1
|
Tsikandelova R, Galo E, Cerniauskas E, Hallam D, Georgiou M, Cerna-Chavez R, Atkinson R, Palmowski P, Burté F, Davies T, Steel DH, McKibbin M, Bond J, Haggarty J, Whitfield P, Korolchuk V, Armstrong L, Yang C, Dorgau B, Kurzawa-Akanbi M, Lako M. Retinal cells derived from patients with DRAM2-dependent CORD21 dystrophy exhibit key lysosomal enzyme deficiency and lysosomal content accumulation. Stem Cell Reports 2024; 19:1107-1121. [PMID: 38964324 PMCID: PMC11368688 DOI: 10.1016/j.stemcr.2024.06.002] [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: 04/04/2024] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 07/06/2024] Open
Abstract
Biallelic mutations in DRAM2 lead to an autosomal recessive cone-rod dystrophy known as CORD21, which typically presents between the third and sixth decades of life. Although DRAM2 localizes to the lysosomes of photoreceptor and retinal pigment epithelium (RPE) cells, its specific role in retinal degeneration has not been fully elucidated. In this study, we generated and characterized retinal organoids (ROs) and RPE cells from induced pluripotent stem cells (iPSCs) derived from two CORD21 patients. Our investigation revealed that CORD21-ROs and RPE cells exhibit abnormalities in lipid metabolism, defects in autophagic flux, accumulation of aberrant lysosomal content, and reduced lysosomal enzyme activity. We identified potential interactions of DRAM2 with vesicular trafficking proteins, suggesting its involvement in this cellular process. These findings collectively suggest that DRAM2 plays a crucial role in maintaining the integrity of photoreceptors and RPE cells by regulating lysosomal function, autophagy, and potentially vesicular trafficking.
Collapse
Affiliation(s)
| | - Eldo Galo
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | - Dean Hallam
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Maria Georgiou
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | | | | | - Florence Burté
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Tracey Davies
- Electron Microscopy Research Services, Newcastle University, Newcastle, UK
| | - David H Steel
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Martin McKibbin
- Leeds Teaching Hospitals NHS Trust, Leeds UK and Leeds Institute for Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Jacquelyn Bond
- Leeds Teaching Hospitals NHS Trust, Leeds UK and Leeds Institute for Medical Research, St. James's University Hospital, University of Leeds, Leeds, UK
| | - Jennifer Haggarty
- Shared Research Facilities, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Phil Whitfield
- Glasgow Polyomics and Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | | | - Lyle Armstrong
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Chunbo Yang
- Biosciences Institute, Newcastle University, Newcastle, UK
| | - Birthe Dorgau
- Biosciences Institute, Newcastle University, Newcastle, UK
| | | | - Majlinda Lako
- Biosciences Institute, Newcastle University, Newcastle, UK.
| |
Collapse
|
2
|
Mu Y, Wang Z, Song L, Ma K, Chen Y, Li P, Yan Z. Modulating lipid bilayer permeability and structure: Impact of hydrophobic chain length, C-3 hydroxyl group, and double bond in sphingosine. J Colloid Interface Sci 2024; 674:513-526. [PMID: 38943912 DOI: 10.1016/j.jcis.2024.06.171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 06/14/2024] [Accepted: 06/23/2024] [Indexed: 07/01/2024]
Abstract
Sphingosine, an amphiphilic molecule, plays a pivotal role as the core structure of sphingolipids, essential constituents of cell membranes. Its unique capability to enhance the permeability of lipid membranes profoundly influences crucial life processes. The molecular structure of sphingosine dictates its mode of entry into lipid bilayers and governs its interactions with lipids, thereby determining membrane permeability. However, the incomplete elucidation of the relationship between the molecular structure of sphingosine and the permeability of lipid membranes persists due to challenges associated with synthesizing sphingosine molecules. A series of sphingosine-derived molecules, featuring diverse hydrophobic chain lengths and distinct headgroup structure, were meticulously designed and successfully synthesized. These molecules were employed to investigate the permeability of large unilamellar vesicles, functioning as model lipid bilayers. With a decrease in the hydrophobic chain length of sphingosine from C15 to C11, the transient leakage ratio of vesicle contents escalated from ∼ 13 % to ∼ 28 %. Although the presence of double bond did not exert a pronounced influence on transient leakage, it significantly affected the continuous leakage ratio. Conversely, modifying the chirality of the C-3 hydroxyl group gives the opposite result. Notably, methylation at the C-3 hydroxyl significantly elevates transient leakage while suppressing the continuous leakage ratio. Additionally, sphingosines that significantly affect vesicle permeability tend to have a more pronounced impact on cell viability. Throughout this leakage process, the charge state of sphingosine-derived molecule aggregates in the solution emerged as a pivotal factor influencing vesicle permeability. Fluorescence lifetime experiments further revealed discernible variations in the effect of sphingosine molecular structure on the mobility of hydrophobic regions within lipid bilayers. These observed distinctions emphasize the impact of molecular structure on intermolecular interactions, extending to the microscopic architecture of membranes, and underscore the significance of subtle alterations in molecular structure and their associated aggregation behaviors in governing membrane permeability.
Collapse
Affiliation(s)
- Yonghang Mu
- State Key Laboratory of Heavy Oil Processing, College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266580, China
| | - Zi Wang
- State Key Laboratory of Heavy Oil Processing, College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266580, China.
| | - Linhua Song
- State Key Laboratory of Heavy Oil Processing, College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266580, China
| | - Kun Ma
- ISIS Facility, Rutherford Appleton Laboratory, STFC, Chilton, Didcot, Oxon OX11 0QX, UK
| | - Yao Chen
- ISIS Facility, Rutherford Appleton Laboratory, STFC, Chilton, Didcot, Oxon OX11 0QX, UK
| | - Peixun Li
- ISIS Facility, Rutherford Appleton Laboratory, STFC, Chilton, Didcot, Oxon OX11 0QX, UK
| | - Zifeng Yan
- State Key Laboratory of Heavy Oil Processing, College of Chemistry and Chemical Engineering, China University of Petroleum, Qingdao 266580, China.
| |
Collapse
|
3
|
Hempelmann P, Lolicato F, Graziadei A, Brown RDR, Spiegel S, Rappsilber J, Nickel W, Höglinger D, Jamecna D. The sterol transporter STARD3 transports sphingosine at ER-lysosome contact sites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.18.557036. [PMID: 37790546 PMCID: PMC10542139 DOI: 10.1101/2023.09.18.557036] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Sphingolipids are important structural components of membranes. Additionally, simple sphingolipids such as sphingosine are highly bioactive and participate in complex subcellular signaling. Sphingolipid deregulation is associated with many severe diseases including diabetes, Parkinson's and cancer. Here, we focus on how sphingosine, generated from sphingolipid catabolism in late endosomes/lysosomes, is reintegrated into the biosynthetic machinery at the endoplasmic reticulum (ER). We characterized the sterol transporter STARD3 as a sphingosine transporter acting at lysosome-ER contact sites. Experiments featuring crosslinkable sphingosine probes, supported by unbiased molecular dynamics simulations, exposed how sphingosine binds to the lipid-binding domain of STARD3. Following the metabolic fate of pre-localized lysosomal sphingosine showed the importance of STARD3 and its actions at contact sites for the integration of sphingosine into ceramide in a cellular context. Our findings provide the first example of interorganellar sphingosine transfer and pave the way for a better understanding of sphingolipid - sterol co-regulation.
Collapse
Affiliation(s)
- Pia Hempelmann
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Fabio Lolicato
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
- Department of Physics, University of Helsinki, FI-00014 Helsinki, Finland
| | - Andrea Graziadei
- Institute for Biotechnology, Technical University Berlin, Gustav Mayer Allee 25, 13355 Berlin
| | - Ryan D R Brown
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA
| | - Juri Rappsilber
- Institute for Biotechnology, Technical University Berlin, Gustav Mayer Allee 25, 13355 Berlin
| | - Walter Nickel
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Doris Höglinger
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| | - Denisa Jamecna
- Heidelberg University Biochemistry Center, Im Neuenheimer Feld 328, 69120 Heidelberg
| |
Collapse
|
4
|
Issleny BM, Jamjoum R, Majumder S, Stiban J. Sphingolipids: From structural components to signaling hubs. Enzymes 2023; 54:171-201. [PMID: 37945171 DOI: 10.1016/bs.enz.2023.07.003] [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] [Indexed: 11/12/2023]
Abstract
In late November 2019, Prof. Lina M. Obeid passed away from cancer, a disease she spent her life researching and studying its intricate molecular underpinnings. Along with her husband, Prof. Yusuf A. Hannun, Obeid laid down the foundations of sphingolipid biochemistry and oversaw its remarkable evolution over the years. Lipids are a class of macromolecules that are primarily associated with cellular architecture. In fact, lipids constitute the perimeter of the cell in such a way that without them, there cannot be cells. Hence, much of the early research on lipids identified the function of this class of biological molecules as merely structural. Nevertheless, unlike proteins, carbohydrates, and nucleic acids, lipids are elaborately diverse as they are not made up of monomers in polymeric forms. This diversity in structure is clearly mirrored by functional pleiotropy. In this chapter, we focus on a major subset of lipids, sphingolipids, and explore their historic rise from merely inert structural components of plasma membranes to lively and necessary signaling molecules that transmit various signals and control many cellular processes. We will emphasize the works of Lina Obeid since she was an integral pillar of the sphingolipid research world.
Collapse
Affiliation(s)
- Batoul M Issleny
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | - Rama Jamjoum
- Department of Pharmacy, Birzeit University, West Bank, Palestine
| | | | - Johnny Stiban
- Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine.
| |
Collapse
|
5
|
Spears ME, Lee N, Hwang S, Park SJ, Carlisle AE, Li R, Doshi MB, Armando AM, Gao J, Simin K, Zhu LJ, Greer PL, Quehenberger O, Torres EM, Kim D. De novo sphingolipid biosynthesis necessitates detoxification in cancer cells. Cell Rep 2022; 40:111415. [PMID: 36170811 PMCID: PMC9552870 DOI: 10.1016/j.celrep.2022.111415] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 07/21/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022] Open
Abstract
Sphingolipids play important signaling and structural roles in cells. Here, we find that during de novo sphingolipid biosynthesis, a toxic metabolite is formed with critical implications for cancer cell survival. The enzyme catalyzing the first step in this pathway, serine palmitoyltransferase complex (SPT), is upregulated in breast and other cancers. SPT is dispensable for cancer cell proliferation, as sphingolipids can be salvaged from the environment. However, SPT activity introduces a liability as its product, 3-ketodihydrosphingosine (3KDS), is toxic and requires clearance via the downstream enzyme 3-ketodihydrosphingosine reductase (KDSR). In cancer cells, but not normal cells, targeting KDSR induces toxic 3KDS accumulation leading to endoplasmic reticulum (ER) dysfunction and loss of proteostasis. Furthermore, the antitumor effect of KDSR disruption can be enhanced by increasing metabolic input (via high-fat diet) to allow greater 3KDS production. Thus, de novo sphingolipid biosynthesis entails a detoxification requirement in cancer cells that can be therapeutically exploited.
Collapse
Affiliation(s)
- Meghan E Spears
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Namgyu Lee
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Sunyoung Hwang
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Anne E Carlisle
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Rui Li
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Mihir B Doshi
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Aaron M Armando
- School of Medicine, Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Jenny Gao
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Karl Simin
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Lihua Julie Zhu
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Oswald Quehenberger
- School of Medicine, Department of Pharmacology, University of California, San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Eduardo M Torres
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA
| | - Dohoon Kim
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA 01604, USA.
| |
Collapse
|
6
|
Carreira AC, Pokorna S, Ventura AE, Walker MW, Futerman AH, Lloyd-Evans E, de Almeida RFM, Silva LC. Biophysical impact of sphingosine and other abnormal lipid accumulation in Niemann-Pick disease type C cell models. Biochim Biophys Acta Mol Cell Biol Lipids 2021; 1866:158944. [PMID: 33892149 DOI: 10.1016/j.bbalip.2021.158944] [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: 10/28/2020] [Revised: 03/08/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
Niemann-Pick disease type C (NPC) is a complex and rare pathology, which is mainly associated to mutations in the NPC1 gene. This disease is phenotypically characterized by the abnormal accumulation of multiple lipid species in the acidic compartments of the cell. Due to the complexity of stored material, a clear molecular mechanism explaining NPC pathophysiology is still not established. Abnormal sphingosine accumulation was suggested as the primary factor involved in the development of NPC, followed by the accumulation of other lipid species. To provide additional mechanistic insight into the role of sphingosine in NPC development, fluorescence spectroscopy and microscopy were used to study the biophysical properties of biological membranes using different cellular models of NPC. Addition of sphingosine to healthy CHO-K1 cells, in conditions where other lipid species are not yet accumulated, caused a rapid decrease in plasma membrane and lysosome membrane fluidity, suggesting a direct effect of sphingosine rather than a downstream event. Changes in membrane fluidity caused by addition of sphingosine were partially sustained upon impaired trafficking and metabolization of cholesterol in these cells, and could recapitulate the decrease in membrane fluidity observed in NPC1 null Chinese Hamster Ovary (CHO) cells (CHO-M12) and in cells with pharmacologically induced NPC phenotype (treated with U18666A). In summary, these results show for the first time that the fluidity of the membranes is altered in models of NPC and that these changes are in part caused by sphingosine, supporting the role of this lipid in the pathophysiology of NPC.
Collapse
Affiliation(s)
- Ana C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Sarka Pokorna
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel; Heyrovský Institute of Physical Chemistry of the Czech Academy of Sciences, Dolejškova 3, 182 23 Prague, Czech Republic
| | - Ana E Ventura
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel; iBB-Institute for Bioengineering and Biosciences, Department of Bioengineering, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, Lisboa, Portugal
| | - Mathew W Walker
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Anthony H Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Emyr Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Museum Avenue, Cardiff, UK
| | - Rodrigo F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal.
| | - Liana C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
7
|
Torres-Luna C, Hu N, Domszy R, Fan X, Yang J, Briber RM, Wang NS, Yang A. Effect of Carbon Chain Length, Ionic Strength, and pH on the In Vitro Release Kinetics of Cationic Drugs from Fatty-Acid-Loaded Contact Lenses. Pharmaceutics 2021; 13:pharmaceutics13071060. [PMID: 34371751 PMCID: PMC8309118 DOI: 10.3390/pharmaceutics13071060] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/01/2021] [Accepted: 07/05/2021] [Indexed: 11/17/2022] Open
Abstract
This paper explores the use of fatty acids in silicone hydrogel contact lenses for extending the release duration of cationic drugs. Drug release kinetics was dependent on the carbon chain length of the fatty acid loaded in the lens, with 12-, 14- and 18-carbon chain length fatty acids increasing the uptake and the release duration of ketotifen fumarate (KTF) and tetracaine hydrochloride (THCL). Drug release kinetics from oleic acid-loaded lenses was evaluated in phosphate buffer saline (PBS) at different ionic strengths (I = 167, 500, 1665 mM); the release duration of KTF and THCL was decreased with increasing ionic strength of the release medium. Furthermore, the release of KTF and THCL in deionized water did not show a burst and was significantly slower compared to that in PBS. The release kinetics of KTF and THCL was significantly faster when the pH of the release medium was decreased from 7.4 towards 5.5 because of the decrease in the relative amounts of oleate anions in the lens mostly populated at the polymer–pore interfaces. The use of boundary charges at the polymer–pore interfaces of a contact lens to enhance drug partition and extend its release is further confirmed by loading cationic phytosphingosine in contact lenses to attract an anionic drug.
Collapse
Affiliation(s)
- Cesar Torres-Luna
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD 20740, USA;
- Lynthera Corporation, 1200 Corporate Blvd., STE 10C, Lancaster, PA 17601, USA; (N.H.); (R.D.); (J.Y.)
| | - Naiping Hu
- Lynthera Corporation, 1200 Corporate Blvd., STE 10C, Lancaster, PA 17601, USA; (N.H.); (R.D.); (J.Y.)
| | - Roman Domszy
- Lynthera Corporation, 1200 Corporate Blvd., STE 10C, Lancaster, PA 17601, USA; (N.H.); (R.D.); (J.Y.)
| | - Xin Fan
- Department of Chemical Engineering, Auburn University, Auburn, AL 36849, USA;
| | - Jeff Yang
- Lynthera Corporation, 1200 Corporate Blvd., STE 10C, Lancaster, PA 17601, USA; (N.H.); (R.D.); (J.Y.)
| | - Robert M. Briber
- Department of Materials Science & Engineering, University of Maryland, College Park, MD 20740, USA;
| | - Nam Sun Wang
- Department of Chemical & Biomolecular Engineering, University of Maryland, College Park, MD 20740, USA;
- Correspondence: (N.S.W.); (A.Y.); Tel.: +1-(301)-405-1910 (N.S.W.); +1-(717)-522-1739 (A.Y.)
| | - Arthur Yang
- Lynthera Corporation, 1200 Corporate Blvd., STE 10C, Lancaster, PA 17601, USA; (N.H.); (R.D.); (J.Y.)
- Correspondence: (N.S.W.); (A.Y.); Tel.: +1-(301)-405-1910 (N.S.W.); +1-(717)-522-1739 (A.Y.)
| |
Collapse
|
8
|
Rohrhofer J, Zwirzitz B, Selberherr E, Untersmayr E. The Impact of Dietary Sphingolipids on Intestinal Microbiota and Gastrointestinal Immune Homeostasis. Front Immunol 2021; 12:635704. [PMID: 34054805 PMCID: PMC8160510 DOI: 10.3389/fimmu.2021.635704] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022] Open
Abstract
The large surfaces of gastrointestinal (GI) organs are well adapted to their diverse tasks of selective nutritional uptake and defense against the external environment. To maintain a functional balance, a vast number of immune cells is located within the mucosa. A strictly regulated immune response is required to impede constant inflammation and to maintain barrier function. An increasing prevalence of GI diseases has been reported in Western societies over the past decades. This surge in GI disorders has been linked to dietary changes followed by an imbalance of the gut microbiome, leading to a chronic, low grade inflammation of the gut epithelium. To counteract the increasing health care costs associated with diseases, it is paramount to understand the mechanisms driving immuno-nutrition, the associations between nutritional compounds, the commensal gut microbiota, and the host immune response. Dietary compounds such as lipids, play a central role in GI barrier function. Bioactive sphingolipids (SLs), e.g. sphingomyelin (SM), sphingosine (Sph), ceramide (Cer), sphingosine-1- phosphate (S1P) and ceramide-1-phosphate (C1P) may derive from dietary SLs ingested through the diet. They are not only integral components of cell membranes, they additionally modulate cell trafficking and are precursors for mediators and second messenger molecules. By regulating intracellular calcium levels, cell motility, cell proliferation and apoptosis, SL metabolites have been described to influence GI immune homeostasis positively and detrimentally. Furthermore, dietary SLs are suggested to induce a shift in the gut microbiota. Modes of action range from competing with the commensal bacteria for intestinal cell attachment to prevention from pathogen invasion by regulating innate and immediate defense mechanisms. SL metabolites can also be produced by gut microorganisms, directly impacting host metabolic pathways. This review aims to summarize recent findings on SL signaling and functional variations of dietary SLs. We highlight novel insights in SL homeostasis and SL impact on GI barrier function, which is directly linked to changes of the intestinal microbiota. Knowledge gaps in current literature will be discussed to address questions relevant for understanding the pivotal role of dietary SLs on chronic, low grade inflammation and to define a balanced and healthy diet for disease prevention and treatment.
Collapse
Affiliation(s)
- Johanna Rohrhofer
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| | - Benjamin Zwirzitz
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Evelyne Selberherr
- Unit of Food Microbiology, Institute of Food Safety, Food Technology and Veterinary Public Health, University of Veterinary Medicine, Vienna, Austria
| | - Eva Untersmayr
- Gastrointestinal Immunology Group, Institute of Pathophysiology and Allergy Research, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Nilam M, Collin S, Karmacharya S, Hennig A, Nau WM. Membrane Permeability and Its Activation Energies in Dependence on Analyte, Lipid, and Phase Type Obtained by the Fluorescent Artificial Receptor Membrane Assay. ACS Sens 2021; 6:175-182. [PMID: 33347764 DOI: 10.1021/acssensors.0c02064] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Time-resolved monitoring of the permeability of analytes is of utmost importance in membrane research. Existing methods are restricted to single-point determinations or flat synthetic membranes, limiting access to biologically relevant kinetic parameters (permeation rate constant, permeation coefficients). We now use the recently introduced fluorescent artificial receptor membrane assay (FARMA) as a method to monitor, in real time, the permeation of indole derivatives through liposomal membranes of different lipid compositions. This method is based on the liposomal encapsulation of a chemosensing ensemble or "fluorescent artificial receptor", consisting of 2,7-dimethyldiazapyrenium as a fluorescent dye and cucurbit[8]uril as the macrocyclic receptor, that responds to the complexation of a permeating aromatic analyte by fluorescence quenching. FARMA does not require a fluorescent labeling of the analytes and allows access to permeability coefficients in the range from 10-8 to 10-4 cm s-1. The effect of temperature on the permeation rate of a series of indole derivatives across the phospholipid membranes was studied. The activation energies for permeation through POPC/POPS phospholipid membranes were in the range of 28-96 kJ mol-1. To study the effect of different lipid phases on the membrane permeability, we performed experiments with DPPC/DOPS vesicles, which showed a phase transition from a gel phase to a liquid-crystalline phase, where the activation energies for the permeation process were expected to show a dramatic change. Accordingly, for the permeation of the indole derivatives into the DPPC/DOPS liposomes, discontinuities were observed in the Arrhenius plots, from which the permeation activation energies for the distinct phases could be determined, for example, for tryptamine 245 kJ mol-1 in the gel phase and 47 kJ mol-1 in the liquid-crystalline phase.
Collapse
Affiliation(s)
- Mohamed Nilam
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
- Institute for Chemistry of New Materials, University of Osnabrück, Barbarastraße 7, 49069 Osnabrück, Germany
| | - Solène Collin
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
| | - Shreya Karmacharya
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
| | - Andreas Hennig
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
- Institute for Chemistry of New Materials, University of Osnabrück, Barbarastraße 7, 49069 Osnabrück, Germany
| | - Werner M. Nau
- Department of Life Sciences and Chemistry, Jacobs University Bremen, Campus Ring 1, 28759 Bremen, Germany
| |
Collapse
|
10
|
Pant DC, Aguilera-Albesa S, Pujol A. Ceramide signalling in inherited and multifactorial brain metabolic diseases. Neurobiol Dis 2020; 143:105014. [PMID: 32653675 DOI: 10.1016/j.nbd.2020.105014] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 06/13/2020] [Accepted: 07/07/2020] [Indexed: 12/14/2022] Open
Abstract
In recent years, research on sphingolipids, particularly ceramides, has attracted increased attention, revealing the important roles and many functions of these molecules in several human neurological disorders. The nervous system is enriched with important classes of sphingolipids, e.g., ceramide and its derivatives, which compose the major portion of this group, particularly in the form of myelin. Ceramides have also emerged as important nodes for lipid signalling, both inside the cell and between cells. Until recently, knowledge about ceramides in the nervous system was limited, but currently, multiple links between ceramide signalling and neurological diseases have been reported. Alterations in the regulation of ceramide pathobiology have been shown to influence the risk of developing neurometabolic diseases. Thus, these molecules are critically important in the maintenance and development of the nervous system and are culprits or major contributors to the development of brain disorders, either inherited or multifactorial. In the present review, we highlight the critical role of ceramide signalling in several different neurological disorders as well as the effects of their perturbations and discuss how this emerging class of bioactive sphingolipids has attracted interest in the field of neurological diseases.
Collapse
Affiliation(s)
- Devesh C Pant
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Sergio Aguilera-Albesa
- Pediatric Neurology Unit, Department of Pediatrics, Navarra Health Service Hospital, Irunlarrea 4, 310620 Pamplona, Spain; Navarrabiomed-Miguel Servet Research Foundation, Pamplona, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, IDIBELL, Hospital Duran i Reynals, Gran Via 199, 08908, L'Hospitalet de Llobregat, Barcelona, Spain; Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Catalonia, Spain; Center for Biomedical Research on Rare Diseases (CIBERER), ISCIII, Madrid, Spain.
| |
Collapse
|
11
|
Allende G, Chávez-Reyes J, Guerrero-Alba R, Vázquez-León P, Marichal-Cancino BA. Advances in Neurobiology and Pharmacology of GPR12. Front Pharmacol 2020; 11:628. [PMID: 32457622 PMCID: PMC7226366 DOI: 10.3389/fphar.2020.00628] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 04/21/2020] [Indexed: 01/19/2023] Open
Abstract
GPR12 is a G protein-coupled orphan receptor genetically related to type 1 and type 2 cannabinoid receptors (CB1 and CB2) which are ancient proteins expressed all over the body. Both cannabinoid receptors, but especially CB1, are involved in neurodevelopment and cognitive processes such as learning, memory, brain reward, coordination, etc. GPR12 shares with CB1 that both are mainly expressed into the brain. Regrettably, very little is known about physiology of GPR12. Concerning its pharmacology, GPR12 seems to be endogenously activated by the lysophospholipids sphingosine-1-phosphate (S1P) and sphingosyl-phosphorylcholine (SPC). Exogenously, GPR12 is a target for the phytocannabinoid cannabidiol (CBD). Functionally, GPR12 seems to be related to neurogenesis and neural inflammation, but its relationship with cognitive functions remains to be characterized. Although GPR12 was initially suggested to be a cannabinoid receptor, it does not meet the five criteria proposed in 2010 by the International Union of Basic and Clinical Pharmacology (IUPHAR). In this review, we analyze all the direct available information in PubMed database about expression, function, and pharmacology of this receptor in central nervous system (CNS) trying to provide a broad overview of its current and prospective neurophysiology. Moreover, in this mini-review we highlight the need to produce more relevant data about the functions of GPR12 in CNS. Hence, this work should motivate further research in this field.
Collapse
Affiliation(s)
- Gonzalo Allende
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, Aguascalientes, Mexico
| | - Jesús Chávez-Reyes
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, Aguascalientes, Mexico
| | - Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, Aguascalientes, Mexico
| | - Priscila Vázquez-León
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, Aguascalientes, Mexico
| | - Bruno A Marichal-Cancino
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Ciudad Universitaria, Aguascalientes, Mexico
| |
Collapse
|
12
|
Gutiérrez-Nájera NA, Saucedo-García M, Noyola-Martínez L, Vázquez-Vázquez C, Palacios-Bahena S, Carmona-Salazar L, Plasencia J, El-Hafidi M, Gavilanes-Ruiz M. Sphingolipid Effects on the Plasma Membrane Produced by Addition of Fumonisin B1 to Maize Embryos. PLANTS (BASEL, SWITZERLAND) 2020; 9:E150. [PMID: 31979343 PMCID: PMC7076497 DOI: 10.3390/plants9020150] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/15/2022]
Abstract
Fumonisin B1 is a mycotoxin produced by Fusarium verticillioides that modifies the membrane properties from animal cells and inhibits complex sphingolipids synthesis through the inhibition of ceramide synthase. The aim of this work was to determine the effect of Fumonisin B1 on the plant plasma membrane when the mycotoxin was added to germinating maize embryos. Fumonisin B1 addition to the embryos diminished plasma membrane fluidity, increased electrolyte leakage, caused a 7-fold increase of sphinganine and a small decrease in glucosylceramide in the plasma membrane, without affecting phytosphingosine levels or fatty acid composition. A 20%-30% inhibition of the plasma membrane H+-ATPase activity was observed when embryos were germinated in the presence of the mycotoxin. Such inhibition was only associated to the decrease in glucosylceramide and the addition of exogenous ceramide to the embryos relieved the inhibition of Fumonisin B1. These results indicate that exposure of the maize embryos for 24 h to Fumonisin B1 allowed the mycotoxin to target ceramide synthase at the endoplasmic reticulum, eliciting an imbalance of endogenous sphingolipids. The latter disrupted membrane properties and inhibited the plasma membrane H+-ATPase activity. Altogether, these results illustrate the mode of action of the pathogen and a plant defense strategy.
Collapse
Affiliation(s)
- Nora A. Gutiérrez-Nájera
- Instituto Nacional de Medicina Genómica. Periférico Sur 4124, Torre 2, 5° piso. Álvaro Obregón 01900, Cd. de México, Mexico;
| | - Mariana Saucedo-García
- Instituto de Ciencias Agropecuarias, Universidad Autónoma del Estado de Hidalgo, Avenida Universidad Km. 1, Rancho Universitario, Tulancingo-Santiago, Tulantepec, Tulancingo 43600, Hidalgo, Mexico;
| | - Liliana Noyola-Martínez
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| | - Christian Vázquez-Vázquez
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| | - Silvia Palacios-Bahena
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| | - Laura Carmona-Salazar
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| | - Javier Plasencia
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| | - Mohammed El-Hafidi
- Departamento de Bioquímica. Instituto Nacional de Cardiología “Ignacio Chávez”. Juan Badiano 1. Tlalpan 14080, Cd. de México, Mexico;
| | - Marina Gavilanes-Ruiz
- Departamento de Bioquímica, Facultad de Química, UNAM. Cd. Universitaria. Coyoacán 04510, Cd. de México, Mexico; (L.N.-M.); (C.V.-V.); (S.P.-B.); (L.C.-S.); (J.P.)
| |
Collapse
|
13
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019:100995. [PMID: 31445071 DOI: 10.1016/j.plipres.2019.100995] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/19/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, 1749-016 Lisboa, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Switzerland; Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN) and IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
14
|
Carreira AC, Santos TC, Lone MA, Zupančič E, Lloyd-Evans E, de Almeida RFM, Hornemann T, Silva LC. Mammalian sphingoid bases: Biophysical, physiological and pathological properties. Prog Lipid Res 2019; 75:100988. [PMID: 31132366 DOI: 10.1016/j.plipres.2019.100988] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Revised: 05/17/2019] [Accepted: 05/21/2019] [Indexed: 12/11/2022]
Abstract
Sphingoid bases encompass a group of long chain amino alcohols which form the essential structure of sphingolipids. Over the last years, these amphiphilic molecules were moving more and more into the focus of biomedical research due to their role as bioactive molecules. In fact, free sphingoid bases interact with specific receptors and target molecules, and have been associated with numerous biological and physiological processes. In addition, they can modulate the biophysical properties of biological membranes. Several human diseases are related to pathological changes in the structure and metabolism of sphingoid bases. Yet, the mechanisms underlying their biological and pathophysiological actions remain elusive. Within this review, we aimed to summarize the current knowledge on the biochemical and biophysical properties of the most common sphingoid bases and to discuss their importance in health and disease.
Collapse
Affiliation(s)
- A C Carreira
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal; Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - T C Santos
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal; Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - M A Lone
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - E Zupančič
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal
| | - E Lloyd-Evans
- Sir Martin Evans Building, School of Biosciences, Cardiff University, Cardiff, UK
| | - R F M de Almeida
- Centro de Química e Bioquímica (CQB) e Centro de Química Estrutural (CQE), Faculdade de Ciências, Universidade de Lisboa, Ed. C8, Campo Grande, Lisboa 1749-016, Portugal
| | - T Hornemann
- Institute for Clinical Chemistry, University Hospital Zurich, Zurich Center for Integrative Human Physiology (ZIHP), University of Zurich, Switzerland
| | - L C Silva
- iMed.ULisboa - Research Institute for Medicines, Faculdade de Farmácia, Universidade de Lisboa, Av. Prof. Gama Pinto, Lisboa 1649-003, Portugal; Centro de Química-Física Molecular - Institute of Nanoscience and Nanotechnology (CQFM-IN), IBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
15
|
Gros M, Amigorena S. Regulation of Antigen Export to the Cytosol During Cross-Presentation. Front Immunol 2019; 10:41. [PMID: 30745902 PMCID: PMC6360170 DOI: 10.3389/fimmu.2019.00041] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 01/09/2019] [Indexed: 02/02/2023] Open
Abstract
Cross-priming refers to the induction of primary cytotoxic CD8+ T cell responses to antigens that are not expressed in antigen presenting cells (APCs) responsible for T cell priming. Cross-priming is achieved through cross-presentation of exogenous antigens derived from tumors, extracellular pathogens or infected neighboring cells on Major Histocompatibility Complex (MHC) class I molecules. Despite extensive research efforts to understand the intracellular pathways involved in antigen cross-presentation, certain critical steps remain elusive and controversial. Here we review recent advances on antigen cross-presentation, focusing on the mechanisms involved in antigen export to the cytosol, a crucial step of this pathway.
Collapse
|
16
|
Zulueta Díaz YDLM, Caby S, Bongarzone ER, Fanani ML. Psychosine remodels model lipid membranes at neutral pH. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:2515-2526. [PMID: 30267657 DOI: 10.1016/j.bbamem.2018.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/24/2018] [Accepted: 09/25/2018] [Indexed: 01/01/2023]
Abstract
β-Galactosylsphingosine or psychosine (PSY) is a single chain sphingolipid with a cationic group, which is degraded in the lysosome lumen by β-galactosylceramidase during sphingolipid biosynthesis. A deficiency of this enzyme activity results in Krabbe's disease and PSY accumulation. This favors its escape to extralysosomal spaces, with its pH changing from acidic to neutral. We studied the interaction of PSY with model lipid membranes in neutral conditions, using phospholipid vesicles and monolayers as classical model systems, as well as a complex lipid mixture that mimics the lipid composition of myelin. At pH 7.4, when PSY is mainly neutral, it showed high surface activity, self-organizing into large structures, probably lamellar in nature, with a CMC of 38 ± 3 μM. When integrated into phospholipid membranes, PSY showed preferential partition into disordered phases, shifting phase equilibrium. The presence of PSY reduces the compactness of the membrane, making it more easily compressible. It also induces lipid domain disruption in vesicles composed of the main myelin lipids. The surface electrostatics of lipid membranes was altered by PSY in a complex manner. A shift to positive zeta potential values evidenced its presence in the vesicles. Furthermore, the increase of surface potential and surface water structuring observed may be a consequence of its location at the interface of the positively charged layer. As Krabbe's disease is a demyelinating process, PSY alteration of the membrane phase state, lateral lipid distribution and surface electrostatics appears important to the understanding of myelin destabilization at the supramolecular level.
Collapse
Affiliation(s)
- Yenisleidy de Las Mercedes Zulueta Díaz
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Ciudad Universitaria, Haya de la Torre y Medina Allende, Córdoba X5000HUA, Argentina
| | - Sofia Caby
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, United States of America; Departamento de Química Biológica, IQUIFIB, Universidad Nacional de Buenos Aires, Buenos Aires, Argentina
| | - María Laura Fanani
- Departamento de Química Biológica Ranwel Caputto, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina; Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), CONICET, Ciudad Universitaria, Haya de la Torre y Medina Allende, Córdoba X5000HUA, Argentina.
| |
Collapse
|
17
|
Abbineni PS, Coorssen JR. Sphingolipids modulate docking, Ca 2+ sensitivity and membrane fusion of native cortical vesicles. Int J Biochem Cell Biol 2018; 104:43-54. [PMID: 30195064 DOI: 10.1016/j.biocel.2018.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/31/2018] [Accepted: 09/01/2018] [Indexed: 12/16/2022]
Abstract
Docking, priming, and membrane fusion of secretory vesicles (i.e. regulated exocytosis) requires lipids and proteins. Sphingolipids, in particular, sphingosine and sphingosine-1-phosphate, have been implicated in the modulation of exocytosis. However, the specific exocytotic steps that sphingolipids modulate and the enzymes that regulate sphingolipid concentrations on native secretory vesicle membranes remain unknown. Here we use tightly coupled functional and molecular analyses of fusion-ready cell surface complexes and cortical vesicles isolated from oocytes to assess the role of sphingolipids in the late, Ca2+-triggered steps of exocytosis. The molecular changes resulting from treatments with sphingolipid modifying compounds coupled with immunoblotting analysis revealed the presence of sphingosine kinase on native vesicles; the presence of a sphingosine-1-phosphate phosphatase is also indicated. Changes in sphingolipid concentrations on vesicles altered their docking/priming, Ca2+-sensitivity, and ability to fuse, indicating that sphingolipid concentrations are tightly regulated and maintained at optimal levels and ratios to ensure efficient exocytosis.
Collapse
Affiliation(s)
- Prabhodh S Abbineni
- Department of Molecular Physiology, and the WSU Molecular Medicine Research Group, School of Medicine, Western Sydney University, Campbelltown, NSW, 2560, Australia
| | - Jens R Coorssen
- Department of Health Sciences, Faculty of Applied Health Sciences, Department of Biology, Faculty of Mathematics and Science, Brock University, St. Catharines, Ontario, Canada.
| |
Collapse
|
18
|
Nieto-Garai JA, Glass B, Bunn C, Giese M, Jennings G, Brankatschk B, Agarwal S, Börner K, Contreras FX, Knölker HJ, Zankl C, Simons K, Schroeder C, Lorizate M, Kräusslich HG. Lipidomimetic Compounds Act as HIV-1 Entry Inhibitors by Altering Viral Membrane Structure. Front Immunol 2018; 9:1983. [PMID: 30233582 PMCID: PMC6131562 DOI: 10.3389/fimmu.2018.01983] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/13/2018] [Indexed: 12/16/2022] Open
Abstract
The envelope of Human Immunodeficiency Virus type 1 (HIV-1) consists of a liquid-ordered membrane enriched in raft lipids and containing the viral glycoproteins. Previous studies demonstrated that changes in viral membrane lipid composition affecting membrane structure or curvature can impair infectivity. Here, we describe novel antiviral compounds that were identified by screening compound libraries based on raft lipid-like scaffolds. Three distinct molecular structures were chosen for mode-of-action studies, a sterol derivative (J391B), a sphingosine derivative (J582C) and a long aliphatic chain derivative (IBS70). All three target the viral membrane and inhibit virus infectivity at the stage of fusion without perturbing virus stability or affecting virion-associated envelope glycoproteins. Their effect did not depend on the expressed envelope glycoproteins or a specific entry route, being equally strong in HIV pseudotypes carrying VSV-G or MLV-Env glycoproteins. Labeling with laurdan, a reporter of membrane order, revealed different membrane structure alterations upon compound treatment of HIV-1, which correlated with loss of infectivity. J582C and IBS70 decreased membrane order in distinctive ways, whereas J391B increased membrane order. The compounds' effects on membrane order were reproduced in liposomes generated from extracted HIV lipids and thus independent both of virion proteins and of membrane leaflet asymmetry. Remarkably, increase of membrane order by J391B required phosphatidylserine, a lipid enriched in the HIV envelope. Counterintuitively, mixtures of two compounds with opposite effects on membrane order, J582C and J391B, did not neutralize each other but synergistically inhibited HIV infection. Thus, altering membrane order, which can occur by different mechanisms, constitutes a novel antiviral mode of action that may be of general relevance for enveloped viruses and difficult to overcome by resistance development.
Collapse
Affiliation(s)
- Jon Ander Nieto-Garai
- Departamento de Bioquímica y Biología Molecular, Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Bilbao, Spain
| | - Bärbel Glass
- Department of Infectious Diseases, Virology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | | | | | - Beate Brankatschk
- JADO Technologies, Dresden, Germany.,Membrane Biochemistry Group, Paul-Langerhans-Institute Dresden, Helmholtz Zentrum München at the University Hospital and Faculty of Medicine Carl Gustav Carus, Dresden, Germany
| | - Sameer Agarwal
- JADO Technologies, Dresden, Germany.,Department of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Kathleen Börner
- Department of Infectious Diseases, Virology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | - F Xabier Contreras
- Departamento de Bioquímica y Biología Molecular, Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Bilbao, Spain.,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | - Hans-Joachim Knölker
- JADO Technologies, Dresden, Germany.,Department of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Claudia Zankl
- JADO Technologies, Dresden, Germany.,Department of Chemistry, Technische Universität Dresden, Dresden, Germany
| | - Kai Simons
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Cornelia Schroeder
- JADO Technologies, Dresden, Germany.,Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany.,Department of Anatomy, Medical Faculty Carl-Gustav-Carus, Technische Universität Dresden, Dresden, Germany
| | - Maier Lorizate
- Departamento de Bioquímica y Biología Molecular, Instituto Biofisika (CSIC, UPV/EHU), Universidad del País Vasco, Bilbao, Spain
| | - Hans-Georg Kräusslich
- Department of Infectious Diseases, Virology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
19
|
Rasouli S, Abdolvahabi A, Croom CM, Plewman DL, Shi Y, Shaw BF. Glycerolipid Headgroups Control Rate and Mechanism of Superoxide Dismutase-1 Aggregation and Accelerate Fibrillization of Slowly Aggregating Amyotrophic Lateral Sclerosis Mutants. ACS Chem Neurosci 2018; 9:1743-1756. [PMID: 29649360 DOI: 10.1021/acschemneuro.8b00086] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Interactions between superoxide dismutase-1 (SOD1) and lipid membranes might be directly involved in the toxicity and intercellular propagation of aggregated SOD1 in amyotrophic lateral sclerosis (ALS), but the chemical details of lipid-SOD1 interactions and their effects on SOD1 aggregation remain unclear. This paper determined the rate and mechanism of nucleation of fibrillar apo-SOD1 catalyzed by liposomal surfaces with identical hydrophobic chains (RCH2(O2C18H33)2), but headgroups of different net charge and hydrophobicity (i.e., R(CH2)N+(CH3)3, RPO4-(CH2)2N+(CH3)3, and RPO4-). Under semiquiescent conditions (within a 96 well microplate, without a gyrating bead), the aggregation of apo-SOD1 into thioflavin-T-positive (ThT(+)) amyloid fibrils did not occur over 120 h in the absence of liposomal surfaces. Anionic liposomes triggered aggregation of apo-SOD1 into ThT(+) amyloid fibrils; cationic liposomes catalyzed fibrillization but at slower rates and across a narrower lipid concentration; zwitterionic liposomes produced nonfibrillar (amorphous) aggregates. The inability of zwitterionic liposomes to catalyze fibrillization and the dependence of fibrillization rate on anionic lipid concentration suggests that membranes catalyze SOD1 fibrillization by a primary nucleation mechanism. Membrane-catalyzed fibrillization was also examined for eight ALS variants of apo-SOD1, including G37R, G93R, D90A, and E100G apo-SOD1 that nucleate slower than or equal to WT SOD1 in lipid-free, nonquiescent amyloid assays. All ALS variants (with one exception) nucleated faster than WT SOD1 in the presence of anionic liposomes, wherein the greatest acceleratory effects were observed among variants with lower net negative surface charge (G37R, G93R, D90A, E100G). The exception was H46R apo-SOD1, which did not form ThT(+) species.
Collapse
Affiliation(s)
- Sanaz Rasouli
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
- Institute of Biomedical Studies, Baylor University, Waco, Texas 76706, United States
| | - Alireza Abdolvahabi
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Corbin M. Croom
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Devon L. Plewman
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Yunhua Shi
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| | - Bryan F. Shaw
- Department of Chemistry and Biochemistry, Baylor University, Waco, Texas 76706, United States
| |
Collapse
|
20
|
Vutukuri R, Brunkhorst R, Kestner RI, Hansen L, Bouzas NF, Pfeilschifter J, Devraj K, Pfeilschifter W. Alteration of sphingolipid metabolism as a putative mechanism underlying LPS-induced BBB disruption. J Neurochem 2017; 144:172-185. [PMID: 29023711 DOI: 10.1111/jnc.14236] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 09/24/2017] [Accepted: 09/25/2017] [Indexed: 12/31/2022]
Abstract
Septic encephalopathy with confusion and agitation occurs early during sepsis and contributes to the severity of the disease. A decrease in the sphingosine-1-phosphate (S1P) blood levels has been shown in patients and in animal models of sepsis. The lipid mediator S1P is known to be involved in endothelial barrier function in a context-dependent manner. We utilized lipopolysaccharide (LPS)-injected mice as a model for septic encephalopathy and first performed tracer permeability assays to assess the blood-brain barrier (BBB) breakdown in vivo. At time points corresponding to the BBB breakdown post LPS injection, we aimed to characterize the regulation of the sphingolipid signaling pathway at the BBB during sepsis. We measured sphingolipid concentrations in blood, in mouse brain microvessels (MBMVs), and brain tissue. We also analyzed the expression of S1P receptors, transporters, and metabolizing enzymes in MBMVs and brain tissue. Primary mouse brain microvascular endothelial cells (MBMECs) were isolated to evaluate the effects of LPS on transendothelial electrical resistance (TEER) as a measure of permeability in vitro. We observed a relevant decrease in S1P levels after LPS injection in all three compartments (blood, MBMVs, brain tissue) that was accompanied by an increased expression of the S1P receptor type 1 and of sphingosine kinase 1 on one hand and of the S1P degrading enzymes lipid phosphate phosphatase 1 (LPP1) and S1P phosphatase 1 on the other hand, as well as a down-regulation of sphingosine kinase 2. Application of LPS to a monolayer of primary MBMECs did not alter TEER, but serum from LPS-treated mice lead to a breakdown of the barrier compared to serum from vehicle-treated mice. We observed profound alterations of the sphingolipid metabolism at the BBB after LPS injection that point toward a therapeutic potential of drugs interfering with this pathway as novel approach for the detrimental overwhelming immune response in sepsis. Read the Editorial Highlight for this article on page 115. Cover Image for this Issue: doi. 10.1111/jnc.14161.
Collapse
Affiliation(s)
- Rajkumar Vutukuri
- Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Robert Brunkhorst
- Department of Neurology, University Hospital Frankfurt and Goethe University, Frankfurt, Germany
| | - Roxane-Isabelle Kestner
- Department of Neurology, University Hospital Frankfurt and Goethe University, Frankfurt, Germany
| | - Lena Hansen
- Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Nerea Ferreiros Bouzas
- Pharmazentrum Frankfurt, Institute for Clinical Pharmacology, Goethe University, Frankfurt, Germany
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt, Germany
| | - Kavi Devraj
- Pharmazentrum Frankfurt, Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt, Germany.,Edinger Institute of Neurology, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Waltraud Pfeilschifter
- Department of Neurology, University Hospital Frankfurt and Goethe University, Frankfurt, Germany
| |
Collapse
|
21
|
Hepatocyte nuclear factor 1A deficiency causes hemolytic anemia in mice by altering erythrocyte sphingolipid homeostasis. Blood 2017; 130:2786-2798. [PMID: 29109103 DOI: 10.1182/blood-2017-03-774356] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 10/25/2017] [Indexed: 12/18/2022] Open
Abstract
The hepatocyte nuclear factor (HNF) family regulates complex networks of metabolism and organ development. Human mutations in its prototypical member HNF1A cause maturity-onset diabetes of the young (MODY) type 3. In this study, we identified an important role for HNF1A in the preservation of erythrocyte membrane integrity, calcium homeostasis, and osmotic resistance through an as-yet unrecognized link of HNF1A to sphingolipid homeostasis. HNF1A-/- mice displayed microcytic hypochromic anemia with reticulocytosis that was partially compensated by avid extramedullary erythropoiesis at all erythroid stages in the spleen thereby excluding erythroid differentiation defects. Morphologically, HNF1A-/- erythrocytes resembled acanthocytes and displayed increased phosphatidylserine exposure, high intracellular calcium, and elevated osmotic fragility. Sphingolipidome analysis by mass spectrometry revealed substantial and tissue-specific sphingolipid disturbances in several tissues including erythrocytes with the accumulation of sphingosine as the most prominent common feature. All HNF1A-/- erythrocyte defects could be simulated by exposure of wild-type (WT) erythrocytes to sphingosine in vitro and attributed in part to sphingosine-induced suppression of the plasma-membrane Ca2+-ATPase activity. Bone marrow transplantation rescued the anemia phenotype in vivo, whereas incubation with HNF1A-/- plasma increased the osmotic fragility of WT erythrocytes in vitro. Our data suggest a non-cell-autonomous erythrocyte defect secondary to the sphingolipid changes caused by HNF1A deficiency. Transcriptional analysis revealed 4 important genes involved in sphingolipid metabolism to be deregulated in HNF1A deficiency: Ormdl1, sphingosine kinase-2, neutral ceramidase, and ceramide synthase-5. The considerable erythrocyte defects in murine HNF1A deficiency encourage clinical studies to explore the hematological consequences of HNF1A deficiency in human MODY3 patients.
Collapse
|
22
|
Development of lysosome-mimicking vesicles to study the effect of abnormal accumulation of sphingosine on membrane properties. Sci Rep 2017. [PMID: 28638081 PMCID: PMC5479847 DOI: 10.1038/s41598-017-04125-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Synthetic systems are widely used to unveil the molecular mechanisms of complex cellular events. Artificial membranes are key examples of models employed to address lipid-lipid and lipid-protein interactions. In this work, we developed a new synthetic system that more closely resembles the lysosome – the lysosome-mimicking vesicles (LMVs) – displaying stable acid-to-neutral pH gradient across the membrane. To evaluate the advantages of this synthetic system, we assessed the distinct effects of sphingosine (Sph) accumulation in membrane structure and biophysical properties of standard liposomes (no pH gradient) and in LMVs with lipid composition tuned to mimic physiological- or NPC1-like lysosomes. Ternary 1-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC)/Sphingomyelin (SM)/Cholesterol (Chol) mixtures with, respectively, low and high Chol/SM levels were prepared. The effect of Sph on membrane permeability and biophysical properties was evaluated by fluorescence spectroscopy, electrophoretic and dynamic light scattering. The results showed that overall Sph has the ability to cause a shift in vesicle surface charge, increase membrane order and promote a rapid increase in membrane permeability. These effects are enhanced in NPC1- LMVs. The results suggest that lysosomal accumulation of these lipids, as observed under pathological conditions, might significantly affect lysosomal membrane structure and integrity, and therefore contribute to the impairment of cell function.
Collapse
|
23
|
Schonauer S, Körschen HG, Penno A, Rennhack A, Breiden B, Sandhoff K, Gutbrod K, Dörmann P, Raju DN, Haberkant P, Gerl MJ, Brügger B, Zigdon H, Vardi A, Futerman AH, Thiele C, Wachten D. Identification of a feedback loop involving β-glucosidase 2 and its product sphingosine sheds light on the molecular mechanisms in Gaucher disease. J Biol Chem 2017; 292:6177-6189. [PMID: 28258214 DOI: 10.1074/jbc.m116.762831] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 03/03/2017] [Indexed: 11/06/2022] Open
Abstract
The lysosomal acid β-glucosidase GBA1 and the non-lysosomal β-glucosidase GBA2 degrade glucosylceramide (GlcCer) to glucose and ceramide in different cellular compartments. Loss of GBA2 activity and the resulting accumulation of GlcCer results in male infertility, whereas mutations in the GBA1 gene and loss of GBA1 activity cause the lipid-storage disorder Gaucher disease. However, the role of GBA2 in Gaucher disease pathology and its relationship to GBA1 is not well understood. Here, we report a GBA1-dependent down-regulation of GBA2 activity in patients with Gaucher disease. Using an experimental approach combining cell biology, biochemistry, and mass spectrometry, we show that sphingosine, the cytotoxic metabolite accumulating in Gaucher cells through the action of GBA2, directly binds to GBA2 and inhibits its activity. We propose a negative feedback loop, in which sphingosine inhibits GBA2 activity in Gaucher cells, preventing further sphingosine accumulation and, thereby, cytotoxicity. Our findings add a new chapter to the understanding of the complex molecular mechanism underlying Gaucher disease and the regulation of β-glucosidase activity in general.
Collapse
Affiliation(s)
- Sophie Schonauer
- From the Minerva Max Planck Research Group, Molecular Physiology, and
| | - Heinz G Körschen
- the Department of Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), 53175 Bonn, Germany
| | - Anke Penno
- the Department of Cell Biology of Lipids, LIMES Institute, University of Bonn, Bonn, Germany
| | - Andreas Rennhack
- the Department of Molecular Sensory Systems, Center of Advanced European Studies and Research (caesar), 53175 Bonn, Germany
| | - Bernadette Breiden
- the LIMES Institute, c/o Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Konrad Sandhoff
- the LIMES Institute, c/o Kekulé-Institute, University of Bonn, 53115 Bonn, Germany
| | - Katharina Gutbrod
- the Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Peter Dörmann
- the Institute of Molecular Physiology and Biotechnology of Plants, University of Bonn, 53115 Bonn, Germany
| | - Diana N Raju
- From the Minerva Max Planck Research Group, Molecular Physiology, and
| | - Per Haberkant
- the Proteomic Core Facility, EMBL Heidelberg, 69117 Heidelberg, Germany
| | - Mathias J Gerl
- the Biochemie-Zentrum (BZH), Ruprecht-Karls-University Heidelberg, 69120 Heidelberg, Germany
| | - Britta Brügger
- the Biochemie-Zentrum (BZH), Ruprecht-Karls-University Heidelberg, 69120 Heidelberg, Germany
| | - Hila Zigdon
- the Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel, and
| | - Ayelet Vardi
- the Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel, and
| | - Anthony H Futerman
- the Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot 76100, Israel, and
| | - Christoph Thiele
- the Department of Cell Biology of Lipids, LIMES Institute, University of Bonn, Bonn, Germany
| | - Dagmar Wachten
- From the Minerva Max Planck Research Group, Molecular Physiology, and .,the Institute of Innate Immunity, University Hospital, University of Bonn, 53127 Bonn, Germany
| |
Collapse
|
24
|
Lima S, Milstien S, Spiegel S. Sphingosine and Sphingosine Kinase 1 Involvement in Endocytic Membrane Trafficking. J Biol Chem 2017; 292:3074-3088. [PMID: 28049734 DOI: 10.1074/jbc.m116.762377] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/22/2016] [Indexed: 12/22/2022] Open
Abstract
The balance between cholesterol and sphingolipids within the plasma membrane has long been implicated in endocytic membrane trafficking. However, in contrast to cholesterol functions, little is still known about the roles of sphingolipids and their metabolites. Perturbing the cholesterol/sphingomyelin balance was shown to induce narrow tubular plasma membrane invaginations enriched with sphingosine kinase 1 (SphK1), the enzyme that converts the bioactive sphingolipid metabolite sphingosine to sphingosine-1-phosphate, and suggested a role for sphingosine phosphorylation in endocytic membrane trafficking. Here we show that sphingosine and sphingosine-like SphK1 inhibitors induced rapid and massive formation of vesicles in diverse cell types that accumulated as dilated late endosomes. However, much smaller vesicles were formed in SphK1-deficient cells. Moreover, inhibition or deletion of SphK1 prolonged the lifetime of sphingosine-induced vesicles. Perturbing the plasma membrane cholesterol/sphingomyelin balance abrogated vesicle formation. This massive endosomal influx was accompanied by dramatic recruitment of the intracellular SphK1 and Bin/Amphiphysin/Rvs domain-containing proteins endophilin-A2 and endophilin-B1 to enlarged endosomes and formation of highly dynamic filamentous networks containing endophilin-B1 and SphK1. Together, our results highlight the importance of sphingosine and its conversion to sphingosine-1-phosphate by SphK1 in endocytic membrane trafficking.
Collapse
Affiliation(s)
- Santiago Lima
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298.
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology and Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298.
| |
Collapse
|
25
|
On the possible structural role of single chain sphingolipids Sphingosine and Sphingosine 1-phosphate in the amyloid-β peptide interactions with membranes. Consequences for Alzheimer’s disease development. Colloids Surf A Physicochem Eng Asp 2016. [DOI: 10.1016/j.colsurfa.2016.04.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
26
|
Zhou K, Blom T. Trafficking and Functions of Bioactive Sphingolipids: Lessons from Cells and Model Membranes. Lipid Insights 2015; 8:11-20. [PMID: 26715852 PMCID: PMC4685176 DOI: 10.4137/lpi.s31615] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/08/2015] [Accepted: 11/10/2015] [Indexed: 12/15/2022] Open
Abstract
Ceramide and sphingosine and their phosphorylated counterparts are recognized as "bioactive sphingolipids" and modulate membrane integrity, the activity of enzymes, or act as ligands of G protein-coupled receptors. The subcellular distribution of the bioactive sphingolipids is central to their function as the same lipid can mediate diametrically opposite effects depending on its location. To ensure that these lipids are present in the right amount and in the appropriate organelles, cells employ selective lipid transport and compartmentalize sphingolipid-metabolizing enzymes to characteristic subcellular sites. Our knowledge of key mechanisms involved in sphingolipid signaling and trafficking has increased substantially in the past decades-thanks to advances in biochemical and cell biological methods. In this review, we focus on the bioactive sphingolipids and discuss how the combination of studies in cells and in model membranes have contributed to our understanding of how they behave and function in living organisms.
Collapse
Affiliation(s)
- Kecheng Zhou
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Tomas Blom
- Department of Anatomy, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
27
|
Fröhlich F, Petit C, Kory N, Christiano R, Hannibal-Bach HK, Graham M, Liu X, Ejsing CS, Farese RV, Walther TC. The GARP complex is required for cellular sphingolipid homeostasis. eLife 2015; 4. [PMID: 26357016 PMCID: PMC4600884 DOI: 10.7554/elife.08712] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/09/2015] [Indexed: 12/11/2022] Open
Abstract
Sphingolipids are abundant membrane components and important signaling molecules in eukaryotic cells. Their levels and localization are tightly regulated. However, the mechanisms underlying this regulation remain largely unknown. In this study, we identify the Golgi-associated retrograde protein (GARP) complex, which functions in endosome-to-Golgi retrograde vesicular transport, as a critical player in sphingolipid homeostasis. GARP deficiency leads to accumulation of sphingolipid synthesis intermediates, changes in sterol distribution, and lysosomal dysfunction. A GARP complex mutation analogous to a VPS53 allele causing progressive cerebello-cerebral atrophy type 2 (PCCA2) in humans exhibits similar, albeit weaker, phenotypes in yeast, providing mechanistic insights into disease pathogenesis. Inhibition of the first step of de novo sphingolipid synthesis is sufficient to mitigate many of the phenotypes of GARP-deficient yeast or mammalian cells. Together, these data show that GARP is essential for cellular sphingolipid homeostasis and suggest a therapeutic strategy for the treatment of PCCA2. DOI:http://dx.doi.org/10.7554/eLife.08712.001 Every cell is enveloped by a membrane that forms a barrier between the cell and its environment. This membrane contains fat molecules called ‘sphingolipids’, which help to maintain the structure of the membrane and enable it to work correctly. These molecules are also used as signals to send information around the interior of the cell and are required for the cell to grow and divide normally. The levels of sphingolipids in the membrane have to be tightly controlled because any imbalance can cause stress to the cell and can lead to serious diseases. Sphingolipids are made inside the cell and are then sent to a compartment called the Golgi before being delivered to the membrane. To regulate the amount of sphingolipids in the membrane, these molecules are routinely returned to the interior of the cell in small structures called endosomes. From here, they can either be broken down or recycled back to the membrane via the Golgi. A group of proteins known as the Golgi-associated retrograde protein complex (or GARP) is involved in the movement of endosomes from the membrane to the Golgi. People that have a mutation in the gene that encodes GARP suffer from a severe neurodegenerative disease known as ‘progressive cerebello-cerebral atrophy type 2’ (PCCA2) in which brain cells die prematurely. Researchers have assumed that the most important role of GARP is to sort proteins, and that the missorting of proteins leads to PCCA2. Here, Frohlich et al. used a combination of genetic analysis and biochemical techniques to study GARP in yeast cells. The experiments show that GARP is critical for sphingolipid recycling, and that a lack of GARP leads to more sphingolipids being degraded, which results in a build-up of toxic molecules. Frohlich et al. generated yeast cells that have the same mutations in the gene that encodes GARP as those in human patients with PCCA2. These cells grew much slower than normal yeast and were less able to transport sphingolipids from the endosome to the Golgi. Like the yeast cells, human cells in which the gene that encodes GARP was less active also accumulated toxic molecules. Together, these findings suggest that a build-up of toxic fat molecules may be responsible for the symptoms observed in PCCA2 patients. A future challenge is to find out whether this also applies to patients with Alzheimer's disease and other conditions that also affect endosomes. DOI:http://dx.doi.org/10.7554/eLife.08712.002
Collapse
Affiliation(s)
- Florian Fröhlich
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Constance Petit
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Nora Kory
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Romain Christiano
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States
| | - Hans-Kristian Hannibal-Bach
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Morven Graham
- Center for Cellular and Molecular Imaging, Yale School of Medicine, New Haven, United States
| | - Xinran Liu
- Center for Cellular and Molecular Imaging, Yale School of Medicine, New Haven, United States.,Department of Cell Biology, Yale School of Medicine, New Haven, United States
| | - Christer S Ejsing
- Department of Biochemistry and Molecular Biology, VILLUM Center for Bioanalytical Sciences, University of Southern Denmark, Odense, Denmark
| | - Robert V Farese
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States.,Broad Institute, Cambridge, United States
| | - Tobias C Walther
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Cell Biology, Harvard Medical School, Boston, United States.,Broad Institute, Cambridge, United States.,Howard Hughes Medical Institute, Harvard T.H. Chan School of Public Health, Boston, United States
| |
Collapse
|
28
|
Carreira AC, Ventura AE, Varela AR, Silva LC. Tackling the biophysical properties of sphingolipids to decipher their biological roles. Biol Chem 2015; 396:597-609. [DOI: 10.1515/hsz-2014-0283] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 01/01/2015] [Indexed: 11/15/2022]
Abstract
Abstract
From the most simple sphingoid bases to their complex glycosylated derivatives, several sphingolipid species were shown to have a role in fundamental cellular events and/or disease. Increasing evidence places lipid-lipid interactions and membrane structural alterations as central mechanisms underlying the action of these lipids. Understanding how these molecules exert their biological roles by studying their impact in the physical properties and organization of membranes is currently one of the main challenges in sphingolipid research. Herein, we review the progress in the state-of-the-art on the biophysical properties of sphingolipid-containing membranes, focusing on sphingosine, ceramides, and glycosphingolipids.
Collapse
|
29
|
Orchestration of membrane receptor signaling by membrane lipids. Biochimie 2015; 113:111-24. [DOI: 10.1016/j.biochi.2015.04.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 04/05/2015] [Indexed: 12/20/2022]
|
30
|
Uchida Y, Kim YI, Park K. Signaling roles of ceramide and its metabolites in cutaneous antimicrobial defense. DERMATOL SIN 2015. [DOI: 10.1016/j.dsi.2015.04.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
31
|
Jiménez-Rojo N, Sot J, Viguera AR, Collado MI, Torrecillas A, Gómez-Fernández JC, Goñi FM, Alonso A. Membrane permeabilization induced by sphingosine: effect of negatively charged lipids. Biophys J 2015; 106:2577-84. [PMID: 24940775 DOI: 10.1016/j.bpj.2014.04.038] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/14/2014] [Accepted: 04/23/2014] [Indexed: 10/25/2022] Open
Abstract
Sphingosine [(2S, 3R, 4E)-2-amino-4-octadecen-1, 3-diol] is the most common sphingoid long chain base in sphingolipids. It is the precursor of important cell signaling molecules, such as ceramides. In the last decade it has been shown to act itself as a potent metabolic signaling molecule, by activating a number of protein kinases. Moreover, sphingosine has been found to permeabilize phospholipid bilayers, giving rise to vesicle leakage. The present contribution intends to analyze the mechanism by which this bioactive lipid induces vesicle contents release, and the effect of negatively charged bilayers in the release process. Fluorescence lifetime measurements and confocal fluorescence microscopy have been applied to observe the mechanism of sphingosine efflux from large and giant unilamellar vesicles; a graded-release efflux has been detected. Additionally, stopped-flow measurements have shown that the rate of vesicle permeabilization increases with sphingosine concentration. Because at the physiological pH sphingosine has a net positive charge, its interaction with negatively charged phospholipids (e.g., bilayers containing phosphatidic acid together with sphingomyelins, phosphatidylethanolamine, and cholesterol) gives rise to a release of vesicular contents, faster than with electrically neutral bilayers. Furthermore, phosphorous 31-NMR and x-ray data show the capacity of sphingosine to facilitate the formation of nonbilayer (cubic phase) intermediates in negatively charged membranes. The data might explain the pathogenesis of Niemann-Pick type C1 disease.
Collapse
Affiliation(s)
- Noemi Jiménez-Rojo
- Unidad de Biofísica (CSIC, UPV/EHU) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain
| | - Jesús Sot
- Unidad de Biofísica (CSIC, UPV/EHU) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain
| | - Ana R Viguera
- Unidad de Biofísica (CSIC, UPV/EHU) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain
| | - M Isabel Collado
- Servicio General de Resonancia Magnética Nuclear, Universidad del País Vasco, Bilbao, Spain
| | - Alejandro Torrecillas
- Sección de Biología Molecular, Servicio de Apoyo a la Investigación, Universidad de Murcia
| | - J C Gómez-Fernández
- Departamento de Bioquímica y Biología Molecular-A, Facultad de Veterinaria, Universidad de Murcia, Regional Campus of International Excellence "Campus Mare Nostrum", Murcia, Spain
| | - Félix M Goñi
- Unidad de Biofísica (CSIC, UPV/EHU) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain
| | - Alicia Alonso
- Unidad de Biofísica (CSIC, UPV/EHU) and Departamento de Bioquímica, Universidad del País Vasco, Bilbao, Spain.
| |
Collapse
|
32
|
Mechanisms of action of substituted β-amino alkanols on Leishmania donovani. Antimicrob Agents Chemother 2014; 59:1211-8. [PMID: 25487805 DOI: 10.1128/aac.04003-14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Leishmaniasis is the protozoan disease second in importance for human health, superseded only by malaria; however, the options for chemotherapeutic treatment are increasingly limited due to drug resistance and toxicity. Under this perspective, a quest for new chemical compounds is urgently needed. An N-substituted 2-aminoalkan-1-ol scaffold has been shown to be a versatile scaffold for antiparasitic activity. Knowledge about its mechanism of action is still rather limited. In this work, we endeavored to define the leishmanicidal profile of such β-amino alkanol derivatives using a set of 15 N-mono- and disubstituted surrogates, tested on Leishmania donovani promastigotes and intracellular amastigotes. The best compound (compound 5), 2-ethylaminododecan-1-ol, had a 50% effective concentration (EC50) of 0.3 μM and a selectivity index of 72 for infected THP-1 cells and was selected for further elucidation of its leishmanicidal mechanism. It induced fast depletion of intracellular ATP content in promastigotes in the absence of vital dye intracellular entry, ruling out plasma membrane permeabilization as its origin. Confocal and transmission electron microscopy analyses showed that compound 5 induced severe mitochondrial swelling and vesiculation. Polarographic analysis using an oxygen electrode demonstrated that complex II of the respiratory chain (succinate reductase) was strongly inhibited by compound 5, identifying this complex as one of the primary targets. Furthermore, for other β-amino alkanols whose structures differed subtly from that of compound 5, plasma membrane permeabilization or interference with membrane traffic was also observed. In all, N-substituted β-amino alkanols were shown as appealing leishmanicidal candidates deserving further exploration.
Collapse
|
33
|
Lysosomotropic agents: impact on lysosomal membrane permeabilization and cell death. Biochem Soc Trans 2014; 42:1460-4. [DOI: 10.1042/bst20140145] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Lysosomes are acidic organelles essential for degradation, signalling and cell homoeostasis. In addition, they play a key role in cell death. Permeabilization of the lysosomal membrane and release of hydrolytic enzymes to the cytosol accompanies apoptosis signalling in several systems. The regulatory mechanism of lysosomal stability is, however, poorly understood. Lipophilic or amphiphilic compounds with a basic moiety will become protonated and trapped within lysosomes, and such lysosomotropic behaviour is also found in many pharmacological drugs. The natural sphingolipid sphingosine exhibits lysosomotropic detergent ability and is an endogenous candidate for controlling lysosomal membrane permeabilization. The lysosomotropic properties of certain detergents might be of use in lysosome-targeting anticancer drugs and drug delivery system in the future. The present review summarizes the current knowledge on the targeting and permeabilizing properties of lysosomotropic detergents from a cellular and physicochemical perspective.
Collapse
|
34
|
Biophysical properties of sphingosine, ceramides and other simple sphingolipids. Biochem Soc Trans 2014; 42:1401-8. [DOI: 10.1042/bst20140159] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Some of the simplest sphingolipids, namely sphingosine, ceramide and their phosphorylated compounds [sphingosine 1-phosphate (Sph-1-P) and ceramide 1-phosphate (Cer-1-P)], are potent metabolic regulators. Each of these lipids modifies in marked and specific ways the physical properties of the cell membranes, in what can be the basis for some of their physiological actions. The present paper is an overview of the mechanisms by which these sphingolipid signals, sphingosine and ceramide, in particular, are able to modify the properties of cell membranes.
Collapse
|
35
|
Jiménez-Rojo N, Viguera AR, Collado MI, Sims KH, Constance C, Hill K, Shaw WA, Goñi FM, Alonso A. Sphingosine induces the aggregation of imine-containing peroxidized vesicles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2071-7. [DOI: 10.1016/j.bbamem.2014.04.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 04/25/2014] [Accepted: 04/28/2014] [Indexed: 01/09/2023]
|
36
|
Repnik U, Hafner Česen M, Turk B. Lysosomal membrane permeabilization in cell death: concepts and challenges. Mitochondrion 2014; 19 Pt A:49-57. [PMID: 24984038 DOI: 10.1016/j.mito.2014.06.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/19/2014] [Accepted: 06/24/2014] [Indexed: 01/06/2023]
Abstract
Late endocytic compartments include late endosomes, lysosomes and hybrid organelles. In the acidic lumen, cargo material derived from endocytosed and phagocytosed extracellular material and autophagy-derived intracellular material is degraded. In the event of lysosomal membrane permeabilization (LMP), the function of endo/lysosomal compartment is affected and the luminal contents are released into the cytosol to various extents. LMP can be a result of osmotic lysis or direct membranolytic activity of the compounds that accumulate in the lumen of endo/lysosomes. In addition to several synthetic compounds, such as dipeptide methyl esters and lysosomotropic detergents, endogenous agents that can cause LMP include ROS and lipid metabolites such as sphingosine and phosphatidic acid. Depending on the cell type and the dose, LMP can initiate the lysosomal apoptotic pathway, pyroptosis or necrosis. LMP can also amplify cell death signaling that was initiated outside the endocytic compartment, and hamper cell recovery via autophagy. However, mechanisms that connect LMP with cell death signaling are poorly understood, with the exception of the proteolytic activation of Bid by aspartic cathepsin D and cysteine cathepsins. Determination of LMP in a cell model system is methodologically challenging. Even more difficult is to prove that LMP is the primary event leading to cell death. Nevertheless, LMP may prove to be a valuable approach in therapy, either as a trigger of cell death or as a mechanism of therapeutic drug release in the case of delivery systems that target the endocytic pathway.
Collapse
Affiliation(s)
- Urška Repnik
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; Department of Biosciences, University of Oslo, Blindernveien 31, 0371 Oslo, Norway
| | - Maruša Hafner Česen
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Department of Biochemistry and Molecular and Structural Biology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKeBiP, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Aškerčeva 5, 1000 Ljubljana, Slovenia.
| |
Collapse
|
37
|
Zupancic E, Carreira AC, de Almeida RFM, Silva LC. Biophysical Implications of Sphingosine Accumulation in Membrane Properties at Neutral and Acidic pH. J Phys Chem B 2014; 118:4858-66. [DOI: 10.1021/jp501167f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Eva Zupancic
- iMed.UL
- Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia da Universidade de Lisboa, 1649-003 Lisboa, Portugal
| | - Ana C. Carreira
- iMed.UL
- Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia da Universidade de Lisboa, 1649-003 Lisboa, Portugal
- Centro
de Química e Bioquímica, DQB, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Rodrigo F. M. de Almeida
- Centro
de Química e Bioquímica, DQB, Faculdade de Ciências da Universidade de Lisboa, Campo Grande, 1749-016 Lisboa, Portugal
| | - Liana C. Silva
- iMed.UL
- Research Institute for Medicines and Pharmaceutical Sciences, Faculdade de Farmácia da Universidade de Lisboa, 1649-003 Lisboa, Portugal
| |
Collapse
|
38
|
Swain J, Borkar SR, Aidhen IS, Mishra AK. A molecular level understanding of interaction between FTY720 (Fingolimod hydrochloride) and DMPC multilamellar vesicles. RSC Adv 2014. [DOI: 10.1039/c4ra02404d] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
This work focuses on the molecular level understanding of interaction between FTY720 (Fingolimod hydrochloride) and dimyristoylphosphatidylcholine (DMPC) multilamellar vesicles (MLVs) as a drug molecule carrier by investigating the structural changes, solubilisation effect and thermotropic phase behaviour.
Collapse
Affiliation(s)
- Jitendriya Swain
- Department of Chemistry
- Indian Institute of Technology Madras
- Chennai 600 036, India
| | - Santosh R. Borkar
- Department of Chemistry
- Indian Institute of Technology Madras
- Chennai 600 036, India
| | | | - Ashok Kumar Mishra
- Department of Chemistry
- Indian Institute of Technology Madras
- Chennai 600 036, India
| |
Collapse
|
39
|
de Melo NR, Abdrahman A, Greig C, Mukherjee K, Thornton C, Ratcliffe NA, Vilcinskas A, Butt TM. Myriocin significantly increases the mortality of a non-mammalian model host during Candida pathogenesis. PLoS One 2013; 8:e78905. [PMID: 24260135 PMCID: PMC3829820 DOI: 10.1371/journal.pone.0078905] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 09/17/2013] [Indexed: 12/14/2022] Open
Abstract
Candida albicans is a major human pathogen whose treatment is challenging due to antifungal drug toxicity, drug resistance and paucity of antifungal agents available. Myrocin (MYR) inhibits sphingosine synthesis, a precursor of sphingolipids, an important cell membrane and signaling molecule component. MYR also has dual immune suppressive and antifungal properties, potentially modulating mammalian immunity and simultaneously reducing fungal infection risk. Wax moth (Galleria mellonella) larvae, alternatives to mice, were used to establish if MYR suppressed insect immunity and increased survival of C. albicans-infected insects. MYR effects were studied in vivo and in vitro, and compared alone and combined with those of approved antifungal drugs, fluconazole (FLC) and amphotericin B (AMPH). Insect immune defenses failed to inhibit C. albicans with high mortalities. In insects pretreated with the drug followed by C. albicans inoculation, MYR+C. albicans significantly increased mortality to 93% from 67% with C. albicans alone 48 h post-infection whilst AMPH+C. albicans and FLC+C. albicans only showed 26% and 0% mortalities, respectively. MYR combinations with other antifungal drugs in vivo also enhanced larval mortalities, contrasting the synergistic antifungal effect of the MYR+AMPH combination in vitro. MYR treatment influenced immunity and stress management gene expression during C. albicans pathogenesis, modulating transcripts putatively associated with signal transduction/regulation of cytokines, I-kappaB kinase/NF-kappaB cascade, G-protein coupled receptor and inflammation. In contrast, all stress management gene expression was down-regulated in FLC and AMPH pretreated C. albicans -infected insects. Results are discussed with their implications for clinical use of MYR to treat sphingolipid-associated disorders.
Collapse
Affiliation(s)
| | - Ahmed Abdrahman
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, United Kingdom
| | - Carolyn Greig
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, United Kingdom
| | - Krishnendu Mukherjee
- Institut für Phytopathologie und Angewandte Zoologie, Abteilung Angewandte Entomologie, Gieβen, Germany
| | - Catherine Thornton
- College of Medicine, Swansea University, Singleton Park, Swansea, United Kingdom
| | - Norman A. Ratcliffe
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, United Kingdom
- Department of Biological Sciences, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Andreas Vilcinskas
- Institut für Phytopathologie und Angewandte Zoologie, Abteilung Angewandte Entomologie, Gieβen, Germany
| | - Tariq M. Butt
- Department of Biosciences, College of Science, Swansea University, Singleton Park, Swansea, United Kingdom
- * E-mail:
| |
Collapse
|
40
|
Zhang O, Hsu FF, Xu W, Pawlowic M, Zhang K. Sphingosine kinase A is a pleiotropic and essential enzyme for Leishmania survival and virulence. Mol Microbiol 2013; 90:489-501. [PMID: 23980754 PMCID: PMC3938578 DOI: 10.1111/mmi.12378] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2013] [Indexed: 01/05/2023]
Abstract
Sphingosine kinase is a key enzyme in sphingolipid metabolism, catalysing the conversion of sphingosine or dihydrosphingosine into sphingosine-1-phosphate or dihydrosphingosine-1-phosphate respectively. In mammals, sphingosine-1-phosphate is a powerful signalling molecule regulating cell growth, differentiation, apoptosis and immunity. Functions of sphingosine kinase or sphingosine-1-phosphate in pathogenic protozoans are virtually unknown. While most organisms possess two closely related sphingosine kinases, only one sphingosine kinase homologue (SKa) can be identified in Leishmania, which are vector-borne protozoan parasites responsible for leishmaniasis. Leishmania SKa is a large, cytoplasmic enzyme capable of phosphorylating both sphingosine and dihydrosphingosine. Remarkably, deletion of SKa leads to catastrophic defects in both the insect stage and mammalian stage of Leishmania parasites. Genetic and biochemical analyses demonstrate that proper expression of SKa is essential for Leishmania parasites to remove toxic metabolites, to survive stressful conditions, and to cause disease in mice. Therefore, SKa is a pleiotropic enzyme with vital roles throughout the life cycle of Leishmania. The essentiality of SKa and its apparent divergence from mammalian counterparts suggests that this enzyme can be selectively targeted to reduce Leishmania infection.
Collapse
Affiliation(s)
- Ou Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Fong-Fu Hsu
- Department of Internal Medicine, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO 63110, USA
| | - Wei Xu
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Mattie Pawlowic
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| | - Kai Zhang
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
| |
Collapse
|
41
|
D'Auria L, Deleu M, Dufour S, Mingeot-Leclercq MP, Tyteca D. Surfactins modulate the lateral organization of fluorescent membrane polar lipids: A new tool to study drug:membrane interaction and assessment of the role of cholesterol and drug acyl chain length. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2064-73. [DOI: 10.1016/j.bbamem.2013.05.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/16/2013] [Accepted: 05/08/2013] [Indexed: 12/11/2022]
|
42
|
Antioxidation Behavior of Milkweed Oil 4-Hydroxy-3-methoxycinnamate Esters in Phospholipid Bilayers. J AM OIL CHEM SOC 2013. [DOI: 10.1007/s11746-013-2323-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
Altered sphingoid base profiles predict compromised membrane structure and permeability in atopic dermatitis. J Dermatol Sci 2013; 72:296-303. [PMID: 24070864 DOI: 10.1016/j.jdermsci.2013.08.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 07/24/2013] [Accepted: 08/01/2013] [Indexed: 01/16/2023]
Abstract
BACKGROUND Ceramide hydrolysis by ceramidase in the stratum corneum (SC) yields both sphingoid bases and free fatty acids (FFA). While FFA are key constituents of the lamellar bilayers that mediate the epidermal permeability barrier, whether sphingoid bases influence permeability barrier homeostasis remains unknown. Pertinently, alterations of lipid profile, including ceramide and ceramidase activities occur in atopic dermatitis (AD). OBJECT We investigated alterations in sphingoid base levels and/or profiles (sphingosine to sphinganine ratio) in the SC of normal vs. AD mice, a model that faithfully replicates human AD, and then whether altered sphingoid base levels and/or profiles influence(s) membrane stability and/or structures. METHODS Unilamellar vesicles (LV), incorporating the three major SC lipids (ceramides/FFA/cholesterol) and different ratios of sphingosine/sphinganine, encapsulating carboxyfluorescein, were used as the model of SC lipids. Membrane stability was measured as release of carboxyfluorescein. Thermal analysis of LV was conducted by differential scanning calorimetry (DSC). RESULTS LV containing AD levels of sphingosine/sphinganine (AD-LV) displayed altered membrane permeability vs. normal-LV. DSC analyses revealed decreases in orthorhombic structures that form tightly packed lamellar structures in AD-LV. CONCLUSION Sphingoid base composition influences lamellar membrane architecture in SC, suggesting that altered sphingoid base profiles could contribute to the barrier abnormality in AD.
Collapse
|
44
|
Lee YM, Lim C, Lee HS, Shin YK, Shin KO, Lee YM, Kim S. Synthesis and Biological Evaluation of a Polyyne-Containing Sphingoid Base Probe as a Chemical Tool. Bioconjug Chem 2013; 24:1324-31. [DOI: 10.1021/bc300684q] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Yun Mi Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu,
Seoul 151-742, Korea
| | - Chaemin Lim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu,
Seoul 151-742, Korea
| | - Hun Seok Lee
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu,
Seoul 151-742, Korea
| | - Young Kee Shin
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu,
Seoul 151-742, Korea
| | - Kyong-Oh Shin
- College
of Pharmacy and MRC, Chungbuk National University, Cheongju 361-763, Korea
| | - Yong-Moon Lee
- College
of Pharmacy and MRC, Chungbuk National University, Cheongju 361-763, Korea
| | - Sanghee Kim
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu,
Seoul 151-742, Korea
| |
Collapse
|
45
|
Abstract
There is growing evidence that cell membranes can contain domains with different lipid and protein compositions and with different physical properties. Furthermore, it is increasingly appreciated that sphingolipids play a crucial role in the formation and properties of ordered lipid domains (rafts) in cell membranes. This review describes recent advances in our understanding of ordered membrane domains in both cells and model membranes. In addition, how the structure of sphingolipids influences their ability to participate in the formation of ordered domains, as well as how sphingolipid structure alters ordered domain properties, is described. The diversity of sphingolipid structure is likely to play an important role in modulating the biologically relevant properties of "rafts" in cell membranes.
Collapse
|
46
|
Janas T, Janas T, Yarus M. Human tRNA(Sec) associates with HeLa membranes, cell lipid liposomes, and synthetic lipid bilayers. RNA (NEW YORK, N.Y.) 2012; 18:2260-2268. [PMID: 23097422 PMCID: PMC3504676 DOI: 10.1261/rna.035352.112] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Accepted: 09/14/2012] [Indexed: 06/01/2023]
Abstract
We have shown previously that simple RNA structures bind pure phospholipid liposomes. However, binding of bona fide cellular RNAs under physiological ionic conditions is shown here for the first time. Human tRNA(Sec) contains a hydrophobic anticodon-loop modification: N⁶-isopentenyladenosine (i⁶A) adjacent to its anticodon. Using a highly specific double-probe hybridization assay, we show mature human tRNA(Sec) specifically retained in HeLa intermediate-density membranes. Further, isolated human tRNA(Sec) rebinds to liposomes from isolated HeLa membrane lipids, to a much greater extent than an unmodified tRNA(Sec) transcript. To better define this affinity, experiments with pure lipids show that liposomes forming rafts or including positively charged sphingosine, or particularly both together, exhibit increased tRNA(Sec) binding. Thus tRNA(Sec) residence on membranes is determined by several factors, such as hydrophobic modification (likely isopentenylation of tRNA(Sec)), lipid structure (particularly lipid rafts), or sphingosine at a physiological concentration in rafted membranes. From prior work, RNA structure and ionic conditions also appear important. tRNA(Sec) dissociation from HeLa liposomes implies a mean membrane residence of 7.6 min at 24°C (t(1/2) = 5.3 min). Clearly RNA with a 5-carbon hydrophobic modification binds HeLa membranes, probably favoring raft domains containing specific lipids, for times sufficient to alter biological fates.
Collapse
Affiliation(s)
- Teresa Janas
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309, USA
- Department of Biotechnology and Molecular Biology, University of Opole, 45-032 Opole, Poland
| | - Tadeusz Janas
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309, USA
- Department of Biotechnology and Molecular Biology, University of Opole, 45-032 Opole, Poland
| | - Michael Yarus
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, Colorado 80309, USA
| |
Collapse
|
47
|
Robciuc A, Hyötyläinen T, Jauhiainen M, Holopainen JM. Hyperosmolarity-induced lipid droplet formation depends on ceramide production by neutral sphingomyelinase 2. J Lipid Res 2012; 53:2286-95. [PMID: 22899568 DOI: 10.1194/jlr.m026732] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Hyperosmolarity (HO) imposes a remarkable stress on membranes, especially in tissues in direct contact with the external environment. Our efforts were focused on revealing stress-induced lipid changes that precede the inflammatory cytokine response in human corneal epithelial cells exposed to increasing osmolarity. We used a lipidomic analysis that detected significant and systematic changes in the lipid profile, highly correlated with sodium concentrations in the medium. Ceramides and triglycerides (TGs) were the most-responsive lipid classes, with gradual increases of up to 2- and 3-fold, respectively, when compared with control. The source of ceramide proved to be sphingomyelin hydrolysis, and neutral sphingomyelinase 2 (NSM2) activity showed a 2-fold increase 1 h after HO stress, whereas transcription increased 3-fold. Both TG accumulation and IL-8 secretion were shown to be dependent on ceramide production by specific knock-down of NSM2. In HCE cells, diglyceride acyltransferase 1 was responsible for the TG synthesis, but the enzyme activity had no effect on cytokine secretion. Hence, NSM2 plays a key role in the cellular response to hyperosmolar stress, and its activity regulates both cytokine secretion and lipid droplet formation.
Collapse
Affiliation(s)
- Alexandra Robciuc
- Helsinki Eye Lab, Department of Ophthalmology, Haartmaninkatu 4 C, University of Helsinki, 00290 Helsinki, Finland
| | | | | | | |
Collapse
|
48
|
Goñi FM, Montes LR, Alonso A. Phospholipases C and sphingomyelinases: Lipids as substrates and modulators of enzyme activity. Prog Lipid Res 2012; 51:238-66. [DOI: 10.1016/j.plipres.2012.03.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 03/23/2012] [Accepted: 03/26/2012] [Indexed: 11/30/2022]
|
49
|
Qadri SM, Bauer J, Zelenak C, Mahmud H, Kucherenko Y, Lee SH, Ferlinz K, Lang F. Sphingosine but not sphingosine-1-phosphate stimulates suicidal erythrocyte death. Cell Physiol Biochem 2011; 28:339-46. [PMID: 21865742 DOI: 10.1159/000331750] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2011] [Indexed: 12/14/2022] Open
Abstract
Sphingosine kinase 1 phosphorylates sphingosine, which is converted to ceramide by ceramide synthetase. Ceramide triggers eryptosis, the suicidal erythrocyte death characterized by cell shrinkage and phosphatidylserine (PS) exposure at the erythrocyte surface. Erythrocytes lack sphingosine phosphate-degrading enzymes and thus store large quantities of sphingosine phosphate. The present study explored the influence of sphingosine and sphingosine phosphate on eryptosis. [Ca(2+)](i), was estimated from Fluo3 fluorescence, cell volume from forward scatter and PS exposure from annexin V-binding in FACS analysis. Sphingosine (0.1 - 10 μM) but not sphingosine-1- phosphate (0.1 - 10 μM) increased [Ca(2+)](i), decreased cell volume and increased PS-exposure. The observations disclose sphingosine, but not sphingosine-1-phosphate, as a strong inducer of eryptosis.
Collapse
Affiliation(s)
- Syed M Qadri
- Department of Physiology, University of Tübingen, Tübingen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Georgieva R, Koumanov K, Momchilova A, Tessier C, Staneva G. Effect of sphingosine on domain morphology in giant vesicles. J Colloid Interface Sci 2010; 350:502-10. [DOI: 10.1016/j.jcis.2010.07.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2010] [Revised: 07/09/2010] [Accepted: 07/10/2010] [Indexed: 02/04/2023]
|