1
|
Dai J, Feng Y, Liao Y, Tan L, Sun Y, Song C, Qiu X, Ding C. Virus infection and sphingolipid metabolism. Antiviral Res 2024; 228:105942. [PMID: 38908521 DOI: 10.1016/j.antiviral.2024.105942] [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: 04/27/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Cellular sphingolipids have vital roles in human virus replication and spread as they are exploited by viruses for cell entry, membrane fusion, genome replication, assembly, budding, and propagation. Intracellular sphingolipid biosynthesis triggers conformational changes in viral receptors and facilitates endosomal escape. However, our current understanding of how sphingolipids precisely regulate viral replication is limited, and further research is required to comprehensively understand the relationships between viral replication and endogenous sphingolipid species. Emerging evidence now suggests that targeting and manipulating sphingolipid metabolism enzymes in host cells is a promising strategy to effectively combat viral infections. Additionally, serum sphingolipid species and concentrations could function as potential serum biomarkers to help monitor viral infection status in different patients. In this work, we comprehensively review the literature to clarify how viruses exploit host sphingolipid metabolism to accommodate viral replication and disrupt host innate immune responses. We also provide valuable insights on the development and use of antiviral drugs in this area.
Collapse
Affiliation(s)
- Jun Dai
- Experimental Animal Center, Zunyi Medical University, Zunyi, 563099, China
| | - Yiyi Feng
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Laboratory of Veterinary Microbiology and Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning, 530004, Guangxi China
| | - Ying Liao
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Lei Tan
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Yingjie Sun
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Cuiping Song
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China
| | - Xusheng Qiu
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China.
| | - Chan Ding
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai 200241, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
2
|
Chen S, Liang J, Chen D, Huang Q, Sun K, Zhong Y, Lin B, Kong J, Sun J, Gong C, Wang J, Gao Y, Zhang Q, Sun H. Cerebrospinal fluid metabolomic and proteomic characterization of neurologic post-acute sequelae of SARS-CoV-2 infection. Brain Behav Immun 2024; 115:209-222. [PMID: 37858739 DOI: 10.1016/j.bbi.2023.10.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 10/08/2023] [Accepted: 10/14/2023] [Indexed: 10/21/2023] Open
Abstract
The mechanism by which SARS-CoV-2 causes neurological post-acute sequelae of SARS-CoV-2 (neuro-PASC) remains unclear. Herein, we conducted proteomic and metabolomic analyses of cerebrospinal fluid (CSF) samples from 21 neuro-PASC patients, 45 healthy volunteers, and 26 inflammatory neurological diseases patients. Our data showed 69 differentially expressed metabolites and six differentially expressed proteins between neuro-PASC patients and healthy individuals. Elevated sphinganine and ST1A1, sphingolipid metabolism disorder, and attenuated inflammatory responses may contribute to the occurrence of neuro-PASC, whereas decreased levels of 7,8-dihydropterin and activation of steroid hormone biosynthesis may play a role in the repair process. Additionally, a biomarker cohort consisting of sphinganine, 7,8-dihydroneopterin, and ST1A1 was preliminarily demonstrated to have high value in diagnosing neuro-PASC. In summary, our study represents the first attempt to integrate the diagnostic benefits of CSF with the methodological advantages of multi-omics, thereby offering valuable insights into the pathogenesis of neuro-PASC and facilitating the work of neuroscientists in disclosing different neurological dimensions associated with COVID-19.
Collapse
Affiliation(s)
- Shilan Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianhao Liang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Dingqiang Chen
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Qiyuan Huang
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Kaijian Sun
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuxia Zhong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Baojia Lin
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jingjing Kong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jiaduo Sun
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou 510180, China
| | - Chengfang Gong
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jun Wang
- Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Ya Gao
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Qingguo Zhang
- Department of Anesthesiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China.
| | - Haitao Sun
- Clinical Biobank Center, Microbiome Medicine Center, Department of Laboratory Medicine, Guangdong Provincial Clinical Research Center for Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Neurosurgery Center, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Key Laboratory of Mental Health of the Ministry of Education, Guangdong-Hong Kong-Macao Greater Bay Area Center for Brain Science and Brain-Inspired Intelligence, Southern Medical University, Guangzhou, China.
| |
Collapse
|
3
|
Ali H, Naseem A, Siddiqui ZI. SARS-CoV-2 Syncytium under the Radar: Molecular Insights of the Spike-Induced Syncytia and Potential Strategies to Limit SARS-CoV-2 Replication. J Clin Med 2023; 12:6079. [PMID: 37763019 PMCID: PMC10531702 DOI: 10.3390/jcm12186079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
SARS-CoV-2 infection induces non-physiological syncytia when its spike fusogenic protein on the surface of the host cells interacts with the ACE2 receptor on adjacent cells. Spike-induced syncytia are beneficial for virus replication, transmission, and immune evasion, and contribute to the progression of COVID-19. In this review, we highlight the properties of viral fusion proteins, mainly the SARS-CoV-2 spike, and the involvement of the host factors in the fusion process. We also highlight the possible use of anti-fusogenic factors as an antiviral for the development of therapeutics against newly emerging SARS-CoV-2 variants and how the fusogenic property of the spike could be exploited for biomedical applications.
Collapse
Affiliation(s)
- Hashim Ali
- Department of Pathology, University of Cambridge, Addenbrookes Hospital, Cambridge CB2 0QQ, UK
| | - Asma Naseem
- Infection, Immunity and Inflammation Research and Teaching Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1DZ, UK
| | - Zaheenul Islam Siddiqui
- Diabetes and Obesity Research Center, NYU Grossman Long Island School of Medicine, New York, NY 11501, USA
| |
Collapse
|
4
|
Chen P, Wu M, He Y, Jiang B, He ML. Metabolic alterations upon SARS-CoV-2 infection and potential therapeutic targets against coronavirus infection. Signal Transduct Target Ther 2023; 8:237. [PMID: 37286535 DOI: 10.1038/s41392-023-01510-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 04/18/2023] [Accepted: 05/19/2023] [Indexed: 06/09/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) caused by coronavirus SARS-CoV-2 infection has become a global pandemic due to the high viral transmissibility and pathogenesis, bringing enormous burden to our society. Most patients infected by SARS-CoV-2 are asymptomatic or have mild symptoms. Although only a small proportion of patients progressed to severe COVID-19 with symptoms including acute respiratory distress syndrome (ARDS), disseminated coagulopathy, and cardiovascular disorders, severe COVID-19 is accompanied by high mortality rates with near 7 million deaths. Nowadays, effective therapeutic patterns for severe COVID-19 are still lacking. It has been extensively reported that host metabolism plays essential roles in various physiological processes during virus infection. Many viruses manipulate host metabolism to avoid immunity, facilitate their own replication, or to initiate pathological response. Targeting the interaction between SARS-CoV-2 and host metabolism holds promise for developing therapeutic strategies. In this review, we summarize and discuss recent studies dedicated to uncovering the role of host metabolism during the life cycle of SARS-CoV-2 in aspects of entry, replication, assembly, and pathogenesis with an emphasis on glucose metabolism and lipid metabolism. Microbiota and long COVID-19 are also discussed. Ultimately, we recapitulate metabolism-modulating drugs repurposed for COVID-19 including statins, ASM inhibitors, NSAIDs, Montelukast, omega-3 fatty acids, 2-DG, and metformin.
Collapse
Affiliation(s)
- Peiran Chen
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Mandi Wu
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China
| | - Yaqing He
- Shenzhen Center for Disease Control and Prevention, Shenzhen, 518055, Guangdong, China
| | - Binghua Jiang
- Cell Signaling and Proteomic Center, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Ming-Liang He
- Department of Biomedical Sciences, City University of Hong Kong, HKSAR, Hong Kong, China.
| |
Collapse
|
5
|
Lysophosphatidylinositol Induced Morphological Changes and Stress Fiber Formation through the GPR55-RhoA-ROCK Pathway. Int J Mol Sci 2022; 23:ijms231810932. [PMID: 36142844 PMCID: PMC9504244 DOI: 10.3390/ijms231810932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/30/2022] Open
Abstract
We previously reported that lysophosphatidylinositol (LPI) functions as an endogenous agonist of GPR55, a novel cannabinoid receptor. However, the physiological roles of LPI-GPR55 have not yet been elucidated in detail. In the present study, we found that LPI induced morphological changes in GPR55-expressing HEK293 cells. LPI induced the cell rounding of GPR55-expressing HEK293 cells but not of empty-vector-transfected cells. LPI also induced the activation of small GTP-binding protein RhoA and increased stress fiber formation in GPR55-expressing HEK293 cells. The inhibition of RhoA and Rho kinase ROCK by the C3 exoenzyme and the ROCK inhibitor reduced LPI-induced cell rounding and stress fiber formation. These results clearly indicated that the LPI-induced morphological changes and the assembly of the cytoskeletons were mediated through the GPR55-RhoA-ROCK pathway.
Collapse
|
6
|
Avota E, Bodem J, Chithelen J, Mandasari P, Beyersdorf N, Schneider-Schaulies J. The Manifold Roles of Sphingolipids in Viral Infections. Front Physiol 2021; 12:715527. [PMID: 34658908 PMCID: PMC8511394 DOI: 10.3389/fphys.2021.715527] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022] Open
Abstract
Sphingolipids are essential components of eukaryotic cells. In this review, we want to exemplarily illustrate what is known about the interactions of sphingolipids with various viruses at different steps of their replication cycles. This includes structural interactions during entry at the plasma membrane or endosomal membranes, early interactions leading to sphingolipid-mediated signal transduction, interactions with internal membranes and lipids during replication, and interactions during virus assembly and budding. Targeted interventions in sphingolipid metabolism - as far as they can be tolerated by cells and organisms - may open novel possibilities to support antiviral therapies. Human immunodeficiency virus type 1 (HIV-1) infections have intensively been studied, but for other viral infections, such as influenza A virus (IAV), measles virus (MV), hepatitis C virus (HCV), dengue virus, Ebola virus, and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), investigations are still in their beginnings. As many inhibitors of sphingolipid metabolism are already in clinical use against other diseases, repurposing studies for applications in some viral infections appear to be a promising approach.
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Janice Chithelen
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Putri Mandasari
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
7
|
Schneider-Schaulies S, Schumacher F, Wigger D, Schöl M, Waghmare T, Schlegel J, Seibel J, Kleuser B. Sphingolipids: Effectors and Achilles Heals in Viral Infections? Cells 2021; 10:cells10092175. [PMID: 34571822 PMCID: PMC8466362 DOI: 10.3390/cells10092175] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/26/2022] Open
Abstract
As viruses are obligatory intracellular parasites, any step during their life cycle strictly depends on successful interaction with their particular host cells. In particular, their interaction with cellular membranes is of crucial importance for most steps in the viral replication cycle. Such interactions are initiated by uptake of viral particles and subsequent trafficking to intracellular compartments to access their replication compartments which provide a spatially confined environment concentrating viral and cellular components, and subsequently, employ cellular membranes for assembly and exit of viral progeny. The ability of viruses to actively modulate lipid composition such as sphingolipids (SLs) is essential for successful completion of the viral life cycle. In addition to their structural and biophysical properties of cellular membranes, some sphingolipid (SL) species are bioactive and as such, take part in cellular signaling processes involved in regulating viral replication. It is especially due to the progress made in tools to study accumulation and dynamics of SLs, which visualize their compartmentalization and identify interaction partners at a cellular level, as well as the availability of genetic knockout systems, that the role of particular SL species in the viral replication process can be analyzed and, most importantly, be explored as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Sibylle Schneider-Schaulies
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Fabian Schumacher
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
| | - Dominik Wigger
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
| | - Marie Schöl
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Trushnal Waghmare
- Institute for Virology and Immunobiology, University of Wuerzburg, 97078 Würzburg, Germany; (S.S.-S.); (M.S.); (T.W.)
| | - Jan Schlegel
- Department for Biotechnology and Biophysics, University of Wuerzburg, 97074 Würzburg, Germany;
| | - Jürgen Seibel
- Department for Organic Chemistry, University of Wuerzburg, 97074 Würzburg, Germany;
| | - Burkhard Kleuser
- Institute of Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, 14195 Berlin, Germany; (F.S.); (D.W.)
- Correspondence: ; Tel.: +49-30-8386-9823
| |
Collapse
|
8
|
Hayashi Y, Tsuchiya K, Yamamoto M, Nemoto-Sasaki Y, Tanigawa K, Hama K, Ueda Y, Tanikawa T, Gohda J, Maeda K, Inoue JI, Yamashita A. N-(4-Hydroxyphenyl) Retinamide Suppresses SARS-CoV-2 Spike Protein-Mediated Cell-Cell Fusion by a Dihydroceramide Δ4-Desaturase 1-Independent Mechanism. J Virol 2021; 95:e0080721. [PMID: 34106748 PMCID: PMC8354230 DOI: 10.1128/jvi.00807-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 05/28/2021] [Indexed: 02/02/2023] Open
Abstract
The membrane fusion between the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and host cells is essential for the initial step of infection; therefore, the host cell membrane components, including sphingolipids, influence the viral infection. We assessed several inhibitors of the enzymes pertaining to sphingolipid metabolism, against SARS-CoV-2 spike protein (S)-mediated cell-cell fusion and viral infection. N-(4-Hydroxyphenyl) retinamide (4-HPR), an inhibitor of dihydroceramide Δ4-desaturase 1 (DES1), suppressed cell-cell fusion and viral infection. The analysis of sphingolipid levels revealed that the inhibition efficiencies of cell-cell fusion and viral infection in 4-HPR-treated cells were consistent with an increased ratio of saturated sphinganine-based lipids to total sphingolipids. We investigated the relationship of DES1 with the inhibition efficiencies of cell-cell fusion. The changes in the sphingolipid profile induced by 4-HPR were mitigated by the supplementation with exogenous cell-permeative ceramide; however, the reduced cell-cell fusion could not be reversed. The efficiency of cell-cell fusion in DES1 knockout (KO) cells was at a level comparable to that in wild-type (WT) cells; however, the ratio of saturated sphinganine-based lipids to the total sphingolipids was higher in DES1 KO cells than in WT cells. 4-HPR reduced cell membrane fluidity without any significant effects on the expression or localization of angiotensin-converting enzyme 2, the SARS-CoV-2 receptor. Therefore, 4-HPR suppresses SARS-CoV-2 S-mediated membrane fusion through a DES1-independent mechanism, and this decrease in membrane fluidity induced by 4-HPR could be the major cause for the inhibition of SARS-CoV-2 infection. IMPORTANCE Sphingolipids could play an important role in SARS-CoV-2 S-mediated membrane fusion with host cells. We studied the cell-cell fusion using SARS-CoV-2 S-expressing cells and sphingolipid-manipulated target cells, with an inhibitor of the sphingolipid metabolism. 4-HPR (also known as fenretinide) is an inhibitor of DES1, and it exhibits antitumor activity and suppresses cell-cell fusion and viral infection. 4-HPR suppresses membrane fusion through a decrease in membrane fluidity, which could possibly be the cause for the inhibition of SARS-CoV-2 infection. There is accumulating clinical data on the safety of 4-HPR. Therefore, it could be a potential candidate drug against COVID-19.
Collapse
Affiliation(s)
| | - Kiyoto Tsuchiya
- AIDS Clinical Center, National Center for Global Health and Medicine Hospital, Tokyo, Japan
| | - Mizuki Yamamoto
- Research Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | - Kotaro Hama
- Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Yusuke Ueda
- Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Takashi Tanikawa
- Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, Saitama, Japan
| | - Jin Gohda
- Research Center for Asian Infectious Diseases, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Kenji Maeda
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | | | | |
Collapse
|
9
|
Dang X, Li Y, Li X, Wang C, Ma Z, Wang L, Fan X, Li Z, Huang D, Xu J, Zhou Z. Lipidomic Profiling Reveals Distinct Differences in Sphingolipids Metabolic Pathway between Healthy Apis cerana cerana larvae and Chinese Sacbrood Disease. INSECTS 2021; 12:insects12080703. [PMID: 34442269 PMCID: PMC8396520 DOI: 10.3390/insects12080703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 07/28/2021] [Accepted: 08/03/2021] [Indexed: 11/16/2022]
Abstract
Chinese sacbrood disease (CSD), which is caused by Chinese sacbrood virus (CSBV), is a major viral disease in Apis cerana cerana larvae. Analysis of lipid composition is critical to the study of CSBV replication. The host lipidome profiling during CSBV infection has not been conducted. This paper identified the lipidome of the CSBV-larvae interaction through high-resolution mass spectrometry. A total of 2164 lipids were detected and divided into 20 categories. Comparison of lipidome between healthy and CSBV infected-larvae showed that 266 lipid species were altered by CSBV infection. Furthermore, qRT-PCR showed that various sphingolipid enzymes and the contents of sphingolipids in the larvae were increased, indicating that sphingolipids may be important for CSBV infection. Importantly, Cer (d14:1 + hO/21:0 + O), DG (41:0e), PE (18:0e/18:3), SM (d20:0/19:1), SM (d37:1), TG (16:0/18:1/18:3), TG (18:1/20:4/21:0) and TG (43:7) were significantly altered in both CSBV_24 h vs. CK_24 h and CSBV_48 h vs. CK_48 h. Moreover, TG (39:6), which was increased by more than 10-fold, could be used as a biomarker for the early detection of CSD. This study provides evidence that global lipidome homeostasis in A. c. cerana larvae is remodeled after CSBV infection. Detailed studies in the future may improve the understanding of the relationship between the sphingolipid pathway and CSBV replication.
Collapse
Affiliation(s)
- Xiaoqun Dang
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Yan Li
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Xiaoqing Li
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Chengcheng Wang
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Zhengang Ma
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Linling Wang
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Xiaodong Fan
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Zhi Li
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Dunyuan Huang
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
| | - Jinshan Xu
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
- Correspondence: (J.X.); (Z.Z.)
| | - Zeyang Zhou
- Chongqing Key Laboratory of Vector Insect, College of Life Science, Chongqing Normal University, Chongqing 401331, China; (X.D.); (Y.L.); (X.L.); (C.W.); (Z.M.); (L.W.); (X.F.); (Z.L.); (D.H.)
- State Key Laboratory of Silkworm Genome Biology, College of Biotechnology, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Microsporidia Infection and Control, Southwest University, Chongqing 400715, China
- Correspondence: (J.X.); (Z.Z.)
| |
Collapse
|
10
|
Ceramide and Related Molecules in Viral Infections. Int J Mol Sci 2021; 22:ijms22115676. [PMID: 34073578 PMCID: PMC8197834 DOI: 10.3390/ijms22115676] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 05/21/2021] [Accepted: 05/21/2021] [Indexed: 02/08/2023] Open
Abstract
Ceramide is a lipid messenger at the heart of sphingolipid metabolism. In concert with its metabolizing enzymes, particularly sphingomyelinases, it has key roles in regulating the physical properties of biological membranes, including the formation of membrane microdomains. Thus, ceramide and its related molecules have been attributed significant roles in nearly all steps of the viral life cycle: they may serve directly as receptors or co-receptors for viral entry, form microdomains that cluster entry receptors and/or enable them to adopt the required conformation or regulate their cell surface expression. Sphingolipids can regulate all forms of viral uptake, often through sphingomyelinase activation, and mediate endosomal escape and intracellular trafficking. Ceramide can be key for the formation of viral replication sites. Sphingomyelinases often mediate the release of new virions from infected cells. Moreover, sphingolipids can contribute to viral-induced apoptosis and morbidity in viral diseases, as well as virus immune evasion. Alpha-galactosylceramide, in particular, also plays a significant role in immune modulation in response to viral infections. This review will discuss the roles of ceramide and its related molecules in the different steps of the viral life cycle. We will also discuss how novel strategies could exploit these for therapeutic benefit.
Collapse
|
11
|
Avota E, Bodem J, Chithelen J, Mandasari P, Beyersdorf N, Schneider-Schaulies J. The Manifold Roles of Sphingolipids in Viral Infections. Front Physiol 2021. [PMID: 34658908 DOI: 10.3389/fphys.2021.71552] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Sphingolipids are essential components of eukaryotic cells. In this review, we want to exemplarily illustrate what is known about the interactions of sphingolipids with various viruses at different steps of their replication cycles. This includes structural interactions during entry at the plasma membrane or endosomal membranes, early interactions leading to sphingolipid-mediated signal transduction, interactions with internal membranes and lipids during replication, and interactions during virus assembly and budding. Targeted interventions in sphingolipid metabolism - as far as they can be tolerated by cells and organisms - may open novel possibilities to support antiviral therapies. Human immunodeficiency virus type 1 (HIV-1) infections have intensively been studied, but for other viral infections, such as influenza A virus (IAV), measles virus (MV), hepatitis C virus (HCV), dengue virus, Ebola virus, and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2), investigations are still in their beginnings. As many inhibitors of sphingolipid metabolism are already in clinical use against other diseases, repurposing studies for applications in some viral infections appear to be a promising approach.
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Janice Chithelen
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Putri Mandasari
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
12
|
Taniguchi M, Okazaki T. Ceramide/Sphingomyelin Rheostat Regulated by Sphingomyelin Synthases and Chronic Diseases in Murine Models. J Lipid Atheroscler 2020; 9:380-405. [PMID: 33024732 PMCID: PMC7521967 DOI: 10.12997/jla.2020.9.3.380] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/16/2020] [Accepted: 07/19/2020] [Indexed: 12/16/2022] Open
Abstract
Ceramide and sphingomyelin (SM) are major components of the double membrane-bound sphingolipids. Ceramide is an essential bioactive lipid involved in numerous cell processes including apoptosis, necrosis, and autophagy-dependent cell death. Inversely, SM regulates opposite cellular processes such as proliferation and migration by changing receptor-mediated signal transduction in the lipid microdomain. SM is generated through a transfer of phosphocholine from phosphatidylcholine to ceramide by SM synthases (SMSs). Research during the past several decades has revealed that the ceramide/SM balance in cellular membranes regulated by SMSs is important to decide the cell fate, survival, and proliferation. In addition, recent experimental studies utilizing SMS knockout mice and murine disease models provide evidence that SMS-regulated ceramide/SM balance is involved in human diseases. Here, we review the basic structural and functional characteristics of SMSs and focus on their cellular functions through the regulation of ceramide/SM balance in membrane microdomains. In addition, we present the pathological or physiological implications of SMSs by analyzing their role in SMS-knockout mice and human disease models. This review finally presents evidence indicating that the regulation of ceramide/SM balance through SMS could be a therapeutic target for human disorders.
Collapse
Affiliation(s)
- Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Kahoku, Japan
| | - Toshiro Okazaki
- Research Institute for Bioresources and Biotechnology, Kanazawa Prefectural University, Nonoichi, Japan
| |
Collapse
|
13
|
Murakami C, Hoshino F, Sakai H, Hayashi Y, Yamashita A, Sakane F. Diacylglycerol kinase δ and sphingomyelin synthase-related protein functionally interact via their sterile α motif domains. J Biol Chem 2020; 295:2932-2947. [PMID: 31980461 DOI: 10.1074/jbc.ra119.012369] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/23/2020] [Indexed: 12/27/2022] Open
Abstract
The δ isozyme of diacylglycerol kinase (DGKδ) plays critical roles in lipid signaling by converting diacylglycerol (DG) to phosphatidic acid (PA). We previously demonstrated that DGKδ preferably phosphorylates palmitic acid (16:0)- and/or palmitoleic acid (16:1)-containing DG molecular species, but not arachidonic acid (20:4)-containing DG species, which are recognized as DGK substrates derived from phosphatidylinositol turnover, in high glucose-stimulated myoblasts. However, little is known about the origin of these DG molecular species. DGKδ and two DG-generating enzymes, sphingomyelin synthase (SMS) 1 and SMS-related protein (SMSr), contain a sterile α motif domain (SAMD). In this study, we found that SMSr-SAMD, but not SMS1-SAMD, co-immunoprecipitates with DGKδ-SAMD. Full-length DGKδ co-precipitated with full-length SMSr more strongly than with SMS1. However, SAMD-deleted variants of SMSr and DGKδ interacted only weakly with full-length DGKδ and SMSr, respectively. These results strongly suggested that DGKδ interacts with SMSr through their respective SAMDs. To determine the functional outcomes of the relationship between DGKδ and SMSr, we used LC-MS/MS to investigate whether overexpression of DGKδ and/or SMSr in COS-7 cells alters the levels of PA species. We found that SMSr overexpression significantly enhances the production of 16:0- or 16:1-containing PA species such as 14:0/16:0-, 16:0/16:0-, 16:0/18:1-, and/or 16:1/18:1-PA in DGKδ-overexpressing COS-7 cells. Moreover, SMSr enhanced DGKδ activity via their SAMDs in vitro Taken together, these results strongly suggest that SMSr is a candidate DG-providing enzyme upstream of DGKδ and that the two enzymes represent a new pathway independent of phosphatidylinositol turnover.
Collapse
Affiliation(s)
- Chiaki Murakami
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Fumi Hoshino
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan
| | - Hiromichi Sakai
- Department of Biosignaling and Radioisotope Experiment, Interdisciplinary Center for Science Research, Organization for Research and Academic Information, Shimane University, Izumo 693-8501, Japan
| | - Yasuhiro Hayashi
- Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan
| | - Atsushi Yamashita
- Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan
| | - Fumio Sakane
- Department of Chemistry, Graduate School of Science, Chiba University, Chiba 263-8522, Japan.
| |
Collapse
|
14
|
Avota E, de Lira MN, Schneider-Schaulies S. Sphingomyelin Breakdown in T Cells: Role of Membrane Compartmentalization in T Cell Signaling and Interference by a Pathogen. Front Cell Dev Biol 2019; 7:152. [PMID: 31457008 PMCID: PMC6700246 DOI: 10.3389/fcell.2019.00152] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 07/22/2019] [Indexed: 12/15/2022] Open
Abstract
Sphingolipids are major components of cellular membranes, and at steady-state level, their metabolic fluxes are tightly controlled. On challenge by external signals, they undergo rapid turnover, which substantially affects the biophysical properties of membrane lipid and protein compartments and, consequently, signaling and morphodynamics. In T cells, external cues translate into formation of membrane microdomains where proximal signaling platforms essential for metabolic reprograming and cytoskeletal reorganization are organized. This review will focus on sphingomyelinases, which mediate sphingomyelin breakdown and ensuing ceramide release that have been implicated in T-cell viability and function. Acting at the sphingomyelin pool at the extrafacial or cytosolic leaflet of cellular membranes, acid and neutral sphingomyelinases organize ceramide-enriched membrane microdomains that regulate T-cell homeostatic activity and, upon stimulation, compartmentalize receptors, membrane proximal signaling complexes, and cytoskeletal dynamics as essential for initiating T-cell motility and interaction with endothelia and antigen-presenting cells. Prominent examples to be discussed in this review include death receptor family members, integrins, CD3, and CD28 and their associated signalosomes. Progress made with regard to experimental tools has greatly aided our understanding of the role of bioactive sphingolipids in T-cell biology at a molecular level and of targets explored by a model pathogen (measles virus) to specifically interfere with their physiological activity.
Collapse
Affiliation(s)
- Elita Avota
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | - Maria Nathalia de Lira
- Institute for Virology and Immunobiology, Julius Maximilian University of Würzburg, Würzburg, Germany
| | | |
Collapse
|
15
|
Wang P, Li Z, Jiang L, Zhou L, Ye D. Design and Synthesis of the Diazirine-based Clickable Photo-affinity Probe Targeting Sphingomyelin Synthase 2. LETT DRUG DES DISCOV 2019. [DOI: 10.2174/1570180816666181106154601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Background:SMS family plays a very important role in sphingolipids metabolism and is involved in the membrane mobility and signaling transduction.Methods:SMS2 subtype was related to a variety of diseases and could be regarded as a promising potential drug target. However, the uncertainty of the binding sites and the molecular mechanism of action limited the development of SMS2 inhibitors. Herein, we discovered a photo-affinity probe PAL-1 targeting SMS2.Results:The enzyme inhibitory activity and the photo-affinity labeling experiments showed that PAL-1 could be mono-labeled on SMS2.Conclusion:In summary, starting from the N-arylbenzamides core structure and the minimalist terminal alkyne-containing diazirine photo-crosslinker, we designed and synthesized a photoaffinity probe PAL-1 targeting SMS2. The enzymatic inhibitory activity study showed that PAL-1 exhibited superior selectivities for SMS2 with an IC50 of 0.37 µM over SMS1.
Collapse
Affiliation(s)
- Penghui Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Zhining Li
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Lulu Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Lu Zhou
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| | - Deyong Ye
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
16
|
Shi XX, Zhang H, Chen M, Zhang YD, Zhu MF, Zhang MJ, Li FQ, Wratten S, Zhou WW, Mao C, Zhu ZR. Two sphingomyelin synthase homologues regulate body weight and sphingomyelin synthesis in female brown planthopper, N. lugens (Stål). INSECT MOLECULAR BIOLOGY 2019; 28:253-263. [PMID: 30375099 DOI: 10.1111/imb.12549] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Although sphingomyelins known to be are lipid constituents of the plasma membrane in vertebrates, much remains obscure about the metabolism of sphingomyelins in insects. With ultra performance liquid chromatography-time-of-flight-tandem mass spectrometry analysis, we revealed for the first time that sphingomyelins are abundant in Nilaparvata lugens (Stål), the brown planthopper (BPH), and their biosynthesis is carried out by sphingomyelin synthase-like protein 2 (SMSL2), which is homologous to sphingomyelin synthase-related protein (SMSr). Unlike other insect species, high concentrations of sphingomyelins rather than ceramide phosphoethanolamines exist in the BPH. Two putative genes, which are homologous to SMSr, are named Nilaparvata lugens SMS-like 1 (NlSMSL1) and 2 (NlSMSL2). Knockdowns of both NlSMSL2 and NlSMSL1 were conducted but only the first decreased concentrations of sphingomyelins in the BPH, indicating that NlSMSL2 plays a role in the biosynthesis of sphingomyelins. Real-time quantitative PCR analysis revealed both NlSMSL1 and NlSMSL2 are highly expressed in BPH adults, with NlSMSL1 specifically highly expressed in reproductive organs (ovaries and testes) whereas NlSMSL2 was highly expressed in the malpighian tubules. The knockdown of NlSMSL1 or NlSMSL2 increased BPH female body weight but not that of males, suggesting sex-specific roles for SMSLs in influencing BPH body weight. The results suggest that NlSMSL2 catalyses the synthesis of sphingomyelins and maintains female BPH body weight through alteration of sphingolipid content.
Collapse
Affiliation(s)
- X-X Shi
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
- Institute of Insect Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - H Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M Chen
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - Y-D Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-F Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - M-J Zhang
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - F-Q Li
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - S Wratten
- Bio-Protection Research Centre, Lincoln University, Lincoln, New Zealand
| | - W-W Zhou
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| | - C Mao
- Department of Medicine and Stony Brook Cancer Center, The State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Z-R Zhu
- State Key Laboratory of Rice Biology, Key Laboratory of Molecular Biology of Crop Pathogens and Insects, Ministry of Agriculture, Hangzhou, Zhejiang, China
| |
Collapse
|
17
|
Hayashi Y, Nemoto-Sasaki Y, Matsumoto N, Hama K, Tanikawa T, Oka S, Saeki T, Kumasaka T, Koizumi T, Arai S, Wada I, Yokoyama K, Sugiura T, Yamashita A. Complex formation of sphingomyelin synthase 1 with glucosylceramide synthase increases sphingomyelin and decreases glucosylceramide levels. J Biol Chem 2018; 293:17505-17522. [PMID: 30242129 PMCID: PMC6231140 DOI: 10.1074/jbc.ra118.002048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 08/21/2018] [Indexed: 12/23/2022] Open
Abstract
Sphingolipids, including sphingomyelin (SM) and glucosylceramide (GlcCer), are generated by the addition of a polar head group to ceramide (Cer). Sphingomyelin synthase 1 (SMS1) and glucosylceramide synthase (GCS) are key enzymes that catalyze the conversion of Cer to SM and GlcCer, respectively. GlcCer synthesis has been postulated to occur mainly in cis-Golgi, and SM synthesis is thought to occur in medial/trans-Golgi; however, SMS1 and GCS are known to partially co-localize in cisternae, especially in medial/trans-Golgi. Here, we report that SMS1 and GCS can form a heteromeric complex, in which the N terminus of SMS1 and the C terminus of GCS are in close proximity. Deletion of the N-terminal sterile α-motif of SMS1 reduced the stability of the SMS1-GCS complex, resulting in a significant reduction in SM synthesis in vivo In contrast, chemical-induced heterodimerization augmented SMS1 activity, depending on an increase in the amount and stability of the complex. Fusion of the SMS1 N terminus to the GCS C terminus via linkers of different lengths increased SM synthesis and decreased GlcCer synthesis in vivo These results suggest that formation of the SMS1-GCS heteromeric complex increases SM synthesis and decreases GlcCer synthesis. Importantly, this regulation of relative Cer levels by the SMS1-GCS complex was confirmed by CRISPR/Cas9-mediated knockout of SMS1 or GCS combined with pharmacological inhibition of Cer transport protein in HEK293T cells. Our findings suggest that complex formation between SMS1 and GCS is part of a critical mechanism controlling the metabolic fate of Cer in the Golgi.
Collapse
Affiliation(s)
- Yasuhiro Hayashi
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Yoko Nemoto-Sasaki
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Naoki Matsumoto
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Kotaro Hama
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Takashi Tanikawa
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Saori Oka
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Tadaaki Saeki
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Tatsuya Kumasaka
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Takanori Koizumi
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Seisuke Arai
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima City, Fukushima 960-1295, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima City, Fukushima 960-1295, Japan
| | - Kazuaki Yokoyama
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Takayuki Sugiura
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| | - Atsushi Yamashita
- From the Faculty of Pharma-Science, Teikyo University, Tokyo 173- 8605, Japan and
| |
Collapse
|
18
|
D'Angelo G, Moorthi S, Luberto C. Role and Function of Sphingomyelin Biosynthesis in the Development of Cancer. Adv Cancer Res 2018; 140:61-96. [PMID: 30060817 DOI: 10.1016/bs.acr.2018.04.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sphingomyelin (SM) biosynthesis represents a complex, finely regulated process, mostly occurring in vertebrates. It is intimately linked to lipid transport and it is ultimately carried out by two enzymes, SM synthase 1 and 2, selectively localized in the Golgi and plasma membrane. In the course of the SM biosynthetic reaction, various lipids are metabolized. Because these lipids have both structural and signaling functions, the SM biosynthetic process has the potential to affect diverse important cellular processes (such as cell proliferation, cell survival, and migration). Thus defects in SM biosynthesis might directly or indirectly impact the normal physiology of the cell and eventually of the organism. In this chapter, we will focus on evidence supporting a role for SM biosynthesis in specific cellular functions and how its dysregulation can affect neoplastic transformation.
Collapse
Affiliation(s)
- Giovanni D'Angelo
- Institute of Protein Biochemistry, National Research Council of Italy, Naples, Italy
| | - Sitapriya Moorthi
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
| | - Chiara Luberto
- Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
19
|
Abstract
Studies of bioactive lipids in general and sphingolipids in particular have intensified over the past several years, revealing an unprecedented and unanticipated complexity of the lipidome and its many functions, which rivals, if not exceeds, that of the genome or proteome. These results highlight critical roles for bioactive sphingolipids in most, if not all, major cell biological responses, including all major cell signalling pathways, and they link sphingolipid metabolism to key human diseases. Nevertheless, the fairly nascent field of bioactive sphingolipids still faces challenges in its biochemical and molecular underpinnings, including defining the molecular mechanisms of pathway and enzyme regulation, the study of lipid-protein interactions and the development of cellular probes, suitable biomarkers and therapeutic approaches.
Collapse
Affiliation(s)
- Yusuf A Hannun
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
| | - Lina M Obeid
- Stony Brook Cancer Center and Department of Medicine, Stony Brook University, New York 11794, USA
- Northport Veterans Affairs Medical Center, Northport, New York 11768, USA
| |
Collapse
|
20
|
Chen Y, Cao Y. The sphingomyelin synthase family: proteins, diseases, and inhibitors. Biol Chem 2017; 398:1319-1325. [PMID: 28742512 DOI: 10.1515/hsz-2017-0148] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/13/2017] [Indexed: 12/18/2022]
Abstract
Sphingomyelin (SM) is among the most important biomolecules in eukaryotes and acts as both constructive components and signal carrier in physiological processes. SM is catalyzed by a membrane protein family, sphingomyelin synthases (SMSs), consisting of three members, SMS1, SMS2 and SMSr. SMSs modulate sphingomyelin and other sphingolipids levels, thereby regulating membrane mobility, ceramide-dependent apoptosis and DAG-dependent signaling pathways. SMSs was found associated with various diseases. Downregulation of SMS2 activity results in protective effects against obesity, atherosclerosis and diabetes and makes SMS2 inhibitors potential medicines. Structural guided specific drug design could be the next breakthrough, discriminating SMS2 from other homologs.
Collapse
|
21
|
Ohnishi T, Hashizume C, Taniguchi M, Furumoto H, Han J, Gao R, Kinami S, Kosaka T, Okazaki T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer. FASEB J 2017; 31:3816-3830. [PMID: 28522594 DOI: 10.1096/fj.201601225rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 04/24/2017] [Indexed: 12/13/2022]
Abstract
Sphingomyelin synthase 2 (SMS2) is the synthetic enzyme of sphingomyelin (SM), which regulates membrane fluidity and microdomain structure. SMS2 plays a role in LPS-induced lung injury and inflammation; however, its role in inflammation-mediated tumorigenesis is unclear. We investigated the effect of SMS2 deficiency on dextran sodium sulfate (DSS)-induced murine colitis and found inhibition of DSS-induced inflammation in SMS2-deficient (SMS2-/-) mice. DSS treatment induced a significant increase in ceramide levels, with a decrease of SM levels in SMS2-/- colon tissue, and demonstrated attenuation of the elevation of both inflammation-related gene expression and proinflammatory cytokines and chemokines, leukocyte infiltration, and MAPK and signal transducer and activator of transcription 3 activation. After undergoing transplantation of wild-type bone marrow, SMS2-/- mice also exhibited inhibition of DSS-induced inflammation in the colon, which suggested that SMS2 deficiency in bone marrow-derived immune cells was not involved in the inhibition of colitis. Finally, in an azoxymethane/DSS-induced cancer model, SMS2 deficiency significantly decreased tumor incidence in the colon. Our results demonstrate that SMS2 deficiency inhibits DSS-induced colitis and subsequent colitis-associated colon cancer via inhibition of colon epithelial cell-mediated inflammation; therefore, inhibition of SMS2 may be a potential therapeutic target for human colitis and colorectal cancer.-Ohnishi, T., Hashizume, C., Taniguchi, M., Furumoto, H., Han, J., Gao, R., Kinami, S., Kosaka, T., Okazaki, T. Sphingomyelin synthase 2 deficiency inhibits the induction of murine colitis-associated colon cancer.
Collapse
Affiliation(s)
- Toshio Ohnishi
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Chieko Hashizume
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Makoto Taniguchi
- Department of Life Science, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| | - Hidehiro Furumoto
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Jia Han
- Division of Medical Oncology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Rongfen Gao
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Shinichi Kinami
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Takeo Kosaka
- Division of General and Digestive Surgery, Department of Medicine, Kanazawa Medical University, Uchinada, Japan
| | - Toshiro Okazaki
- Division of Hematology/Immunology, Department of Medicine, Kanazawa Medical University, Uchinada, Japan;
- Department of Advanced Medicine, Medical Research Institute, Kanazawa Medical University, Uchinada, Japan
| |
Collapse
|
22
|
Qi XY, Cao Y, Li YL, Mo MG, Zhou L, Ye DY. Discovery of the selective sphingomyelin synthase 2 inhibitors with the novel structure of oxazolopyridine. Bioorg Med Chem Lett 2017; 27:3511-3515. [DOI: 10.1016/j.bmcl.2017.05.074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 05/19/2017] [Accepted: 05/22/2017] [Indexed: 11/30/2022]
|
23
|
Jiao WJ, Li FQ, Bai YL, Shi XX, Zhu MF, Zhang MJ, Mao CG, Zhu ZR. Rice Stripe Virus Infection Alters mRNA Levels of Sphingolipid-Metabolizing Enzymes and Sphingolipids Content in Laodelphax striatellus. JOURNAL OF INSECT SCIENCE (ONLINE) 2017; 17:iew111. [PMID: 28130458 PMCID: PMC5270407 DOI: 10.1093/jisesa/iew111] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Indexed: 05/05/2023]
Abstract
Sphingolipids and their metabolites have been implicated in viral infection and replication in mammal cells but how their metabolizing enzymes in the host are regulated by viruses remains largely unknown. Here we report the identification of 12 sphingolipid genes and their regulation by Rice stripe virus in the small brown planthopper (Laodelphax striatellus Fallén), a serious pest of rice throughout eastern Asia. According to protein sequence similarity, we identified 12 sphingolipid enzyme genes in L. striatellus. By comparing their mRNA levels in viruliferous versus nonviruliferous L. striatellus at different life stages by qPCR, we found that RSV infection upregulated six genes (LsCGT1, LsNAGA1, LsSGPP, LsSMPD4, LsSMS, and LsSPT) in most stages of L. striatellus Especially, four genes (LsCGT1, LsSMPD2, LsNAGA1, and LsSMS) and another three genes (LsNAGA1, LsSGPP, and LsSMS) were significantly upregulated in viruliferous third-instar and fourth-instar nymphs, respectively. HPLC-MS/MS results showed that RSV infection increased the levels of various ceramides, such as Cer18:0, Cer20:0, and Cer22:0 species, in third and fourth instar L. striatellus nymphs. Together, these results demonstrate that RSV infection alters the transcript levels of various sphingolipid enzymes and the contents of sphingolipids in L. striatellus, indicating that sphingolipids may be important for RSV infection or replication in L. striatellus.
Collapse
Affiliation(s)
- Wen-Juan Jiao
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Fei-Qiang Li
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Yue-Liang Bai
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiao-Xiao Shi
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mu-Fei Zhu
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Min-Jing Zhang
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| | - Cun-Gui Mao
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Zeng-Rong Zhu
- State Key Laboratory of Rice Biology/Key Laboratory of Agricultural Entomology Ministry of Agriculture/Institute of Insect Sciences, Zhejiang University, Hangzhou 310058, China
| |
Collapse
|
24
|
Hayashi Y, Nemoto-Sasaki Y, Matsumoto N, Tanikawa T, Oka S, Tanaka Y, Arai S, Wada I, Sugiura T, Yamashita A. Carboxyl-terminal Tail-mediated Homodimerizations of Sphingomyelin Synthases Are Responsible for Efficient Export from the Endoplasmic Reticulum. J Biol Chem 2016; 292:1122-1141. [PMID: 27927984 DOI: 10.1074/jbc.m116.746602] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Revised: 11/16/2016] [Indexed: 11/06/2022] Open
Abstract
Sphingomyelin synthase (SMS) is the key enzyme for cross-talk between bioactive sphingolipids and glycerolipids. In mammals, SMS consists of two isoforms: SMS1 is localized in the Golgi apparatus, whereas SMS2 is localized in both the Golgi and plasma membranes. SMS2 seems to exert cellular functions through protein-protein interactions; however, the existence and functions of quaternary structures of SMS1 and SMS2 remain unclear. Here we demonstrate that both SMS1 and SMS2 form homodimers. The SMSs have six membrane-spanning domains, and the N and C termini of both proteins face the cytosolic side of the Golgi apparatus. Chemical cross-linking and bimolecular fluorescence complementation revealed that the N- and/or C-terminal tails of the SMSs were in close proximity to those of the other SMS in the homodimer. Homodimer formation was significantly decreased by C-terminal truncations, SMS1-ΔC22 and SMS2-ΔC30, indicating that the C-terminal tails of the SMSs are primarily responsible for homodimer formation. Moreover, immunoprecipitation using deletion mutants revealed that the C-terminal tail of SMS2 mainly interacted with the C-terminal tail of its homodimer partner, whereas the C-terminal tail of SMS1 mainly interacted with a site other than the C-terminal tail of its homodimer partner. Interestingly, homodimer formation occurred in the endoplasmic reticulum (ER) membrane before trafficking to the Golgi apparatus. Reduced homodimerization caused by C-terminal truncations of SMSs significantly reduced ER-to-Golgi transport. Our findings suggest that the C-terminal tails of SMSs are involved in homodimer formation, which is required for efficient transport from the ER.
Collapse
Affiliation(s)
- Yasuhiro Hayashi
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Yoko Nemoto-Sasaki
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Naoki Matsumoto
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Takashi Tanikawa
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Saori Oka
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Yusuke Tanaka
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Seisuke Arai
- the Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Hikarigaoka-1, Fukushima City, Fukushima 960-1295, Japan
| | - Ikuo Wada
- the Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Hikarigaoka-1, Fukushima City, Fukushima 960-1295, Japan
| | - Takayuki Sugiura
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| | - Atsushi Yamashita
- From the Faculty of Pharma Sciences, Teikyo University, Kaga 2-11-1, Itabashi-ku, Tokyo 173-8605, Japan and
| |
Collapse
|
25
|
Grammatikos G, Dietz J, Ferreiros N, Koch A, Dultz G, Bon D, Karakasiliotis I, Lutz T, Knecht G, Gute P, Herrmann E, Zeuzem S, Mavromara P, Sarrazin C, Pfeilschifter J. Persistence of HCV in Acutely-Infected Patients Depletes C24-Ceramide and Upregulates Sphingosine and Sphinganine Serum Levels. Int J Mol Sci 2016; 17:E922. [PMID: 27304952 PMCID: PMC4926455 DOI: 10.3390/ijms17060922] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Revised: 05/27/2016] [Accepted: 05/31/2016] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) substantially affects lipid metabolism, and remodeling of sphingolipids appears to be essential for HCV persistence in vitro. The aim of the current study is the evaluation of serum sphingolipid variations during acute HCV infection. We enrolled prospectively 60 consecutive patients with acute HCV infection, most of them already infected with human immunodeficiency virus (HIV), and serum was collected at the time of diagnosis and longitudinally over a six-month period until initiation of antiviral therapy or confirmed spontaneous clearance. Quantification of serum sphingolipids was performed by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Spontaneous clearance was observed in 11 out of 60 patients (18.3%), a sustained viral response (SVR) in 43 out of 45 patients (95.5%) receiving an antiviral treatment after follow-up, whereas persistence of HCV occurred in six out of 60 patients (10%). C24-ceramide (C24-Cer)-levels increased at follow-up in patients with spontaneous HCV eradication (p < 0.01), as compared to baseline. Sphingosine and sphinganine values were significantly upregulated in patients unable to clear HCV over time compared to patients with spontaneous clearance of HCV infection on follow-up (p = 0.013 and 0.006, respectively). In summary, the persistence of HCV after acute infection induces a downregulation of C24Cer and a simultaneous elevation of serum sphingosine and sphinganine concentrations.
Collapse
Affiliation(s)
- Georgios Grammatikos
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
- Medizinische Klinik 1, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Julia Dietz
- Medizinische Klinik 1, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Nerea Ferreiros
- Pharmazentrum Frankfurt, Institut für klinische Pharmakologie, Goethe University Hospital, 60590 Frankfurt am Main, Germany.
| | - Alexander Koch
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Georg Dultz
- Medizinische Klinik 1, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Dimitra Bon
- Institute of Biostatistics and Mathematical Modelling, Department of Medicine, Goethe University, 60590 Frankfurt am Main, Germany.
| | | | - Thomas Lutz
- Infektiologikum, 60590 Frankfurt am Main, Germany.
| | - Gaby Knecht
- Infektiologikum, 60590 Frankfurt am Main, Germany.
| | - Peter Gute
- Infektiologikum, 60590 Frankfurt am Main, Germany.
| | - Eva Herrmann
- Pharmazentrum Frankfurt, Institut für klinische Pharmakologie, Goethe University Hospital, 60590 Frankfurt am Main, Germany.
| | - Stefan Zeuzem
- Medizinische Klinik 1, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Penelope Mavromara
- Molecular Virology, Hellenic Pasteur Institute, 11521 Athens, Greece.
- Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece.
| | - Christoph Sarrazin
- Medizinische Klinik 1, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| | - Josef Pfeilschifter
- Pharmazentrum Frankfurt, Institut für Allgemeine Pharmakologie, Goethe University Hospital, Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany.
| |
Collapse
|
26
|
Kishimoto T, Ishitsuka R, Kobayashi T. Detectors for evaluating the cellular landscape of sphingomyelin- and cholesterol-rich membrane domains. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:812-829. [PMID: 26993577 DOI: 10.1016/j.bbalip.2016.03.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/09/2016] [Accepted: 03/09/2016] [Indexed: 12/11/2022]
Abstract
Although sphingomyelin and cholesterol are major lipids of mammalian cells, the detailed distribution of these lipids in cellular membranes remains still obscure. However, the recent development of protein probes that specifically bind sphingomyelin and/or cholesterol provides new information about the landscape of the lipid domains that are enriched with sphingomyelin or cholesterol or both. Here, we critically summarize the tools to study distribution and dynamics of sphingomyelin and cholesterol. This article is part of a Special Issue entitled: The cellular lipid landscape edited by Tim P. Levine and Anant K. Menon.
Collapse
Affiliation(s)
| | - Reiko Ishitsuka
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan
| | - Toshihide Kobayashi
- Lipid Biology Laboratory, RIKEN, Wako, Saitama 351-0198, Japan; INSERM U1060, Université Lyon 1, Villeurbanne 69621, France.
| |
Collapse
|
27
|
P2X1 Receptor Antagonists Inhibit HIV-1 Fusion by Blocking Virus-Coreceptor Interactions. J Virol 2015; 89:9368-82. [PMID: 26136569 DOI: 10.1128/jvi.01178-15] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/23/2015] [Indexed: 01/03/2023] Open
Abstract
UNLABELLED HIV-1 Env glycoprotein-mediated fusion is initiated upon sequential binding of Env to CD4 and the coreceptor CXCR4 or CCR5. Whereas these interactions are thought to be necessary and sufficient to promote HIV-1 fusion, other host factors can modulate this process. Previous studies reported potent inhibition of HIV-1 fusion by selective P2X1 receptor antagonists, including NF279, and suggested that these receptors play a role in HIV-1 entry. Here we investigated the mechanism of antiviral activity of NF279 and found that this compound does not inhibit HIV-1 fusion by preventing the activation of P2X1 channels but effectively blocks the binding of the virus to CXCR4 or CCR5. The notion of an off-target effect of NF279 on HIV-1 fusion is supported by the lack of detectable expression of P2X1 receptors in cells used in fusion experiments and by the fact that the addition of ATP or the enzymatic depletion of ATP in culture medium does not modulate viral fusion. Importantly, NF279 fails to inhibit HIV-1 fusion with cell lines and primary macrophages when added at an intermediate stage downstream of Env-CD4-coreceptor engagement. Conversely, in the presence of NF279, HIV-1 fusion is arrested downstream of CD4 binding but prior to coreceptor engagement. NF279 also antagonizes the signaling function of CCR5, CXCR4, and another chemokine receptor, as evidenced by the suppression of calcium responses elicited by specific ligands and by recombinant gp120. Collectively, our results demonstrate that NF279 is a dual HIV-1 coreceptor inhibitor that interferes with the functional engagement of CCR5 and CXCR4 by Env. IMPORTANCE Inhibition of P2X receptor activity suppresses HIV-1 fusion and replication, suggesting that P2X signaling is involved in HIV-1 entry. However, mechanistic experiments conducted in this study imply that P2X1 receptor is not expressed in target cells or involved in viral fusion. Instead, we found that inhibition of HIV-1 fusion by a specific P2X1 receptor antagonist, NF279, is due to the blocking of virus interactions with both the CXCR4 and CCR5 coreceptors. The ability of NF279 to abrogate cellular calcium signaling induced by the respective chemokines showed that this compound acts as a dual-coreceptor antagonist. P2X1 receptor antagonists could thus represent a new class of dual-coreceptor inhibitors with a structure and a mechanism of action that are distinct from those of known HIV-1 coreceptor antagonists.
Collapse
|