal Medicine, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Korea
- Correspondence: (H.K.J.); (J.-s.B.); Tel.: +82-53-950-5966 (H.K.J.); +82-53-420-4815 (J.-s.B.); Fax: +82-53-950-5955 (H.K.J.); +82-53-424-3349 (J.-s.B.)
| | - Jae-sung Bae
- KNU Alzheimer’s Disease Research Institute, Kyungpook National University, Daegu 41566, Korea
- Department of Physiology, School of Medicine, Kyungpook National University, Daegu 41944, Korea
- Correspondence: (H.K.J.); (J.-s.B.); Tel.: +82-53-950-5966 (H.K.J.); +82-53-420-4815 (J.-s.B.); Fax: +82-53-950-5955 (H.K.J.); +82-53-424-3349 (J.-s.B.)
| |
Collapse
|
6
|
Song JH, Kim GT, Park KH, Park WJ, Park TS. Bioactive Sphingolipids as Major Regulators of Coronary Artery Disease. Biomol Ther (Seoul) 2021; 29:373-383. [PMID: 33903284 PMCID: PMC8255146 DOI: 10.4062/biomolther.2020.218] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/31/2022] Open
Abstract
Atherosclerosis is the deposition of plaque in the main arteries. It is an inflammatory condition involving the accumulation of macrophages and various lipids (low-density lipoprotein [LDL] cholesterol, ceramide, S1P). Moreover, endothelial cells, macrophages, leukocytes, and smooth muscle cells are the major players in the atherogenic process. Sphingolipids are now emerging as important regulators in various pathophysiological processes, including the atherogenic process. Various sphingolipids exist, such as the ceramides, ceramide-1-phosphate, sphingosine, sphinganine, sphingosine-1-phosphate (S1P), sphingomyelin, and hundreds of glycosphingolipids. Among these, ceramides, glycosphingolipids, and S1P play important roles in the atherogenic processes. The atherosclerotic plaque consists of higher amounts of ceramide, glycosphingolipids, and sphingomyelin. The inhibition of the de novo ceramide biosynthesis reduces the development of atherosclerosis. S1P regulates atherogenesis via binding to the S1P receptor (S1PR). Among the five S1PRs (S1PR1-5), S1PR1 and S1PR3 mainly exert anti-atherosclerotic properties. This review mainly focuses on the effects of ceramide and S1P via the S1PR in the development of atherosclerosis. Moreover, it discusses the recent findings and potential therapeutic implications in atherosclerosis.
Collapse
Affiliation(s)
- Jae-Hwi Song
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Goon-Tae Kim
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| | - Kyung-Ho Park
- Department of Nutrition, Hallym University, Chuncheon 24252, Republic of Korea
| | - Woo-Jae Park
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Tae-Sik Park
- Department of Life Science, Gachon University, Sungnam 13120, Republic of Korea
| |
Collapse
|
7
|
Bielsa N, Casasampere M, Aseeri M, Casas J, Delgado A, Abad JL, Fabriàs G. Discovery of deoxyceramide analogs as highly selective ACER3 inhibitors in live cells. Eur J Med Chem 2021; 216:113296. [PMID: 33677352 DOI: 10.1016/j.ejmech.2021.113296] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/27/2021] [Accepted: 02/12/2021] [Indexed: 12/13/2022]
Abstract
Acid (AC), neutral (NC) and alkaline ceramidase 3 (ACER3) are the most ubiquitous ceramidases and their therapeutic interest as targets in cancer diseases has been well sustained. This supports the importance of discovering potent and specific inhibitors for further use in combination therapies. Although several ceramidase inhibitors have been reported, most of them target AC and a few focus on NC. In contrast, well characterized ACER3 inhibitors are lacking. Here we report on the synthesis and screening of two series of 1-deoxy(dihydro)ceramide analogs on the three enzymes. Activity was determined using fluorogenic substrates in recombinant human NC (rhNC) and both lysates and intact cells enriched in each enzyme. None of the molecules elicited a remarkable AC inhibitory activity in either experimental setup, while using rhNC, several compounds of both series were active as non-competitive inhibitors with Ki values between 1 and 5 μM. However, a dramatic loss of potency occurred in NC-enriched cell lysates and no activity was elicited in intact cells. Interestingly, several compounds of Series 2 inhibited ACER3 dose-dependently in both cell lysates and intact cells with IC50's around 20 μM. In agreement with their activity in live cells, they provoked a significant increase in the amounts of ceramides. Overall, this study identifies highly selective ACER3 activity blockers in intact cells, opening the door to further medicinal chemistry efforts aimed at developing more potent and specific compounds.
Collapse
Affiliation(s)
- Núria Bielsa
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Mireia Casasampere
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Mazen Aseeri
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII, 28029, Madrid, Spain
| | - Antonio Delgado
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Department of Pharmacology, Toxicology and Medicinal Chemistry, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC). Faculty of Pharmacy. University of Barcelona (UB). Avda. Joan XXIII 27-31, 08028, Barcelona, Spain
| | - José Luis Abad
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain.
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules, Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18, 08034, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD), ISCIII, 28029, Madrid, Spain.
| |
Collapse
|
8
|
Skácel J, Slusher BS, Tsukamoto T. Small Molecule Inhibitors Targeting Biosynthesis of Ceramide, the Central Hub of the Sphingolipid Network. J Med Chem 2021; 64:279-297. [PMID: 33395289 PMCID: PMC8023021 DOI: 10.1021/acs.jmedchem.0c01664] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Ceramides are composed of a sphingosine and a single fatty acid connected by an amide linkage. As one of the major classes of biologically active lipids, ceramides and their upstream and downstream metabolites have been implicated in several pathological conditions including cancer, neurodegeneration, diabetes, microbial pathogenesis, obesity, and inflammation. Consequently, tremendous efforts have been devoted to deciphering the dynamics of metabolic pathways involved in ceramide biosynthesis. Given that several distinct enzymes can produce ceramide, different enzyme targets have been pursued depending on the underlying disease mechanism. The main objective of this review is to provide a comprehensive overview of small molecule inhibitors reported to date for each of these ceramide-producing enzymes from a medicinal chemistry perspective.
Collapse
Affiliation(s)
- Jan Skácel
- Johns Hopkins Drug Discovery and Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Barbara S. Slusher
- Johns Hopkins Drug Discovery and Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| | - Takashi Tsukamoto
- Johns Hopkins Drug Discovery and Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21205, United States
| |
Collapse
|
9
|
Martín‐Leal A, Blanco R, Casas J, Sáez ME, Rodríguez‐Bovolenta E, de Rojas I, Drechsler C, Real LM, Fabrias G, Ruíz A, Castro M, Schamel WWA, Alarcón B, van Santen HM, Mañes S. CCR5 deficiency impairs CD4 + T-cell memory responses and antigenic sensitivity through increased ceramide synthesis. EMBO J 2020; 39:e104749. [PMID: 32525588 PMCID: PMC7396835 DOI: 10.15252/embj.2020104749] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022] Open
Abstract
CCR5 is not only a coreceptor for HIV-1 infection in CD4+ T cells, but also contributes to their functional fitness. Here, we show that by limiting transcription of specific ceramide synthases, CCR5 signaling reduces ceramide levels and thereby increases T-cell antigen receptor (TCR) nanoclustering in antigen-experienced mouse and human CD4+ T cells. This activity is CCR5-specific and independent of CCR5 co-stimulatory activity. CCR5-deficient mice showed reduced production of high-affinity class-switched antibodies, but only after antigen rechallenge, which implies an impaired memory CD4+ T-cell response. This study identifies a CCR5 function in the generation of CD4+ T-cell memory responses and establishes an antigen-independent mechanism that regulates TCR nanoclustering by altering specific lipid species.
Collapse
Affiliation(s)
- Ana Martín‐Leal
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| | - Raquel Blanco
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| | - Josefina Casas
- Department of Biological ChemistryInstitute of Advanced Chemistry of Catalonia (IQAC‐CSIC)BarcelonaSpain
- CIBER Liver and Digestive Diseases (CIBER‐EDH)Instituto de Salud Carlos IIIMadridSpain
| | - María E Sáez
- Centro Andaluz de Estudios Bioinformáticos (CAEBi)SevilleSpain
| | - Elena Rodríguez‐Bovolenta
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Itziar de Rojas
- Alzheimer Research CenterMemory Clinic of the Fundació ACEInstitut Català de Neurociències AplicadesBarcelonaSpain
| | - Carina Drechsler
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
- Department of ImmunologyFaculty of BiologyUniversity of FreiburgFreiburgGermany
- Institute for Pharmaceutical SciencesUniversity of FreiburgFreiburgGermany
| | - Luis Miguel Real
- Unit of Infectious Diseases and MicrobiologyHospital Universitario de ValmeSevilleSpain
- Department of Biochemistry, Molecular Biology and ImmunologySchool of MedicineUniversidad de MálagaMálagaSpain
| | - Gemma Fabrias
- Department of Biological ChemistryInstitute of Advanced Chemistry of Catalonia (IQAC‐CSIC)BarcelonaSpain
- CIBER Liver and Digestive Diseases (CIBER‐EDH)Instituto de Salud Carlos IIIMadridSpain
| | - Agustín Ruíz
- Alzheimer Research CenterMemory Clinic of the Fundació ACEInstitut Català de Neurociències AplicadesBarcelonaSpain
- CIBER Enfermedades Neurodegenerativas (CIBERNED)Instituto de Salud Carlos IIIMadridSpain
| | - Mario Castro
- Interdisciplinary Group of Complex SystemsEscuela Técnica Superior de IngenieríaUniversidad Pontificia ComillasMadridSpain
| | - Wolfgang WA Schamel
- Signaling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
- Department of ImmunologyFaculty of BiologyUniversity of FreiburgFreiburgGermany
- Centre for Chronic Immunodeficiency (CCI)University of FreiburgFreiburgGermany
| | - Balbino Alarcón
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Hisse M van Santen
- Department of Cell Biology and ImmunologyCentro de Biología Molecular Severo Ochoa (CBMSO/CSIC)MadridSpain
| | - Santos Mañes
- Department of Immunology and OncologyCentro Nacional de Biotecnología (CNB/CSIC)MadridSpain
| |
Collapse
|
10
|
Muñoz-Guardiola P, Casas J, Megías-Roda E, Solé S, Perez-Montoyo H, Yeste-Velasco M, Erazo T, Diéguez-Martínez N, Espinosa-Gil S, Muñoz-Pinedo C, Yoldi G, Abad JL, Segura MF, Moran T, Romeo M, Bosch-Barrera J, Oaknin A, Alfón J, Domènech C, Fabriàs G, Velasco G, Lizcano JM. The anti-cancer drug ABTL0812 induces ER stress-mediated cytotoxic autophagy by increasing dihydroceramide levels in cancer cells. Autophagy 2020; 17:1349-1366. [PMID: 32397857 DOI: 10.1080/15548627.2020.1761651] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
ABTL0812 is a first-in-class small molecule with anti-cancer activity, which is currently in clinical evaluation in a phase 2 trial in patients with advanced endometrial and squamous non-small cell lung carcinoma (NCT03366480). Previously, we showed that ABTL0812 induces TRIB3 pseudokinase expression, resulting in the inhibition of the AKT-MTORC1 axis and macroautophagy/autophagy-mediated cancer cell death. However, the precise molecular determinants involved in the cytotoxic autophagy caused by ABTL0812 remained unclear. Using a wide range of biochemical and lipidomic analyses, we demonstrated that ABTL0812 increases cellular long-chain dihydroceramides by impairing DEGS1 (delta 4-desaturase, sphingolipid 1) activity, which resulted in sustained ER stress and activated unfolded protein response (UPR) via ATF4-DDIT3-TRIB3 that ultimately promotes cytotoxic autophagy in cancer cells. Accordingly, pharmacological manipulation to increase cellular dihydroceramides or incubation with exogenous dihydroceramides resulted in ER stress, UPR and autophagy-mediated cancer cell death. Importantly, we have optimized a method to quantify mRNAs in blood samples from patients enrolled in the ongoing clinical trial, who showed significant increased DDIT3 and TRIB3 mRNAs. This is the first time that UPR markers are reported to change in human blood in response to any drug treatment, supporting their use as pharmacodynamic biomarkers for compounds that activate ER stress in humans. Finally, we found that MTORC1 inhibition and dihydroceramide accumulation synergized to induce autophagy and cytotoxicity, phenocopying the effect of ABTL0812. Given the fact that ABTL0812 is under clinical development, our findings support the hypothesis that manipulation of dihydroceramide levels might represents a new therapeutic strategy to target cancer.Abbreviations: 4-PBA: 4-phenylbutyrate; AKT: AKT serine/threonine kinase; ATG: autophagy related; ATF4: activating transcription factor 4; Cer: ceramide; DDIT3: DNA damage inducible transcript 3; DEGS1: delta 4-desaturase, sphingolipid 1; dhCer: dihydroceramide; EIF2A: eukaryotic translation initiation factor 2 alpha; EIF2AK3: eukaryotic translation initiation factor 2 alpha kinase 3; ER: endoplasmic reticulum; HSPA5: heat shock protein family A (Hsp70) member 5; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; MEF: mouse embryonic fibroblast; MTORC1: mechanistic target of rapamycin kinase complex 1; NSCLC: non-small cell lung cancer; THC: Δ9-tetrahydrocannabinol; TRIB3: tribbles pseudokinase 3; XBP1: X-box binding protein 1; UPR: unfolded protein response.
Collapse
Affiliation(s)
- Pau Muñoz-Guardiola
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain.,Ability Pharmaceuticals, SL, Cerdanyola Del Vallès, Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD) ISCII, Madrid, Spain
| | - Elisabet Megías-Roda
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain.,Ability Pharmaceuticals, SL, Cerdanyola Del Vallès, Barcelona, Spain
| | - Sònia Solé
- Ability Pharmaceuticals, SL, Cerdanyola Del Vallès, Barcelona, Spain
| | | | | | - Tatiana Erazo
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| | - Nora Diéguez-Martínez
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| | - Sergio Espinosa-Gil
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| | - Cristina Muñoz-Pinedo
- Cell Death and Metabolism Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Guillermo Yoldi
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| | - Jose L Abad
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD) ISCII, Madrid, Spain
| | - Miguel F Segura
- Group of Translational Research in Child and Adolescent Cancer, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| | - Teresa Moran
- Medical Oncology Department, Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Universitat Autònoma de Barcelona, Applied Research Group in Oncology (B-ARGO), Badalona, Spain
| | - Margarita Romeo
- Medical Oncology Department, Catalan Institute of Oncology, Hospital Germans Trias I Pujol, Universitat Autònoma de Barcelona, Applied Research Group in Oncology (B-ARGO), Badalona, Spain
| | - Joaquim Bosch-Barrera
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), Dr. Josep Trueta University Hospital and Girona Biomedical Research Institute (IDIBGI), Girona, Spain
| | - Ana Oaknin
- Medical Oncology Department, Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose Alfón
- Ability Pharmaceuticals, SL, Cerdanyola Del Vallès, Barcelona, Spain
| | - Carles Domènech
- Ability Pharmaceuticals, SL, Cerdanyola Del Vallès, Barcelona, Spain
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules (RUBAM), Department of Biological Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona, Barcelona, Spain; Liver and Digestive Diseases Networking Biomedical Research Centre (CIBEREHD) ISCII, Madrid, Spain
| | - Guillermo Velasco
- Department of Biochemistry and Molecular Biology, School of Biology, Complutense University; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain
| | - Jose M Lizcano
- Protein Kinases and Signal Transduction Laboratory, Departament De Bioquímica I Biologia Molecular and Institut De Neurociències, Universitat Autònoma De Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
11
|
Wigger D, Gulbins E, Kleuser B, Schumacher F. Monitoring the Sphingolipid de novo Synthesis by Stable-Isotope Labeling and Liquid Chromatography-Mass Spectrometry. Front Cell Dev Biol 2019; 7:210. [PMID: 31632963 PMCID: PMC6779703 DOI: 10.3389/fcell.2019.00210] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022] Open
Abstract
Sphingolipids are a class of lipids that share a sphingoid base backbone. They exert various effects in eukaryotes, ranging from structural roles in plasma membranes to cellular signaling. De novo sphingolipid synthesis takes place in the endoplasmic reticulum (ER), where the condensation of the activated C16 fatty acid palmitoyl-CoA and the amino acid L-serine is catalyzed by serine palmitoyltransferase (SPT). The product, 3-ketosphinganine, is then converted into more complex sphingolipids by additional ER-bound enzymes, resulting in the formation of ceramides. Since sphingolipid homeostasis is crucial to numerous cellular functions, improved assessment of sphingolipid metabolism will be key to better understanding several human diseases. To date, no assay exists capable of monitoring de novo synthesis sphingolipid in its entirety. Here, we have established a cell-free assay utilizing rat liver microsomes containing all the enzymes necessary for bottom-up synthesis of ceramides. Following lipid extraction, we were able to track the different intermediates of the sphingolipid metabolism pathway, namely 3-ketosphinganine, sphinganine, dihydroceramide, and ceramide. This was achieved by chromatographic separation of sphingolipid metabolites followed by detection of their accurate mass and characteristic fragmentations through high-resolution mass spectrometry and tandem-mass spectrometry. We were able to distinguish, unequivocally, between de novo synthesized sphingolipids and intrinsic species, inevitably present in the microsome preparations, through the addition of stable isotope-labeled palmitate-d3 and L-serine-d3. To the best of our knowledge, this is the first demonstration of a method monitoring the entirety of ER-associated sphingolipid biosynthesis. Proof-of-concept data was provided by modulating the levels of supplied cofactors (e.g., NADPH) or the addition of specific enzyme inhibitors (e.g., fumonisin B1). The presented microsomal assay may serve as a useful tool for monitoring alterations in sphingolipid de novo synthesis in cells or tissues. Additionally, our methodology may be used for metabolism studies of atypical substrates - naturally occurring or chemically tailored - as well as novel inhibitors of enzymes involved in sphingolipid de novo synthesis.
Collapse
Affiliation(s)
- Dominik Wigger
- Department of Toxicology, University of Potsdam, Nuthetal, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany.,Department of Surgery, University of Cincinnati, Cincinnati, OH, United States
| | - Burkhard Kleuser
- Department of Toxicology, University of Potsdam, Nuthetal, Germany
| | - Fabian Schumacher
- Department of Toxicology, University of Potsdam, Nuthetal, Germany.,Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
12
|
Zhao XY, Wang JW, Yin Y, Li K, Zhang M, Yan FP. Effect of Tong Xie Yao Fang on endogenous metabolites in urine of irritable bowel syndrome model rats. World J Gastroenterol 2019; 25:5134-5151. [PMID: 31558862 PMCID: PMC6747287 DOI: 10.3748/wjg.v25.i34.5134] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 07/30/2019] [Accepted: 08/19/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tong Xie Yao Fang is a representative traditional Chinese prescription for the treatment of liver and spleen deficiency, abdominal pain and diarrhea. It has a unique function in the treatment of gastrointestinal dysfunction including irritable bowel syndrome (IBS), is a common functional bowel disease. Its main symptoms are recurrent abdominal pain, diarrhea, constipation or alternations between diarrhea and constipation.
There are obvious differences in metabolites between TCM syndromes. By comparing the body fluid metabolism maps of model animals, metabolomics can discover disease biomarkers, analyze the differences in metabolic pathways and understand the pathological process and the metabolic pathways of substances in the body. Thus, the evaluation of animal models tends to be comprehensive and objective. This may provide further understanding between the interaction between Tong Xie Yao Fang and the IBS model.
AIM To evaluate the effect of Tong Xie Yao Fang on IBS rats by using metabolomics method.
METHODS Wistar rats were used to establish IBS models, and then randomly divided into four groups: A model control group and three Tong Xie Yao Fang treatment groups (high, medium and low doses). A normal, non-IBS group was established. The rats were treated for 2 wk. On days 0 and 14 of the experimental model, urine was collected for 12 h and was analyzed by ultra-performance liquid chromatography quadrupole time-of-flight mass spectrometry. Nine potential biomarkers were identified, and six major metabolic pathways were found to be related to IBS.
RESULTS In the study of metabonomics, nine potential biomarkers including L-serine, 4-methylgallic acid, L-threonine, succinylacetone, prolyl-hydroxyproline, valyl-serine, acetyl citrate, marmesin rutinoside and 5-hydroxy-L-tryptophan were identified in urine, which were assigned to amino acids, organic acids, succinyl and glycosides. Furthermore, the metabolic pathway of L-serine, L-threonine and 5-hydroxy-L-tryptophan was found in the Kyoto Encyclopedia of Genes and Genomes, which mainly involved the metabolism of cysteine and methionine, vitamin B6 metabolism, serotonin synapse, tryptophan metabolism, sphingolipid metabolism, digestion, absorption of protein and amino acid metabolism. These pathways are related to intestinal dysfunction, inflammatory syndrome, nervous system dysfunction and other diseases.
CONCLUSION Tong Xie Yao Fang has pharmacological effects on IBS, and its mechanism may be related to the metabolism of the nine potential biomarkers identified above in urine.
Collapse
Affiliation(s)
- Xue-Ying Zhao
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| | - Jian-Wei Wang
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| | - Yue Yin
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| | - Kai Li
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| | - Miao Zhang
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| | - Fu-Ping Yan
- School of Basic Medical Sciences, Heilongjiang University of Chinese Medicine, Harbin 150040, Heilongjiang Province, China
| |
Collapse
|
13
|
Snider JM, Luberto C, Hannun YA. Approaches for probing and evaluating mammalian sphingolipid metabolism. Anal Biochem 2019; 575:70-86. [PMID: 30917945 DOI: 10.1016/j.ab.2019.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 01/02/2023]
Abstract
Sphingolipid metabolism plays a critical role in regulating processes that control cellular fate. This dynamic pathway can generate and degrade the central players: ceramide, sphingosine and sphingosine-1-phosphate in almost any membrane in the cell, adding an unexpected level of complexity in deciphering signaling events. While in vitro assays have been developed for most enzymes in SL metabolism, these assays are setup for optimal activity conditions and can fail to take into account regulatory components such as compartmentalization, substrate limitations, and binding partners that can affect cellular enzymatic activity. Therefore, many in-cell assays have been developed to derive results that are authentic to the cellular situation which may give context to alteration in SL mass. This review will discuss approaches for utilizing probes for mammalian in-cell assays to interrogate most enzymatic steps central to SL metabolism. The use of inhibitors in conjunction with these probes can verify the specificity of cellular assays as well as provide valuable insight into flux in the SL network. The use of inhibitors specific to each of the central sphingolipid enzymes are also discussed to assist researchers in further interrogation of these pathways.
Collapse
Affiliation(s)
- Justin M Snider
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Chiara Luberto
- The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Departments of Biochemistry, Pathology and Pharmacology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
14
|
Ceramide Imbalance and Impaired TLR4-Mediated Autophagy in BMDM of an ORMDL3-Overexpressing Mouse Model. Int J Mol Sci 2019; 20:ijms20061391. [PMID: 30897694 PMCID: PMC6470650 DOI: 10.3390/ijms20061391] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 03/14/2019] [Accepted: 03/18/2019] [Indexed: 01/07/2023] Open
Abstract
Increased orosomucoid-like 3 (ORMDL3) expression levels, due to single nucleotide polymorphisms (SNPs), have been associated with several inflammatory diseases, including asthma and inflammatory bowel diseases. ORMDL proteins inhibit serine palmitoyltransferase (SPT), the first rate-limiting enzyme in de novo sphingolipid synthesis and alter cellular calcium homeostasis. Both processes are essential for immune response. The present study addresses ORMDL3 protein involvement in macrophage physiology using an overexpressing knock-in mouse model. Ceramide content was notably different in the bone-marrow-derived macrophages (BMDM) from the transgenic mouse model compared with the wild type (WT) macrophages. Our data revealed an alteration of de novo production of sphinganine upon BMDM activation in the transgenic mouse. Gene-expression analysis showed that alteration in ORMDL3 expression levels did not affect activation or macrophage polarization. Nevertheless, we studied phagocytosis and autophagy—crucial processes that are dependent on lipid membrane composition. Phagocytosis in transgenic macrophages was not affected by ORMDL3 overexpression, but we did find a reduction in toll-like receptor 4 (TLR-4)-mediated autophagy. Both genetic and functional studies have pointed to autophagy as an essential pathway involved in inflammation. We believe that our work provides new insights into the functional link between ORMDL3 expression and inflammatory diseases.
Collapse
|
15
|
Ge YH, Chen YY, Zhou GS, Liu X, Tang YP, Liu R, Liu P, Li N, Yang J, Wang J, Yue SJ, Zhou H, Duan JA. A Novel Antithrombotic Protease from Marine Worm Sipunculus Nudus. Int J Mol Sci 2018; 19:ijms19103023. [PMID: 30287737 PMCID: PMC6213608 DOI: 10.3390/ijms19103023] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/23/2018] [Accepted: 09/28/2018] [Indexed: 12/15/2022] Open
Abstract
Sipunculus nudus, an old marine species, has great potential for use as functional seafood due to its various bioactivities. Its potential antithrombotic activity pushed us to isolate the bio-active components bio-guided by tracking fibrinolytic activity. As a result, a novel protease named as SK (the kinase obtained from S. nudus) was obtained, which possessed a molecular weight of 28,003.67 Da and 15 N-terminal amino acid sequences of PFPVPDPFVWDTSFQ. SK exerted inhibitory effects on thrombus formation through improving the coagulation system with dose-effect relationship within a certain range. Furthermore, in most cases SK got obviously better effect than that of urokinase. With the help of untargeted mass spectrometry-based metabolomics profiling, arachidonic acid, sphingolipid, and nicotinate and nicotinamide mechanism pathways were found to be important pathways. They revealed that the effect mechanism of SK on common carotid arterial thrombosis induced by FeCl3 was achieved by inhibiting vessel contraction, platelet aggregation, adhesion, and release, correcting endothelial cell dysfunction and retarding process of thrombus formation. This study demonstrated SK was a promising thrombolytic agent on the basis of its comprehensive activities on thrombosis, and it should get further exploitation and utilization.
Collapse
Affiliation(s)
- Ya-Hui Ge
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yan-Yan Chen
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Gui-Sheng Zhou
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Xin Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Rui Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Pei Liu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Na Li
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Avenida Wai Long, Taipa 999078, Macau, China.
| | - Jie Yang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Jing Wang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Shi-Jun Yue
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, and Shaanxi Key Laboratory of Chinese Medicine Fundamentals and New Drugs Research, and Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an 712046, China.
| | - Huiping Zhou
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond, VA 23298, USA.
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, and National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
16
|
Pou A, Abad JL, Ordóñez YF, Garrido M, Casas J, Fabriàs G, Delgado A. From the configurational preference of dihydroceramide desaturase-1 towards Δ 6-unsaturated substrates to the discovery of a new inhibitor. Chem Commun (Camb) 2018; 53:4394-4397. [PMID: 28379228 DOI: 10.1039/c6cc08268h] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dihydroceramide desaturase 1 (Des1) catalyzes the last step of ceramide synthesis de novo, thus regulating the physiologically relevant balance between dihydrosphingolipids and sphingolipids. Here we report on the configurational preference of Des1 towards isomeric Δ6-unsaturated dihydroceramide analogs and the discovery of a potent Des1 inhibitor.
Collapse
Affiliation(s)
- Ana Pou
- Spanish National Research Council (CSIC), Institute of Advanced Chemistry of Catalonia (IQAC-CSIC), Research Unit on Bioactive Molecules (RUBAM), Department of Biomedicinal Chemistry, Jordi Girona 18-26, 08034-Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
17
|
Dihydroceramide Desaturase 1 Inhibitors Reduce Amyloid-β Levels in Primary Neurons from an Alzheimer's Disease Transgenic Model. Pharm Res 2018; 35:49. [PMID: 29411122 DOI: 10.1007/s11095-017-2312-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 11/16/2017] [Indexed: 02/07/2023]
Abstract
PURPOSE The induction of autophagy has recently been explored as a promising therapeutic strategy to combat Alzheimer's disease. Among many other factors, there is evidence that ceramides/dihydroceramides act as mediators of autophagy, although the exact mechanisms underlying such effects are poorly understood. Here, we describe how two dihydroceramide desaturase inhibitors (XM461 and XM462) trigger autophagy and reduce amyloid secretion by neurons. METHODS Neurons isolated from wild-type and APP/PS1 transgenic mice were exposed to the two dihydroceramide desaturase inhibitors to assess their effect on these cell's protein and lipid profiles. RESULTS Both dihydroceramide desaturase inhibitors increased the autophagic vesicles in wild-type neurons, reflected as an increase in LC3-II, and this was correlated with the accumulation of dihydroceramides and dihydrosphingomyelins. Exposing APP/PS1 transgenic neurons to these inhibitors also produced a 50% reduction in amyloid secretion and/or production. The lipidomic defects triggered by these dihydroceramide desaturase inhibitors were correlated with a loss of S6K activity, witnessed by the changes in S6 phosphorylation, which strongly suggested a reduction of mTORC1 activity. CONCLUSIONS The data obtained strongly suggest that dihydroceramide desaturase 1 activity may modulate autophagy and mTORC1 activity in neurons, inhibiting amyloid secretion and S6K activity. As such, it is tantalizing to propose that dihydroceramide desaturase 1 may be an important therapeutic target to combat amyloidosis.
Collapse
|
18
|
Sands SA, LeVine SM. Substrate reduction therapy for Krabbe's disease. J Neurosci Res 2017; 94:1261-72. [PMID: 27638608 DOI: 10.1002/jnr.23791] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/19/2016] [Accepted: 05/18/2016] [Indexed: 01/30/2023]
Abstract
Krabbe's disease (KD) is a lysosomal storage disorder in which galactosylceramide, a major glycosphingolipid of myelin, and psychosine (galactose-sphingosine) cannot be adequately metabolized because of a deficiency in galactosylceramidase. Substrate reduction therapy (SRT) has been tested in preclinical studies. The premise of SRT is to reduce the synthesis of substrates that are not adequately digested so that the substrate burden is lowered, resulting in less accumulation of unmetabolized material. SRT is used for Gaucher's disease, in which inhibitors of the terminal biosynthetic step are used. Unfortunately, an inhibitor for the final step of galactosylceramide biosynthesis, i.e., UDP glycosyltransferase 8 (a.k.a. UDP-galactose ceramide galactosyltransferase), has not been found. Approaches that inhibit an earlier biosynthetic step or that lessen the substrate burden by other means, such as genetic manipulations, have been tested in the twitcher mouse model of KD. Either as a stand-alone therapy or in combination with other approaches, SRT slowed the disease course, indicating that this approach has potential therapeutic value. For instance, in individuals with adult-onset disease, SRT theoretically could lessen the production of substrates so that residual enzymatic activity could adequately manage the lower substrate burden. In more severe forms of disease, SRT theoretically could be part of a combination therapy. However, SRT has the potential to impair normal function by reducing the synthesis of galactosylceramide to levels that impede myelin function, or SRT could have other deleterious effects. Thus, multiple issues need to be resolved before this approach is ready for testing in humans. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Scott A Sands
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - Steven M LeVine
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas.
| |
Collapse
|
19
|
Lee AY, Lee JW, Kim JE, Mock HJ, Park S, Kim S, Hong SH, Kim JY, Park EJ, Kang KS, Kim KP, Cho MH. Dihydroceramide is a key metabolite that regulates autophagy and promotes fibrosis in hepatic steatosis model. Biochem Biophys Res Commun 2017; 494:460-469. [PMID: 29066349 DOI: 10.1016/j.bbrc.2017.10.110] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/20/2017] [Indexed: 12/20/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is an increasingly common chronic liver disease worldwide. Sphingolipids are a family of lipids that play essential roles as critical regulators in metabolic disorders. Some sphingolipids are known key factors in metabolic dysfunction. However, the precise effect of dihydroceramide on NAFLD remains unknown. Here, we report how dihydroceramide in autophagosome accumulation activates fibrogenesis in human liver Chang cells treated with free fatty acids (FFA). According to LC/MS lipid profiling, FFA increased the levels of sphingolipids and triacylglycerol (TG). To demonstrate the potential role of dihydroceramide metabolism in autophagy, several sphingolipid synthesis inhibitors were used. Increased dihydroceramide led to impairment of autophagic flux, resulting in increased TG storage in lipid droplets (LD) and upregulated expression of fibrosis markers. Hepatic stellate cells (HSCs, LX-2 cells) were co-cultured with Chang cells to assess the potential fibrogenic response to dihydroceramide, Treatment with rapamycin recovered autophagic flux in Chang cells and fibrogenesis in the co-culture system. Our results identified a critical function of dihydroceramide metabolism in autophagy. It could play an important role in the progression of NAFLD associated with lipid over-accumulation. Therefore, preventing autophagic flux by regulating dihydroceramide could be a potential strategic approach for providing therapy for NAFLD.
Collapse
Affiliation(s)
- Ah Young Lee
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Jae Won Lee
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Ji-Eun Kim
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada
| | - Hyuck Jun Mock
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea
| | - Sungjin Park
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Departmentof Pharmacology and Medical Science, Metabolic Diseases and Cell Signaling Laboratory, Research Institute for Medical Sciences, College of Medicine, Chungnam National University, Daejeon 35015, Republic of Korea
| | - Sanghwa Kim
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Division of Basic Radiation Bioscience, Korea Institute of Radiological & Medical Science, Seoul 01812, Republic of Korea
| | - Seong-Ho Hong
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Bio medicine Lab., CKD Research Institute, Yongin 16995, Republic of Korea
| | - Ji-Young Kim
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Eun-Jung Park
- Department of Brain Science, Ajou University School of Medicine, 164, World Cup-ro, Youngtong-gu, Suwon, 16499, Republic of Korea
| | - Kyung-Sun Kang
- Adult Stem Cell Research Center, College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea
| | - Kwang Pyo Kim
- Department of Applied Chemistry, College of Applied Science, Kyung Hee University, Yongin 17104, Republic of Korea.
| | - Myung-Haing Cho
- Laboratory of Toxicology, Research Institute for Veterinary Science and College of Veterinary Medicine, Seoul National University, Seoul 08826, Republic of Korea; Graduate School of Convergence Science and Technology, Seoul National University, Suwon 16229, Republic of Korea; Graduate Group of Tumor Biology, Seoul National University, Seoul 08826, Republic of Korea; Advanced Institute of Convergence Technology, Seoul National University, Suwon 16229, Republic of Korea; Institute of GreenBio Science Technology, Seoul National University, Pyeongchang 25354, Republic of Korea.
| |
Collapse
|
20
|
Campisi GM, Signorelli P, Rizzo J, Ghilardi C, Antognetti J, Caretti A, Lazarević JS, Strettoi E, Novelli E, Ghidoni R, Rubino FM, Paroni R. Determination of the serine palmitoyl transferase inhibitor myriocin by electrospray and Q-trap mass spectrometry. Biomed Chromatogr 2017. [PMID: 28621883 DOI: 10.1002/bmc.4026] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Myriocin is a potent inhibitor of serine-palmitoyl-transferase, the first and rate-determining enzyme in the sphingolipids biosynthetic pathway. This study developed, validated and applied a LC-MS/MS method to measure myriocin in minute specimens of animal tissue. The chemical analog 14-OH-myriocin was used as the internal standard. The two molecules were extracted from the tissue homogenate by solid-phase extraction, separated by gradient reversed-phase liquid chromatography and measured by negative ion electrospray mass spectrometry in the triple quadrupole. Detection was accomplished by multiple reaction monitoring, employing the most representative transitions, 400@104 and 402@104 for myriocin and 14-OH-myriocin, respectively. The typical limit of detection and lower limit of quantitation of the optimized method were 0.9 pmol/mL (~0.016 pmol injected) and 2.3 pmol/mL, respectively, and the method was linear up to 250 pmol/mL range (r2 = 0.9996). The intra- and between-day repeatability afforded a coefficient of variation ≤7.0%. Applications included quantification of myriocin in mouse lungs after 24 h from administration of ~4 nmol by intra-tracheal delivery. Measured levels ranged from 4.11 (median; 2.3-7.4 IQR, n = 4) to 11.7 (median; 7.6-22.7 interquartile range (IQR), n = 6) pmol/lung depending on the different formulations used. Myriocin was also measured in retinas of mice treated by intravitreal injection and ranged from 0.045 (less than the limit of detection) to 0.35 pmol/retina.
Collapse
Affiliation(s)
| | - Paola Signorelli
- Laboratory of Biochemistry and Molecular Biology, University of Milan, Milan, Italy
| | - Jessica Rizzo
- Laboratory of Analytical Chemistry and Clinical Biochemistry, University of Milan, Milan, Italy
| | - Claudio Ghilardi
- Laboratory of Analytical Chemistry and Clinical Biochemistry, University of Milan, Milan, Italy
| | - Jacopo Antognetti
- Laboratory of Analytical Chemistry and Clinical Biochemistry, University of Milan, Milan, Italy
| | - Anna Caretti
- Laboratory of Biochemistry and Molecular Biology, University of Milan, Milan, Italy
| | - Jelena S Lazarević
- Department of Chemistry, Faculty of Medical Sciences, University of Nisˇ, Nis, Serbia
| | - Enrica Strettoi
- Neuroscience Institute, Consiglio Nazionale delle Ricerche, Pisa, Italy
| | - Elena Novelli
- G.B. Bietti Foundation for Ophthalmology, Rome, Italy
| | - Riccardo Ghidoni
- Laboratory of Biochemistry and Molecular Biology, University of Milan, Milan, Italy
| | - Federico Maria Rubino
- Laboratory for Analytical Toxicology and Metabolomics and International Centre for Rural Health, Department of Health Science, University of Milan, Milan, Italy
| | - Rita Paroni
- Laboratory of Analytical Chemistry and Clinical Biochemistry, University of Milan, Milan, Italy
| |
Collapse
|
21
|
Zulueta A, Caretti A, Campisi GM, Brizzolari A, Abad JL, Paroni R, Signorelli P, Ghidoni R. Inhibitors of ceramide de novo biosynthesis rescue damages induced by cigarette smoke in airways epithelia. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2017; 390:753-759. [PMID: 28409208 DOI: 10.1007/s00210-017-1375-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 04/06/2017] [Indexed: 02/03/2023]
Abstract
Exposure to cigarette smoke represents the most important risk factor for the development of chronic obstructive pulmonary disease (COPD). COPD is characterized by chronic inflammation of the airways, imbalance of proteolytic activity resulting in the destruction of lung parenchyma, alveolar hypoxia, oxidative stress, and apoptosis. Sphingolipids are structural membrane components whose metabolism is altered during stress. Known as apoptosis and inflammation inducer, the sphingolipid ceramide was found to accumulate in COPD airways and its plasma concentration increased as well. The present study investigates the role of sphingolipids in the cigarette smoke-induced damage of human airway epithelial cells. Lung epithelial cells were pre-treated with sphingolipid synthesis inhibitors (myriocin or XM462) and then exposed to a mixture of nicotine, acrolein, formaldehyde, and acetaldehyde, the major toxic cigarette smoke components. The inflammatory and proteolytic responses were investigated by analysis of the mRNA expression (RT-PCR) of cytokines IL-1β and IL-8, and matrix metalloproteinase-9 and of the protein expression (ELISA) of IL-8. Ceramide intracellular amounts were measured by LC-MS technique. Ferric-reducing antioxidant power test and superoxide anion radical scavenging activity assay were used to assess the antioxidant power of the inhibitors of ceramide synthesis. We here show that ceramide synthesis is enhanced under treatment with a cigarette smoke mixture correlating with increased expression of inflammatory cytokines and matrix metalloproteinase 9. The use of inhibitors of ceramide synthesis protected from smoke induced damages such as inflammation, oxidative stress, and proteolytic imbalance in airways epithelia.
Collapse
Affiliation(s)
- Aida Zulueta
- Health Sciences Department, Biochemistry & Mol. Biology Lab., University of Milan, Via Di Rudinì 8, 20142, Milan, Italy.
| | - Anna Caretti
- Health Sciences Department, Biochemistry & Mol. Biology Lab., University of Milan, Via Di Rudinì 8, 20142, Milan, Italy
| | - Giuseppe Matteo Campisi
- Health Sciences Department, Clinical Biochemistry &Mass Spectrometry Lab, University of Milan, Milan, Italy
| | - Andrea Brizzolari
- Health Sciences Department, Biochemistry & Mol. Biology Lab., University of Milan, Via Di Rudinì 8, 20142, Milan, Italy
| | - Jose Luis Abad
- Department of Biomed. Chem., IQAC/CSIC, Research Unit on Bioactive Molecules, Barcelona, Spain
| | - Rita Paroni
- Health Sciences Department, Clinical Biochemistry &Mass Spectrometry Lab, University of Milan, Milan, Italy
| | - Paola Signorelli
- Health Sciences Department, Biochemistry & Mol. Biology Lab., University of Milan, Via Di Rudinì 8, 20142, Milan, Italy
| | - Riccardo Ghidoni
- Health Sciences Department, Biochemistry & Mol. Biology Lab., University of Milan, Via Di Rudinì 8, 20142, Milan, Italy
| |
Collapse
|
22
|
Casasampere M, Ordóñez YF, Casas J, Fabrias G. Dihydroceramide desaturase inhibitors induce autophagy via dihydroceramide-dependent and independent mechanisms. Biochim Biophys Acta Gen Subj 2016; 1861:264-275. [PMID: 27894925 DOI: 10.1016/j.bbagen.2016.11.033] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/10/2016] [Accepted: 11/23/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND Autophagy consists on the delivery of cytoplasmic material and organelles to lysosomes for degradation. Research on autophagy is a growing field because deciphering the basic mechanisms of autophagy is key to understanding its role in health and disease, and to paving the way to discovering novel therapeutic strategies. Studies with chemotherapeutic drugs and pharmacological tools support a role for dihydroceramides as mediators of autophagy. However, their effect on the autophagy outcome (cell survival or death) is more controversial. METHODS We have examined the capacity of structurally varied Des1 inhibitors to stimulate autophagy (LC3-II analysis), to increase dihydroceramides (mass spectrometry) and to reduce cell viability (SRB) in T98G and U87MG glioblastoma cells under different experimental conditions. RESULTS The compounds activity on autophagy induction took place concomitantly with accumulation of dihydroceramides, which occurred by both stimulation of ceramide synthesis de novo and reduction of Des1 activity. However, autophagy was also induced by the test compounds after preincubation with myriocin and in cells with a reduced capacity to produce dihydroceramides (U87DND). Autophagy inhibition with 3-methyladenine in the de novo dihydroceramide synthesis competent U87MG cells increased cytotoxicity, while genetic inhibition of autophagy in U87DND cells, poorly efficient at synthesizing dihydroceramides, augmented resistance to the test compounds. CONCLUSION Dihydroceramide desaturase 1 inhibitors activate autophagy via both dihydroceramide-dependent and independent pathways and the balance between the two pathways influences the final cell fate. GENERAL SIGNIFICANCE The cells capacity to biosynthesize dihydroceramides must be taken into account in proautophagic Des1 inhibitors-including therapies.
Collapse
Affiliation(s)
- Mireia Casasampere
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Yadira F Ordóñez
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Josefina Casas
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain
| | - Gemma Fabrias
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Departament de Química Biomèdica, Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain..
| |
Collapse
|
23
|
Ordóñez YF, González J, Bedia C, Casas J, Abad JL, Delgado A, Fabrias G. 3-Ketosphinganine provokes the accumulation of dihydroshingolipids and induces autophagy in cancer cells. MOLECULAR BIOSYSTEMS 2016; 12:1166-73. [PMID: 26928714 DOI: 10.1039/c5mb00852b] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Although several reports describe the metabolic fate of sphingoid bases and their analogs, as well as their action and that of their phosphates as regulators of sphingolipid metabolizing-enzymes, similar studies for 3-ketosphinganine (KSa), the product of the first committed step in de novo sphingolipid biosynthesis, have not been reported. In this article we show that 3-ketosphinganine (KSa) and its dideuterated analog at C4 (d2KSa) are metabolized to produce high levels of dihydrosphingolipids in HGC27, T98G and U87MG cancer cells. In contrast, either direct C1 O-phosphorylation or N-acylation of d2KSa to produce dideuterated ketodihydrosphingolipids does not occur. We also show that cells respond to d2KSa treatment with induction of autophagy. Time-course experiments agree with sphinganine, sphinganine 1-phosphate and dihydroceramides being the mediators of autophagy stimulated by d2KSa. Enzyme inhibition studies support that inhibition of Des1 by 3-ketobases is caused by their dihydroceramide metabolites. However, this effect contributes to increasing dihydrosphingolipid levels only at short incubation times, since cells respond to long time exposure to 3-ketobases with Des1 overexpression. The translation of these overall effects into cell fate is discussed.
Collapse
Affiliation(s)
- Yadira F Ordóñez
- Consejo Superior de Investigaciones Científicas (CSIC), Institut de Química Avançada de Catalunya (IQAC-CSIC), Research Unit on Bioactive Molecules (RUBAM), Jordi Girona 18-26, 08034 Barcelona, Spain.
| | | | | | | | | | | | | |
Collapse
|
24
|
Aurelio L, Scullino CV, Pitman MR, Sexton A, Oliver V, Davies L, Rebello RJ, Furic L, Creek DJ, Pitson SM, Flynn BL. From Sphingosine Kinase to Dihydroceramide Desaturase: A Structure-Activity Relationship (SAR) Study of the Enzyme Inhibitory and Anticancer Activity of 4-((4-(4-Chlorophenyl)thiazol-2-yl)amino)phenol (SKI-II). J Med Chem 2016; 59:965-84. [PMID: 26780304 DOI: 10.1021/acs.jmedchem.5b01439] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The sphingosine kinase (SK) inhibitor, SKI-II, has been employed extensively in biological investigations of the role of SK1 and SK2 in disease and has demonstrated impressive anticancer activity in vitro and in vivo. However, interpretations of results using this pharmacological agent are complicated by several factors: poor SK1/2 selectivity, additional activity as an inducer of SK1-degradation, and off-target effects, including its recently identified capacity to inhibit dihydroceramide desaturase-1 (Des1). In this study, we have delineated the structure-activity relationship (SAR) for these different targets and correlated them to that required for anticancer activity and determined that Des1 inhibition is primarily responsible for the antiproliferative effects of SKI-II and its analogues. In the course of these efforts, a series of novel SK1, SK2, and Des1 inhibitors have been generated, including compounds with significantly greater anticancer activity.
Collapse
Affiliation(s)
- Luigi Aurelio
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Carmen V Scullino
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Melissa R Pitman
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Anna Sexton
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Victoria Oliver
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Lorena Davies
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Richard J Rebello
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Luc Furic
- Cancer Program, Monash Biomedicine Discovery Institute and Department of Anatomy and Developmental Biology, Clayton, Victoria 3800, Australia
| | - Darren J Creek
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| | - Stuart M Pitson
- Centre for Cancer Biology, University of South Australia and SA Pathology , Frome Road, Adelaide South Australia 5000, Australia
| | - Bernard L Flynn
- Monash Institute of Pharmaceutical Science, Monash University , 381 Royal Parade, Parkville, Victoria 3052, Australia
| |
Collapse
|
25
|
Inhibition of ceramide de novo synthesis as a postischemic strategy to reduce myocardial reperfusion injury. Basic Res Cardiol 2016; 111:12. [PMID: 26786259 DOI: 10.1007/s00395-016-0533-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 01/11/2016] [Indexed: 12/17/2022]
Abstract
The injury caused by myocardial reperfusion after ischemia can be contained by interventions aimed at reducing the inflammation and the oxidative stress that underlie exacerbation of tissue damage. Sphingolipids are a class of structural and signaling lipid molecules; among them, the inflammation mediator ceramide accumulates in the myocardium upon ischemia/reperfusion. Here, we show that, after transient coronary occlusion in mice, an increased de novo ceramide synthesis takes place at reperfusion in the ischemic area surrounding necrosis (area at risk). This correlates with the enhanced expression of the first and rate-limiting enzyme of the de novo pathway, serine palmitoyltransferase (SPT). The intraventricular administration at reperfusion of myriocin, an inhibitor of SPT, significantly protected the area at risk from damage, reducing the infarcted area by 40.9 % relative to controls not treated with the drug. In the area at risk, myriocin downregulated ceramide, reduced the content in other mediators of inflammation and reactive oxygen species, and activated the Nrf2-HO1 cytoprotective response. We conclude that an enhanced ceramide synthesis takes part in ischemia/reperfusion injury and that myriocin treatment can be proposed as a strategy for myocardial pharmacological postconditioning.
Collapse
|
26
|
A selective ATP-competitive sphingosine kinase inhibitor demonstrates anti-cancer properties. Oncotarget 2016; 6:7065-83. [PMID: 25788259 PMCID: PMC4466670 DOI: 10.18632/oncotarget.3178] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/25/2015] [Indexed: 12/20/2022] Open
Abstract
The dynamic balance of cellular sphingolipids, the sphingolipid rheostat, is an important determinant of cell fate, and is commonly deregulated in cancer. Sphingosine 1-phosphate is a signaling molecule with anti-apoptotic, pro-proliferative and pro-angiogenic effects, while conversely, ceramide and sphingosine are pro-apoptotic. The sphingosine kinases (SKs) are key regulators of this sphingolipid rheostat, and are attractive targets for anti-cancer therapy. Here we report a first-in-class ATP-binding site-directed small molecule SK inhibitor, MP-A08, discovered using an approach of structural homology modelling of the ATP-binding site of SK1 and in silico docking with small molecule libraries. MP-A08 is a highly selective ATP competitive SK inhibitor that targets both SK1 and SK2. MP-A08 blocks pro-proliferative signalling pathways, induces mitochondrial-associated apoptosis in a SK-dependent manner, and reduces the growth of human lung adenocarcinoma tumours in a mouse xenograft model by both inducing tumour cell apoptosis and inhibiting tumour angiogenesis. Thus, this selective ATP competitive SK inhibitor provides a promising candidate for potential development as an anti-cancer therapy, and also, due to its different mode of inhibition to other known SK inhibitors, both validates the SKs as targets for anti-cancer therapy, and represents an important experimental tool to study these enzymes.
Collapse
|
27
|
Perdoni F, Signorelli P, Cirasola D, Caretti A, Galimberti V, Biggiogera M, Gasco P, Musicanti C, Morace G, Borghi E. Antifungal activity of Myriocin on clinically relevant Aspergillus fumigatus strains producing biofilm. BMC Microbiol 2015; 15:248. [PMID: 26519193 PMCID: PMC4628231 DOI: 10.1186/s12866-015-0588-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 10/23/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The human pathogenic mold Aspergillus fumigatus is able to form a complex biofilm embedded in extracellular matrix. Biofilms confer antimicrobial resistance and it is well known that aspergillosis is often refractory to the conventional antifungal therapy. The treatment of biofilm-related infections poses a significant clinical challenge on a daily basis, promoting the search for new therapeutic agents. Our aim was to exploit the modulation of sphingolipid mediators as new therapeutic target to overcome antifungal resistance in biofilm-related infections. RESULTS Antifungal susceptibility testing was performed on 20 clinical isolates of Aspergillus fumigatus and one reference strain (A. fumigatus Af293) according the EUCAST protocol. Sessile MICs were assessed on 24-h preformed-biofilm by means of XTT-reduction assay. Myriocin (0.25-64 mg/L), a commercial sphingolipid synthesis inhibitor, was used. The MEC50 value (mg/L) of Myriocin was 8 (range 4-16) for both planktonic and sessile cells. Drug-induced morphological alterations were analyzed by optical and electron microscopy (TEM) on 24h preformed A. fumigatus Af293 biofilms. An evident hyphal damage, resulting in short, stubby, and highly branched hyphae was observed by optical microscopy. At 24h, TEM studies showed important morphological alterations, such as invaginations of the cell membrane, modification in the vacuolar system and presence of multilamellar bodies, in some cases within vacuoles. CONCLUSIONS The direct antifungal activity, observed on both planktonic and sessile fungi, suggests that inhibition of sphingolipid synthesis could represent a new target to fight biofilm-related A. fumigatus resistance.
Collapse
Affiliation(s)
- Federica Perdoni
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| | - Paola Signorelli
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| | - Daniela Cirasola
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| | - Anna Caretti
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| | - Valentina Galimberti
- Department of Biology and Biotechnology, Università degli Studi di Pavia, Pavia, Italy.
| | - Marco Biggiogera
- Department of Biology and Biotechnology, Università degli Studi di Pavia, Pavia, Italy.
| | | | | | - Giulia Morace
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| | - Elisa Borghi
- Department of Health Sciences, Università degli Studi di Milano, Polo Universitario San Paolo, Blocco C, ottavo piano, via di Rudinì 8, 20142, Milan, Italy.
| |
Collapse
|
28
|
Casasampere M, Ordoñez YF, Pou A, Casas J. Inhibitors of dihydroceramide desaturase 1: Therapeutic agents and pharmacological tools to decipher the role of dihydroceramides in cell biology. Chem Phys Lipids 2015; 197:33-44. [PMID: 26248324 DOI: 10.1016/j.chemphyslip.2015.07.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 07/29/2015] [Accepted: 07/31/2015] [Indexed: 02/07/2023]
Abstract
Dihydroceramide desaturase (Des1) is the last enzyme in the de novo synthesis of ceramides (Cer). It catalyzes the insertion of a double bond into dihydroceramides (dhCer) to convert them to Cer, both of which are further metabolized to more complex (dihydro) sphingolipids. For many years dhCer have received poor attention, mainly due to their supposed lack of biological activity. It was not until about ten years ago that the concept that dhCer might have regulatory roles in biology emerged for the first time. Since then, multiple publications have established that dhCer are implicated in a wide spectrum of biological processes. Physiological and pathophysiological functions of dhCer have been recently reviewed. In this review we will focus on the biochemical features of Des1 and on its inhibition by different compounds with presumably different modes of action.
Collapse
Affiliation(s)
- Mireia Casasampere
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Yadira F Ordoñez
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Ana Pou
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), Jordi Girona 18-26, E-08034 Barcelona, Spain.
| |
Collapse
|
29
|
Garcia V, Le Faouder P, Dupuy A, Levade T, Ballereau S, Génisson Y. A novel jaspine B-ceramide hybrid modulates sphingolipid metabolism. Chem Biodivers 2015; 12:1115-25. [PMID: 26172331 DOI: 10.1002/cbdv.201400357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Indexed: 11/09/2022]
Abstract
A new sphingolipid hybrid molecule was designed to assemble, within a tail-to-tail double-chain structure, the ceramide hydrophilic moiety and the tetrahydrofuran pharmacophore of jaspine B, a natural product known to interfere with sphingolipid metabolism. This compound was prepared through acylation of sphingosine with a jaspine B derivative bearing a COOH group in the terminal position of the aliphatic backbone. This new hybrid molecule was evaluated for its capacities to affect melanoma cell viability and sphingolipid metabolism. While retaining the cytotoxicity of ceramide itself, this compound was shown to lower the sphingomyelin cellular levels and significantly enhance the production of sphingosine-1-phosphate, thus representing a novel sphingolipid metabolism modulator.
Collapse
Affiliation(s)
- Virginie Garcia
- INSERM UMR1037, CRCT (Centre de Recherches en Canćerolgie de Toulouse), Oncopole de Toulouse, 2, avenue Hubert Curien, CS 53717, 31037 Toulouse cedex 1
| | - Pauline Le Faouder
- Lipidomic Core Facility, MetaToul platform, UMR1048 INSERM-Université Paul Sabatier-Toulouse III, CHU Rangueil, BP, FR-84225, Toulouse
| | - Aude Dupuy
- Lipidomic Core Facility, MetaToul platform, UMR1048 INSERM-Université Paul Sabatier-Toulouse III, CHU Rangueil, BP, FR-84225, Toulouse
| | - Thierry Levade
- INSERM UMR1037, CRCT (Centre de Recherches en Canćerolgie de Toulouse), Oncopole de Toulouse, 2, avenue Hubert Curien, CS 53717, 31037 Toulouse cedex 1
| | - Stéphanie Ballereau
- LSPCMIB, UMR-5068, CNRS, Université de Toulouse, UPS, FR-31062, Toulouse, (phone: +33-561-556299; fax: +33-561-556011).
| | - Yves Génisson
- LSPCMIB, UMR-5068, CNRS, Université de Toulouse, UPS, FR-31062, Toulouse, (phone: +33-561-556299; fax: +33-561-556011).
| |
Collapse
|
30
|
Signorelli P, Fabiani C, Brizzolari A, Paroni R, Casas J, Fabriàs G, Rossi D, Ghidoni R, Caretti A. Natural Grape Extracts Regulate Colon Cancer Cells Malignancy. Nutr Cancer 2015; 67:494-503. [DOI: 10.1080/01635581.2015.1004591] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Paola Signorelli
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| | - Carlotta Fabiani
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| | - Andrea Brizzolari
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| | - Rita Paroni
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| | - Josefina Casas
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Catalan Institute of Advanced Chemistry, Barcelona, Spain
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules, Department of Biomedicinal Chemistry, Catalan Institute of Advanced Chemistry, Barcelona, Spain
| | - Dario Rossi
- Immobiliare Ca’ Novella srl, Alessandria, Italy
| | - Riccardo Ghidoni
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| | - Anna Caretti
- Department of Health Sciences, University of Milan, Milan, Italy, and San Paolo Hospital, Milan, Italy
| |
Collapse
|
31
|
Rodriguez-Cuenca S, Barbarroja N, Vidal-Puig A. Dihydroceramide desaturase 1, the gatekeeper of ceramide induced lipotoxicity. Biochim Biophys Acta Mol Cell Biol Lipids 2015; 1851:40-50. [DOI: 10.1016/j.bbalip.2014.09.021] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/25/2022]
|
32
|
Kiefer K, Carreras-Sureda A, García-López R, Rubio-Moscardó F, Casas J, Fabriàs G, Vicente R. Coordinated regulation of the orosomucoid-like gene family expression controls de novo ceramide synthesis in mammalian cells. J Biol Chem 2014; 290:2822-30. [PMID: 25519910 DOI: 10.1074/jbc.m114.595116] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The orosomucoid-like (ORMDL) protein family is involved in the regulation of de novo sphingolipid synthesis, calcium homeostasis, and unfolded protein response. Single nucleotide polymorphisms (SNPs) that increase ORMDL3 expression have been associated with various immune/inflammatory diseases, although the pathophysiological mechanisms underlying this association are poorly understood. ORMDL proteins are claimed to be inhibitors of the serine palmitoyltransferase (SPT). However, it is not clear whether individual ORMDL expression levels have an impact on ceramide synthesis. The present study addressed the interaction with and regulation of SPT activity by ORMDLs to clarify their pathophysiological relevance. We have measured ceramide production in HEK293 cells incubated with palmitate as a direct substrate for SPT reaction. Our results showed that a coordinated overexpression of the three isoforms inhibits the enzyme completely, whereas individual ORMDLs are not as effective. Immunoprecipitation and fluorescence resonance energy transfer (FRET) studies showed that mammalian ORMDLs form oligomeric complexes that change conformation depending on cellular sphingolipid levels. Finally, using macrophages as a model, we demonstrate that mammalian cells modify ORMDL genes expression levels coordinately to regulate the de novo ceramide synthesis pathway. In conclusion, we have shown a physiological modulation of SPT activity by general ORMDL expression level regulation. Moreover, because single ORMDL3 protein alteration produces an incomplete inhibition of SPT activity, this work argues against the idea that ORMDL3 pathophysiology could be explained by a simple on/off mechanism on SPT activity.
Collapse
Affiliation(s)
- Kerstin Kiefer
- From the Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain and
| | - Amado Carreras-Sureda
- From the Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain and
| | - Roberto García-López
- From the Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain and
| | - Fanny Rubio-Moscardó
- From the Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain and
| | - Josefina Casas
- Research Unit on Bioactive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC), 08034 Barcelona, Spain
| | - Gemma Fabriàs
- Research Unit on Bioactive Molecules (RUBAM), Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia (IQAC), 08034 Barcelona, Spain
| | - Rubén Vicente
- From the Laboratory of Molecular Physiology and Channelopathies, Department of Experimental and Health Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain and
| |
Collapse
|
33
|
Jones EE, Dworski S, Canals D, Casas J, Fabrias G, Schoenling D, Levade T, Denlinger C, Hannun YA, Medin JA, Drake RR. On-tissue localization of ceramides and other sphingolipids by MALDI mass spectrometry imaging. Anal Chem 2014; 86:8303-11. [PMID: 25072097 PMCID: PMC4139181 DOI: 10.1021/ac501937d] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
![]()
A novel MALDI-FTICR imaging mass
spectrometry (MALDI-IMS) workflow
is described for on-tissue detection, spatial localization, and structural
confirmation of low abundance bioactive ceramides and other sphingolipids.
Increasingly, altered or elevated levels of sphingolipids, sphingolipid
metabolites, and sphingolipid metabolizing enzymes have been associated
with a variety of disorders such as diabetes, obesity, lysosomal storage
disorders, and cancer. Ceramide, which serves as a metabolic hub in
sphingolipid metabolism, has been linked to cancer signaling pathways
and to metabolic regulation with involvement in autophagy, cell-cycle
arrest, senescence, and apoptosis. Using kidney tissues from a new
Farber disease mouse model in which ceramides of all acyl chain lengths
and other sphingolipid metabolites accumulate in tissues, specific
ceramides and sphingomyelins were identified by on-tissue isolation
and fragmentation, coupled with an on-tissue digestion by ceramidase
or sphingomyelinase. Multiple glycosphingolipid species were also
detected. The newly generated library of sphingolipid ions was then
applied to MALDI-IMS of human lung cancer tissues. Multiple tumor
specific ceramide and sphingomyelin species were detected and confirmed
by on-tissue enzyme digests and structural confirmation. High-resolution
MALDI-IMS in combination with novel on-tissue ceramidase and sphingomyelinase
enzyme digestions makes it now possible to rapidly visualize the distribution
of bioactive ceramides and sphingomyelin in tissues.
Collapse
Affiliation(s)
- E Ellen Jones
- Department of Cell and Molecular Pharmacology and MUSC Proteomics Center, Medical University of South Carolina , 173 Ashley Avenue, Charleston, South Carolina 29425, United States
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Cingolani F, Casasampere M, Sanllehí P, Casas J, Bujons J, Fabrias G. Inhibition of dihydroceramide desaturase activity by the sphingosine kinase inhibitor SKI II. J Lipid Res 2014; 55:1711-20. [PMID: 24875537 PMCID: PMC4109765 DOI: 10.1194/jlr.m049759] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 05/27/2014] [Indexed: 01/05/2023] Open
Abstract
Sphingosine kinase inhibitor (SKI) II has been reported as a dual inhibitor of sphingosine kinases (SKs) 1 and 2 and has been extensively used to prove the involvement of SKs and sphingosine-1-phosphate (S1P) in cellular processes. Dihydroceramide desaturase (Des1), the last enzyme in the de novo synthesis of ceramide (Cer), regulates the balance between dihydroceramides (dhCers) and Cers. Both SKs and Des1 have interest as therapeutic targets. Here we show that SKI II is a noncompetitive inhibitor (Ki = 0.3 μM) of Des1 activity with effect also in intact cells without modifying Des1 protein levels. Molecular modeling studies support that the SKI II-induced decrease in Des1 activity could result from inhibition of NADH-cytochrome b5 reductase. SKI II, but not the SK1-specific inhibitor PF-543, provoked a remarkable accumulation of dhCers and their metabolites, while both SKI II and PF-543 reduced S1P to almost undetectable levels. SKI II, but not PF543, reduced cell proliferation with accumulation of cells in the G0/G1 phase. SKI II, but not PF543, induced autophagy. These overall findings should be taken into account when using SKI II as a pharmacological tool, as some of the effects attributed to decreased S1P may actually be caused by augmented dhCers and/or their metabolites.
Collapse
Affiliation(s)
- Francesca Cingolani
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Mireia Casasampere
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Pol Sanllehí
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
- Faculty of Pharmacy, Unit of Pharmaceutical Chemistry (Associated Unit to CSIC), University of Barcelona, E-08028 Barcelona, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Jordi Bujons
- Biological Chemistry and Molecular Modeling, Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| | - Gemma Fabrias
- Research Unit on BioActive Molecules (RUBAM), Departments of Biomedicinal Chemistry Institute for Advanced Chemistry of Catalonia (IQAC-CSIC), 08034 Barcelona, Spain
| |
Collapse
|
35
|
Caretti A, Bragonzi A, Facchini M, De Fino I, Riva C, Gasco P, Musicanti C, Casas J, Fabriàs G, Ghidoni R, Signorelli P. Anti-inflammatory action of lipid nanocarrier-delivered myriocin: therapeutic potential in cystic fibrosis. Biochim Biophys Acta Gen Subj 2013; 1840:586-94. [PMID: 24141140 DOI: 10.1016/j.bbagen.2013.10.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 09/07/2013] [Accepted: 10/10/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND Sphingolipids take part in immune response and can initiate and/or sustain inflammation. Various inflammatory diseases have been associated with increased ceramide content, and pharmacological reduction of ceramide diminishes inflammation damage in vivo. Inflammation and susceptibility to microbial infection are two elements in a vicious circle. Recently, sphingolipid metabolism inhibitors were used to reduce infection. Cystic fibrosis (CF) is characterized by a hyper-inflammation and an excessive innate immune response, which fails to evolve into adaptive immunity and to eradicate infection. Chronic infections result in lung damage and patient morbidity. Notably, ceramide content in mucosa airways is higher in CF mouse models and in patients than in control mice or healthy subjects. METHODS The therapeutic potential of myriocin, an inhibitor of the sphingolipid de novo synthesis rate limiting enzyme (Serine Palmitoyl Transferase, SPT),was investigated in CF cells and mice models. RESULTS We treated CF human respiratory epithelial cells with myriocin, This treatment resulted in reduced basal, as well as TNFα-stimulated, inflammation. In turn, TNFα induced an increase in SPT in these cells, linking de novo synthesis of ceramide to inflammation. Furthermore, myriocin-loaded nanocarrier, injected intratrachea prior to P. aeruginosa challenge, enabled a significant reduction of lung infection and reduced inflammation. CONCLUSIONS The presented data suggest that de novo ceramide synthesis is constitutively enhanced in CF mucosa and that it can be envisaged as pharmacological target for modulating inflammation and restoring effective innate immunity against acute infection. GENERAL SIGNIFICANCE Myriocin stands as a powerful immunomodulatory agent for inflammatory and infectious diseases.
Collapse
Affiliation(s)
- Anna Caretti
- Department of Health Sciences, University of Milan, San Paolo Hospital, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Alayoubi AM, Wang JCM, Au BCY, Carpentier S, Garcia V, Dworski S, El-Ghamrasni S, Kirouac KN, Exertier MJ, Xiong ZJ, Privé GG, Simonaro CM, Casas J, Fabrias G, Schuchman EH, Turner PV, Hakem R, Levade T, Medin JA. Systemic ceramide accumulation leads to severe and varied pathological consequences. EMBO Mol Med 2013; 5:827-42. [PMID: 23681708 PMCID: PMC3779446 DOI: 10.1002/emmm.201202301] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2012] [Revised: 04/04/2013] [Accepted: 04/05/2013] [Indexed: 12/27/2022] Open
Abstract
Farber disease (FD) is a severe inherited disorder of lipid metabolism characterized by deficient lysosomal acid ceramidase (ACDase) activity, resulting in ceramide accumulation. Ceramide and metabolites have roles in cell apoptosis and proliferation. We introduced a single-nucleotide mutation identified in human FD patients into the murine Asah1 gene to generate the first model of systemic ACDase deficiency. Homozygous Asah1P361R/P361R animals showed ACDase defects, accumulated ceramide, demonstrated FD manifestations and died within 7–13 weeks. Mechanistically, MCP-1 levels were increased and tissues were replete with lipid-laden macrophages. Treatment of neonates with a single injection of human ACDase-encoding lentivector diminished the severity of the disease as highlighted by enhanced growth, decreased ceramide, lessened cellular infiltrations and increased lifespans. This model of ACDase deficiency offers insights into the pathophysiology of FD and the roles of ACDase, ceramide and related sphingolipids in cell signaling and growth, as well as facilitates the development of therapy.
Collapse
|
37
|
Abstract
Sphingolipid-metabolizing enzymes are becoming targets for chemotherapeutic development with an increasing interest in the recent years. In this chapter we introduce the sphingolipid family of lipids, and the role of individual species in cell homeostasis. We also discuss their roles in several rare diseases and overall, in cancer transformation. We follow the biosynthesis pathway of the sphingolipid tree, focusing on the enzymes in order to understand how using small molecule inhibitors makes it possible to modulate cancer progression. Finally, we describe the most used and historically significant inhibitors employed in cancer research, their relationships to sphingolipid metabolism, and some promising results found in this field.
Collapse
Affiliation(s)
- Daniel Canals
- Department of Medicine, University of Stony Brook, Stony Brook, New York 11794
| | - Yusuf A. Hannun
- Health Science Center, Stony Brook University, 100 Nicolls Road, L-4, 178, Stony Brook, NY 11794, USA
| |
Collapse
|
38
|
Pratt S, Wansadhipathi-Kannangara NK, Bruce CR, Mina JG, Shams-Eldin H, Casas J, Hanada K, Schwarz RT, Sonda S, Denny PW. Sphingolipid synthesis and scavenging in the intracellular apicomplexan parasite, Toxoplasma gondii. Mol Biochem Parasitol 2012; 187:43-51. [PMID: 23246819 PMCID: PMC3629565 DOI: 10.1016/j.molbiopara.2012.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2012] [Revised: 10/03/2012] [Accepted: 11/29/2012] [Indexed: 12/26/2022]
Abstract
Sphingolipids are essential components of eukaryotic cell membranes, particularly the plasma membrane, and are involved in a diverse array of signal transduction pathways. Mammals produce sphingomyelin (SM) as the primary complex sphingolipid via the well characterised SM synthase. In contrast yeast, plants and some protozoa utilise an evolutionarily related inositol phosphorylceramide (IPC) synthase to synthesise IPC. This activity has no mammalian equivalent and IPC synthase has been proposed as a target for anti-fungals and anti-protozoals. However, detailed knowledge of the sphingolipid biosynthetic pathway of the apicomplexan protozoan parasites was lacking. In this study bioinformatic analyses indicated a single copy orthologue of the putative SM synthase from the apicomplexan Plasmodium falciparum (the causative agent of malaria) was a bona fide sphingolipid synthase in the related model parasite, Toxoplasma gondii (TgSLS). Subsequently, TgSLS was indicated, by complementation of a mutant cell line, to be a functional orthologue of the yeast IPC synthase (AUR1p), demonstrating resistance to the well characterised AUR1p inhibitor aureobasidin A. In vitro, recombinant TgSLS exhibited IPC synthase activity and, for the first time, the presence of IPC was demonstrated in T. gondii lipid extracts by mass spectrometry. Furthermore, host sphingolipid biosynthesis was indicated to influence, but be non-essential for, T. gondii proliferation, suggesting that whilst scavenging does take place de novo sphingolipid synthesis may be important for parasitism.
Collapse
Affiliation(s)
- Steven Pratt
- Biophysical Sciences Institute, Department of Chemistry and School of Biological Sciences, University Science Laboratories, Durham DH1 3LE, UK
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Lige B, Romano JD, Sampels V, Sonda S, Joiner KA, Coppens I. Introduction of caveolae structural proteins into the protozoan Toxoplasma results in the formation of heterologous caveolae but not caveolar endocytosis. PLoS One 2012; 7:e51773. [PMID: 23272165 PMCID: PMC3522706 DOI: 10.1371/journal.pone.0051773] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 11/08/2012] [Indexed: 11/30/2022] Open
Abstract
Present on the plasma membrane of most metazoans, caveolae are specialized microdomains implicated in several endocytic and trafficking mechanisms. Caveolins and the more recently discovered cavins are the major protein components of caveolae. Previous studies reported that caveolar invaginations can be induced de novo on the surface of caveolae-negative mammalian cells upon heterologous expression of caveolin-1. However, it remains undocumented whether other components in the transfected cells participate in caveolae formation. To address this issue, we have exploited the protozoan Toxoplasma as a heterologous expression system to provide insights into the minimal requirements for caveogenesis and caveolar endocytosis. Upon expression of caveolin-1, Toxoplasma accumulates prototypical exocytic caveolae 'precursors' in the cytoplasm. Toxoplasma expressing caveolin-1 alone, or in conjunction with cavin-1, neither develops surface-located caveolae nor internalizes caveolar ligands. These data suggest that the formation of functional caveolae at the plasma membrane in Toxoplasma and, by inference in all non-mammalian cells, requires effectors other than caveolin-1 and cavin-1. Interestingly, Toxoplasma co-expressing caveolin-1 and cavin-1 displays an impressive spiraled network of membranes containing the two proteins, in the cytoplasm. This suggests a synergistic activity of caveolin-1 and cavin-1 in the morphogenesis and remodeling of membranes, as illustrated for Toxoplasma.
Collapse
Affiliation(s)
- Bao Lige
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Julia D. Romano
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Vera Sampels
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| | - Sabrina Sonda
- Institute of Parasitology, University of Zurich, Zurich, Switzerland
| | - Keith A. Joiner
- Arizona Health Science Center, University of Arizona College of Medicine, Tucson, Arizona, United States of America
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health Baltimore, Maryland, United States of America
| |
Collapse
|
40
|
Dihydroceramide delays cell cycle G1/S transition via activation of ER stress and induction of autophagy. Int J Biochem Cell Biol 2012; 44:2135-43. [DOI: 10.1016/j.biocel.2012.08.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 08/18/2012] [Accepted: 08/29/2012] [Indexed: 12/31/2022]
|
41
|
Facchetti G, Zampieri M, Altafini C. Predicting and characterizing selective multiple drug treatments for metabolic diseases and cancer. BMC SYSTEMS BIOLOGY 2012; 6:115. [PMID: 22932283 PMCID: PMC3744170 DOI: 10.1186/1752-0509-6-115] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 08/13/2012] [Indexed: 12/13/2022]
Abstract
BACKGROUND In the field of drug discovery, assessing the potential of multidrug therapies is a difficult task because of the combinatorial complexity (both theoretical and experimental) and because of the requirements on the selectivity of the therapy. To cope with this problem, we have developed a novel method for the systematic in silico investigation of synergistic effects of currently available drugs on genome-scale metabolic networks. RESULTS The algorithm finds the optimal combination of drugs which guarantees the inhibition of an objective function, while minimizing the side effect on the other cellular processes. Two different applications are considered: finding drug synergisms for human metabolic diseases (like diabetes, obesity and hypertension) and finding antitumoral drug combinations with minimal side effect on the normal human cell. The results we obtain are consistent with some of the available therapeutic indications and predict new multiple drug treatments. A cluster analysis on all possible interactions among the currently available drugs indicates a limited variety on the metabolic targets for the approved drugs. CONCLUSION The in silico prediction of drug synergisms can represent an important tool for the repurposing of drugs in a realistic perspective which considers also the selectivity of the therapy. Moreover, for a more profitable exploitation of drug-drug interactions, we have shown that also experimental drugs which have a different mechanism of action can be reconsider as potential ingredients of new multicompound therapeutic indications. Needless to say the clues provided by a computational study like ours need in any case to be thoroughly evaluated experimentally.
Collapse
Affiliation(s)
- Giuseppe Facchetti
- Statistical and Biological Physics Department, SISSA (International School for Advanced Studies), Via Bonomea 265 - 34136, Trieste, Italy
| | - Mattia Zampieri
- Institute of Molecular Systems Biology, ETH (Eidgenoessische Technische Hochschule), Wolfgang Pauli Str. 16 - 8093, Zurich, Switzerland
| | - Claudio Altafini
- Functional Analysis DepartmentSISSA (International School for Advanced Studies), , Via Bonomea 265 - 34136, Trieste, Italy
| |
Collapse
|
42
|
Bikman BT, Guan Y, Shui G, Siddique MM, Holland WL, Kim JY, Fabriàs G, Wenk MR, Summers SA. Fenretinide prevents lipid-induced insulin resistance by blocking ceramide biosynthesis. J Biol Chem 2012; 287:17426-17437. [PMID: 22474281 PMCID: PMC3366851 DOI: 10.1074/jbc.m112.359950] [Citation(s) in RCA: 114] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 03/30/2012] [Indexed: 01/01/2023] Open
Abstract
Fenretinide is a synthetic retinoid that is being tested in clinical trials for the treatment of breast cancer and insulin resistance, but its mechanism of action has been elusive. Recent in vitro data indicate that fenretinide inhibits dihydroceramide desaturase, an enzyme involved in the biosynthesis of lipotoxic ceramides that antagonize insulin action. Because of this finding, we assessed whether fenretinide could improve insulin sensitivity and glucose homeostasis in vitro and in vivo by controlling ceramide production. The effect of fenretinide on insulin action and the cellular lipidome was assessed in a number of lipid-challenged models including cultured myotubes and isolated muscles strips incubated with exogenous fatty acids and mice fed a high-fat diet. Insulin action was evaluated in the various models by measuring glucose uptake or disposal and the activation of Akt/PKB, a serine/threonine kinase that is obligate for insulin-stimulated anabolism. The effects of fenretinide on cellular lipid levels were assessed by LC-MS/MS. Fenretinide negated lipid-induced insulin resistance in each of the model systems assayed. Simultaneously, the drug depleted cells of ceramide, while promoting the accumulation of the precursor dihydroceramide, a substrate for the reaction catalyzed by Des1. These data suggest that fenretinide improves insulin sensitivity, at least in part, by inhibiting Des1 and suggest that therapeutics targeting this enzyme may be a viable therapeutic means for normalizing glucose homeostasis in the overweight and diabetic.
Collapse
Affiliation(s)
- Benjamin T Bikman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, Utah 84602; Program in Cardiovascular and Metabolic Diseases, Duke-National University of Singapore Graduate Medical School, Singapore 169857.
| | - Yuguang Guan
- Program in Cardiovascular and Metabolic Diseases, Duke-National University of Singapore Graduate Medical School, Singapore 169857
| | - Guanghou Shui
- Department of Biochemistry and, National University of Singapore, Singapore 119077; Life Sciences Institute, National University of Singapore, Singapore 119077
| | - M Mobin Siddique
- Program in Cardiovascular and Metabolic Diseases, Duke-National University of Singapore Graduate Medical School, Singapore 169857
| | - William L Holland
- Department of Internal Medicine, University of Texas Southwestern Medical School, Dallas, Texas 75390
| | - Ji Yun Kim
- National University of Singapore High School of Mathematics and Science, Singapore 129957
| | - Gemma Fabriàs
- Research Unit on BioActive Molecules (RUBAM), Departament de Química Orgànica Biològica, Institut d'Investigacions Químiques i Ambientals de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), 08034 Barcelona, Spain
| | - Markus R Wenk
- Department of Biochemistry and, National University of Singapore, Singapore 119077; Life Sciences Institute, National University of Singapore, Singapore 119077; Department of Biological Sciences, National University of Singapore, Singapore 169857
| | - Scott A Summers
- Program in Cardiovascular and Metabolic Diseases, Duke-National University of Singapore Graduate Medical School, Singapore 169857; Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina 27710
| |
Collapse
|
43
|
Camacho L, Simbari F, Garrido M, Abad JL, Casas J, Delgado A, Fabriàs G. 3-Deoxy-3,4-dehydro analogs of XM462. Preparation and activity on sphingolipid metabolism and cell fate. Bioorg Med Chem 2012; 20:3173-9. [PMID: 22537678 DOI: 10.1016/j.bmc.2012.03.073] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 03/26/2012] [Accepted: 03/30/2012] [Indexed: 12/16/2022]
Abstract
Three analogs of the dihydroceramide desaturase inhibitor XM462 are reported. The compounds inhibit both dihydroceramide desaturase and acid ceramidase, but with different potencies depending on the N-acyl moiety. Other enzymes of sphingolipid metabolism, such as neutral ceramidase, acid sphingomyelinase, acid glucosylceramide hydrolase, sphingomyelin synthase and glucosylceramide synthase, are not affected. The effect on the sphingolipidome of the two best inhibitors, namely (R,E)-N-(1-hydroxy-4-(tridecylthio)but-3-en-2-yl)octanamide (RBM2-1B) and (R,E)-N-(1-hydroxy-4-(tridecylthio)but-3-en-2-yl)pivalamide (RBM2-1D), is in accordance with the results obtained in the enzyme assays. These two compounds reduce cell viability in A549 and HCT116 cell lines with similar potencies and both induced apoptotic cell death to similar levels than C8-Cer in HCT116 cells. The possible therapeutic implications of the activities of these compounds are discussed.
Collapse
Affiliation(s)
- Luz Camacho
- Research Unit on Bioactive Molecules, Department of Biomedicinal Chemistry, Institute for Advanced Chemistry of Catalonia, Spanish Council for Scientific Research, Jordi Girona 18-26, 08034 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
44
|
Spassieva SD, Rahmaniyan M, Bielawski J, Clarke CJ, Kraveka JM, Obeid LM. Cell density-dependent reduction of dihydroceramide desaturase activity in neuroblastoma cells. J Lipid Res 2012; 53:918-928. [PMID: 22377532 DOI: 10.1194/jlr.m019075] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We applied a metabolic approach to investigate the role of sphingolipids in cell density-induced growth arrest in neuroblastoma cells. Our data revealed that sphingolipid metabolism in neuroblastoma cells significantly differs depending on the cells' population context. At high cell density, cells exhibited G0/G1 cell-cycle arrest and reduced ceramide, monohexosylceramide, and sphingomyelin, whereas dihydroceramide was significantly increased. In addition, our metabolic-labeling experiments showed that neuroblastoma cells at high cell density preferentially synthesized very long chain (VLC) sphingolipids and dramatically decreased synthesis of sphingosine-1-phosphate (S1P). Moreover, densely populated neuroblastoma cells showed increased message levels of both anabolic and catabolic enzymes of the sphingolipid pathway. Notably, our metabolic-labeling experiments indicated reduced dihydroceramide desaturase activity at confluence, which was confirmed by direct measurement of dihydroceramide desaturase activity in situ and in vitro. Importantly, we could reduce dihydroceramide desaturase activity in low-density cells by applying conditional media from high-density cells, as well as by adding reducing agents, such as DTT and L-cysteine to the media. In conclusion, our data suggest a role of the sphingolipid pathway, dihydroceramides desaturase in particular, in confluence-induced growth arrest in neuroblastoma cells.
Collapse
Affiliation(s)
- Stefka D Spassieva
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425
| | - Mehrdad Rahmaniyan
- Department of Pediatrics Division of Hematology/Oncology, Medical University of South Carolina, Charleston, SC 29425; and
| | - Jacek Bielawski
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425; and
| | - Christopher J Clarke
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425; and
| | - Jacqueline M Kraveka
- Department of Pediatrics Division of Hematology/Oncology, Medical University of South Carolina, Charleston, SC 29425; and
| | - Lina M Obeid
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425; Division of General Internal Medicine, Ralph H. Johnson Veterans Affairs Hospital, Charleston, SC 29401.
| |
Collapse
|
45
|
Dihydroceramide desaturase and dihydrosphingolipids: debutant players in the sphingolipid arena. Prog Lipid Res 2011; 51:82-94. [PMID: 22200621 DOI: 10.1016/j.plipres.2011.12.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Sphingolipids are a wide family of lipids that share common sphingoid backbones, including (2S,3R)-2-amino-4-octadecane-1,3-diol (dihydrosphingosine) and (2S,3R,4E)-2-amino-4-octadecene-1,3-diol (sphingosine). The metabolism and biological functions of sphingolipids derived from sphingosine have been the subject of many reviews. In contrast, dihydrosphingolipids have received poor attention, mainly due to their supposed lack of biological activity. However, the reported biological effects of active site directed dihydroceramide desaturase inhibitors and the involvement of dihydrosphingolipids in the response of cells to known therapeutic agents support that dihydrosphingolipids are not inert but are in fact biologically active and underscore the importance of elucidating further the metabolic pathways and cell signaling networks involved in the biological activities of dihydrosphingolipids. Dihydroceramide desaturase is the enzyme involved in the conversion of dihydroceramide into ceramide and it is crucial in the regulation of the balance between sphingolipids and dihydrosphingolipids. Furthermore, given the enzyme requirement for O₂ and the NAD(P)H cofactor, the cellular redox balance and dihydroceramide desaturase activity may reciprocally influence each other. In this review both dihydroceramide desaturase and the biological functions of dihydrosphingolipids are addressed and perspectives on this field are discussed.
Collapse
|
46
|
Merrill AH. Sphingolipid and glycosphingolipid metabolic pathways in the era of sphingolipidomics. Chem Rev 2011; 111:6387-422. [PMID: 21942574 PMCID: PMC3191729 DOI: 10.1021/cr2002917] [Citation(s) in RCA: 588] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Indexed: 12/15/2022]
Affiliation(s)
- Alfred H Merrill
- School of Biology, and the Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, Georgia 30332-0230, USA.
| |
Collapse
|
47
|
Devlin CM, Lahm T, Hubbard WC, Van Demark M, Wang KC, Wu X, Bielawska A, Obeid LM, Ivan M, Petrache I. Dihydroceramide-based response to hypoxia. J Biol Chem 2011; 286:38069-38078. [PMID: 21914808 DOI: 10.1074/jbc.m111.297994] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
To understand the mechanisms of ceramide-based responses to hypoxia, we performed a mass spectrometry-based survey of ceramide species elicited by a wide range of hypoxic conditions (0.2-5% oxygen). We describe a rapid, time-dependent, marked up-regulation of dihydroceramides (DHCs) in mammalian cells and in the lungs of hypoxic rats. The increase affected all DHC species and was proportional with the depth and duration of hypoxia, ranging from 2- (1 h) to 10-fold (24 h), with complete return to normal after 1 h of reoxygenation at the expense of increased ceramides. We demonstrate that a DHC-based response to hypoxia occurs in a hypoxia-inducible factor-independent fashion and is catalyzed by the DHC desaturase (DEGS) in the de novo ceramide pathway. Both the impact of hypoxia on DHC molecular species and its inhibitory effect on cell proliferation were reproduced by knockdown of DEGS1 or DEGS2 by siRNA during normoxia. Conversely, overexpression of DEGS1 or DEGS2 attenuated the DHC accumulation and increased cell proliferation during hypoxia. Based on the amplitude and kinetics of DHC accumulation, the enzymatic desaturation of DHCs fulfills the criteria of an oxygen sensor across physiological hypoxic conditions, regulating the balance between biologically active components of ceramide metabolism.
Collapse
Affiliation(s)
- Cecilia M Devlin
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202
| | - Tim Lahm
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202; R. L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana 46202
| | - Walter C Hubbard
- Department of Medicine, The Johns Hopkins University, Baltimore, Maryland 21286
| | - Mary Van Demark
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202
| | - Kevin C Wang
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202
| | - Xue Wu
- Department of Microbiology and Immunology, Indiana University, Indianapolis, Indiana 46202
| | - Alicja Bielawska
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Lina M Obeid
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425
| | - Mircea Ivan
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202; Department of Microbiology and Immunology, Indiana University, Indianapolis, Indiana 46202.
| | - Irina Petrache
- Department of Medicine, Indiana University, Indianapolis, Indiana 46202; R. L. Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana 46202.
| |
Collapse
|
48
|
Rahmaniyan M, Curley RW, Obeid LM, Hannun YA, Kraveka JM. Identification of dihydroceramide desaturase as a direct in vitro target for fenretinide. J Biol Chem 2011; 286:24754-64. [PMID: 21543327 DOI: 10.1074/jbc.m111.250779] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The dihydroceramide desaturase (DES) enzyme is responsible for inserting the 4,5-trans-double bond to the sphingolipid backbone of dihydroceramide. We previously demonstrated that fenretinide (4-HPR) inhibited DES activity in SMS-KCNR neuroblastoma cells. In this study, we investigated whether 4-HPR acted directly on the enzyme in vitro. N-C8:0-d-erythro-dihydroceramide (C(8)-dhCer) was used as a substrate to study the conversion of dihydroceramide into ceramide in vitro using rat liver microsomes, and the formation of tritiated water after the addition of the tritiated substrate was detected and used to measure DES activity. NADH served as a cofactor. The apparent K(m) for C(8)-dhCer and NADH were 1.92 ± 0.36 μm and 43.4 ± 6.47 μm, respectively; and the V(max) was 3.16 ± 0.24 and 4.11 ± 0.18 nmol/min/g protein. Next, the effects of 4-HPR and its metabolites on DES activity were investigated. 4-HPR was found to inhibit DES in a dose-dependent manner. At 20 min, the inhibition was competitive; however, longer incubation times demonstrated the inhibition to be irreversible. Among the major metabolites of 4-HPR, 4-oxo-N-(4-hydroxyphenyl)retinamide (4-oxo-4-HPR) showed the highest inhibitory effect with substrate concentration of 0.5 μm, with an IC(50) of 1.68 μm as compared with an IC(50) of 2.32 μm for 4-HPR. N-(4-Methoxyphenyl)retinamide (4-MPR) and 4-Oxo-N-(4-methoxyphenyl)retinamide (4-oxo-4-MPR) had minimal effects on DES activity. A known competitive inhibitor of DES, C(8)-cyclopropenylceramide was used as a positive control. These studies define for the first time a direct in vitro target for 4-HPR and suggest that inhibitors of DES may be used as therapeutic interventions to regulate ceramide desaturation and consequent function.
Collapse
Affiliation(s)
- Mehrdad Rahmaniyan
- Division of Hematology/Oncology, Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
49
|
Dihydroceramide desaturase inhibition by a cyclopropanated dihydroceramide analog in cultured keratinocytes. J Lipids 2010; 2011:724015. [PMID: 21490810 PMCID: PMC3066699 DOI: 10.1155/2011/724015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/21/2010] [Accepted: 10/27/2010] [Indexed: 01/27/2023] Open
Abstract
Most mammalian sphingolipids contain a 4,5-(E)-double bond. We report on the chemical synthesis of a dihydroceramide derivative that prevents the introduction of the double bond into sphingolipids. Minimal alteration of the parent structure by formally replacing the hydrogen atoms in the 5- and in the 6-position of the sphinganine backbone by a methylene group leads to an inhibitor of dihydroceramide desaturase in cultured cells. In the presence of 10–50 μM of compound (1), levels of biosynthetically formed dihydroceramide and—surprisingly—also of phytoceramide are elevated at the expense of ceramide. The cells respond to the lack of unsaturated sphingolipids by an elevation of mRNAs of enzymes required for sphingosine formation. At the same time, the analysis of proliferation and differentiation markers indicates that the sphingolipid double bond is required to keep the cells in a differentiated state.
Collapse
|
50
|
Vieira CR, Munoz-Olaya JM, Sot J, Jiménez-Baranda S, Izquierdo-Useros N, Abad JL, Apellániz B, Delgado R, Martinez-Picado J, Alonso A, Casas J, Nieva JL, Fabriás G, Mañes S, Goñi FM. Dihydrosphingomyelin Impairs HIV-1 Infection by Rigidifying Liquid-Ordered Membrane Domains. ACTA ACUST UNITED AC 2010; 17:766-75. [DOI: 10.1016/j.chembiol.2010.05.023] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Revised: 05/13/2010] [Accepted: 05/14/2010] [Indexed: 12/20/2022]
|