1
|
Amiar S, Johnson KA, Husby ML, Marzi A, Stahelin RV. A fatty acid-ordered plasma membrane environment is critical for Ebola virus matrix protein assembly and budding. J Lipid Res 2024; 65:100663. [PMID: 39369791 PMCID: PMC11565396 DOI: 10.1016/j.jlr.2024.100663] [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: 06/11/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/08/2024] Open
Abstract
Plasma membrane (PM) domains and order phases have been shown to play a key role in the assembly, release, and entry of several lipid-enveloped viruses. In the present study, we provide a mechanistic understanding of the Ebola virus (EBOV) matrix protein VP40 interaction with PM lipids and their effect on VP40 oligomerization, a crucial step for viral assembly and budding. VP40 matrix formation is sufficient to induce changes in the PM fluidity. We demonstrate that the distance between the lipid headgroups, the fatty acid tail saturation, and the PM order are important factors for the stability of VP40 binding and oligomerization at the PM. The use of FDA-approved drugs to fluidize the PM destabilizes the viral matrix assembly leading to a reduction in budding efficiency. Overall, these findings support an EBOV assembly mechanism that reaches beyond lipid headgroup specificity by using ordered PM lipid regions independent of cholesterol.
Collapse
Affiliation(s)
- Souad Amiar
- Borch Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, IN; Purdue Institute of Inflammation, Immunology, and Infectious Disease (PI4D), Purdue University, West Lafayette, IN
| | - Kristen A Johnson
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN
| | - Monica L Husby
- Borch Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, IN
| | - Andrea Marzi
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT
| | - Robert V Stahelin
- Borch Department of Medicinal Chemistry & Molecular Pharmacology, Purdue University, West Lafayette, IN; Purdue Institute of Inflammation, Immunology, and Infectious Disease (PI4D), Purdue University, West Lafayette, IN.
| |
Collapse
|
2
|
Swissa E, Monsonego U, Yang LT, Schori L, Kamintsky L, Mirloo S, Burger I, Uzzan S, Patel R, Sudmant PH, Prager O, Kaufer D, Friedman A. Cortical plasticity is associated with blood-brain barrier modulation. eLife 2024; 12:RP89611. [PMID: 39024007 PMCID: PMC11257677 DOI: 10.7554/elife.89611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2024] Open
Abstract
Brain microvessels possess the unique properties of a blood-brain barrier (BBB), tightly regulating the passage of molecules from the blood to the brain neuropil and vice versa. In models of brain injury, BBB dysfunction and the associated leakage of serum albumin to the neuropil have been shown to induce pathological plasticity, neuronal hyper-excitability, and seizures. The effect of neuronal activity on BBB function and whether it plays a role in plasticity in the healthy brain remain unclear. Here we show that neuronal activity induces modulation of microvascular permeability in the healthy brain and that it has a role in local network reorganization. Combining simultaneous electrophysiological recording and vascular imaging with transcriptomic analysis in rats, and functional and BBB-mapping MRI in human subjects, we show that prolonged stimulation of the limb induces a focal increase in BBB permeability in the corresponding somatosensory cortex that is associated with long-term synaptic plasticity. We further show that the increased microvascular permeability depends on neuronal activity and involves caveolae-mediated transcytosis and transforming growth factor β signaling. Our results reveal a role of BBB modulation in cortical plasticity in the healthy brain, highlighting the importance of neurovascular interactions for sensory experience and learning.
Collapse
Affiliation(s)
- Evyatar Swissa
- Department of Brain and Cognitive Sciences, The School of Brain Sciences and Cognition, Zlotowski Center for Neuroscience, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Uri Monsonego
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Lynn T Yang
- Department of Integrative Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Lior Schori
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Lyna Kamintsky
- Department of Medical Neuroscience, Dalhousie UniversityHalifaxCanada
| | - Sheida Mirloo
- Department of Medical Neuroscience, Dalhousie UniversityHalifaxCanada
| | - Itamar Burger
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Sarit Uzzan
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Rishi Patel
- Department of Integrative Biology, University of California, BerkeleyBerkeleyUnited States
| | - Peter H Sudmant
- Department of Integrative Biology, University of California, BerkeleyBerkeleyUnited States
| | - Ofer Prager
- Department of Brain and Cognitive Sciences, The School of Brain Sciences and Cognition, Zlotowski Center for Neuroscience, Ben-Gurion University of the NegevBeer-ShevaIsrael
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
| | - Daniela Kaufer
- Department of Integrative Biology, University of California, BerkeleyBerkeleyUnited States
- Helen Wills Neuroscience Institute, University of California, BerkeleyBerkeleyUnited States
| | - Alon Friedman
- Department of Brain and Cognitive Sciences, The School of Brain Sciences and Cognition, Zlotowski Center for Neuroscience, Ben-Gurion University of the NegevBeer-ShevaIsrael
- Department of Physiology and Cell Biology, Faculty of Health Sciences, Ben-Gurion University of the NegevBeer-ShevaIsrael
- Department of Medical Neuroscience, Dalhousie UniversityHalifaxCanada
| |
Collapse
|
3
|
Cejas JDP, Rosa AS, González Paz AN, Disalvo EA, Frías MDLA. Impact of chlorogenic acid on surface and phase properties of cholesterol-enriched phosphatidylcholine membranes. Arch Biochem Biophys 2024; 753:109913. [PMID: 38286353 DOI: 10.1016/j.abb.2024.109913] [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: 10/13/2023] [Revised: 11/26/2023] [Accepted: 01/25/2024] [Indexed: 01/31/2024]
Abstract
This study analyses the insertion of Chlorogenic acid (CGA) in phosphatidylcholine (PC) membranes enriched with cholesterol (Chol). While cholesterol decreases the area per lipid and increases the dipole potential, CGA increases and decreases these values, respectively. When CGA is inserted into cholesterol-containing DMPC membranes, these effects cancel out, resulting in values that overlap with those of DMPC monolayers without Chol and CGA. The presence of CGA also compensates the increase of dipole potential produced by Chol which can be explain as a consequence of the orientation of CGA molecule at the interphase opposing the cholesterol dipole moieties and water dipoles. This compensatory effect is less effective when lipids lack carbonyl groups (CO). When monolayers are composed by unsaturated PCs the Chol compensation is found at higher concentrations of CGA due to the direct interaction between CGA and Chol. These results suggest that cholesterol modulates the interaction and distribution of CGA in the lipid membrane, which may have implications for its biological activity.
Collapse
Affiliation(s)
- Jimena Del P Cejas
- Applied Biophysics and Food Research Center (Centro de Investigaciones en Biofísica Aplicada y Alimentos, CIBAAL, National University of Santiago del Estero and CONICET), Laboratory of Biointerphases and Biomimetic Systems, RN 9 - Km 1125, 4206, Santiago del Estero, Argentina
| | - Antonio S Rosa
- Applied Biophysics and Food Research Center (Centro de Investigaciones en Biofísica Aplicada y Alimentos, CIBAAL, National University of Santiago del Estero and CONICET), Laboratory of Biointerphases and Biomimetic Systems, RN 9 - Km 1125, 4206, Santiago del Estero, Argentina
| | - Agustín N González Paz
- Applied Biophysics and Food Research Center (Centro de Investigaciones en Biofísica Aplicada y Alimentos, CIBAAL, National University of Santiago del Estero and CONICET), Laboratory of Biointerphases and Biomimetic Systems, RN 9 - Km 1125, 4206, Santiago del Estero, Argentina
| | - Edgardo A Disalvo
- Applied Biophysics and Food Research Center (Centro de Investigaciones en Biofísica Aplicada y Alimentos, CIBAAL, National University of Santiago del Estero and CONICET), Laboratory of Biointerphases and Biomimetic Systems, RN 9 - Km 1125, 4206, Santiago del Estero, Argentina
| | - María de Los A Frías
- Applied Biophysics and Food Research Center (Centro de Investigaciones en Biofísica Aplicada y Alimentos, CIBAAL, National University of Santiago del Estero and CONICET), Laboratory of Biointerphases and Biomimetic Systems, RN 9 - Km 1125, 4206, Santiago del Estero, Argentina.
| |
Collapse
|
4
|
Kovács D, Gay AS, Debayle D, Abélanet S, Patel A, Mesmin B, Luton F, Antonny B. Lipid exchange at ER-trans-Golgi contact sites governs polarized cargo sorting. J Cell Biol 2024; 223:e202307051. [PMID: 37991810 PMCID: PMC10664280 DOI: 10.1083/jcb.202307051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 10/18/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023] Open
Abstract
Oxysterol binding protein (OSBP) extracts cholesterol from the ER to deliver it to the TGN via counter exchange and subsequent hydrolysis of the phosphoinositide PI(4)P. Here, we show that this pathway is essential in polarized epithelial cells where it contributes not only to the proper subcellular distribution of cholesterol but also to the trans-Golgi sorting and trafficking of numerous plasma membrane cargo proteins with apical or basolateral localization. Reducing the expression of OSBP, blocking its activity, or inhibiting a PI4Kinase that fuels OSBP with PI(4)P abolishes the epithelial phenotype. Waves of cargo enrichment in the TGN in phase with OSBP and PI(4)P dynamics suggest that OSBP promotes the formation of lipid gradients along the TGN, which helps cargo sorting. During their transient passage through the trans-Golgi, polarized plasma membrane proteins get close to OSBP but fail to be sorted when OSBP is silenced. Thus, OSBP lipid exchange activity is decisive for polarized cargo sorting and distribution in epithelial cells.
Collapse
Affiliation(s)
- Dávid Kovács
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Anne-Sophie Gay
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Delphine Debayle
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Sophie Abélanet
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Amanda Patel
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Bruno Mesmin
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Frédéric Luton
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| | - Bruno Antonny
- Université Côte d’Azur and CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Valbonne, France
| |
Collapse
|
5
|
Kwan HLR, Chan ZCK, Bi X, Kutkowska J, Prószyński TJ, Chan CB, Lee CW. Nerve-independent formation of membrane infoldings at topologically complex postsynaptic apparatus by caveolin-3. SCIENCE ADVANCES 2023; 9:eadg0183. [PMID: 37327338 PMCID: PMC10275590 DOI: 10.1126/sciadv.adg0183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/12/2023] [Indexed: 06/18/2023]
Abstract
Junctional folds are unique membrane specializations developed progressively during the postnatal maturation of vertebrate neuromuscular junctions (NMJs), but how they are formed remains elusive. Previous studies suggested that topologically complex acetylcholine receptor (AChR) clusters in muscle cultures undergo a series of transformations, resembling the postnatal maturation of NMJs in vivo. We first demonstrated the presence of membrane infoldings at AChR clusters in cultured muscles. Live-cell super-resolution imaging further revealed that AChRs are gradually redistributed to the crest regions and spatially segregated from acetylcholinesterase along the elongating membrane infoldings over time. Mechanistically, lipid raft disruption or caveolin-3 knockdown not only inhibits membrane infolding formation at aneural AChR clusters and delays agrin-induced AChR clustering in vitro but also affects junctional fold development at NMJs in vivo. Collectively, this study demonstrated the progressive development of membrane infoldings via nerve-independent, caveolin-3-dependent mechanisms and identified their roles in AChR trafficking and redistribution during the structural maturation of NMJs.
Collapse
Affiliation(s)
- Hui-Lam Rachel Kwan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Zora Chui-Kuen Chan
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Xinyi Bi
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Justyna Kutkowska
- Łukasiewicz Research Network – PORT Polish Center for Technology Development, Wrocław, Poland
| | - Tomasz J. Prószyński
- Łukasiewicz Research Network – PORT Polish Center for Technology Development, Wrocław, Poland
| | - Chi Bun Chan
- School of Biological Sciences, Faculty of Science, The University of Hong Kong, Hong Kong, China
| | - Chi Wai Lee
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
- Golden Meditech Centre for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong, China
| |
Collapse
|
6
|
Rosenhouse-Dantsker A, Gazgalis D, Logothetis DE. PI(4,5)P 2 and Cholesterol: Synthesis, Regulation, and Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:3-59. [PMID: 36988876 DOI: 10.1007/978-3-031-21547-6_1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) is the most abundant membrane phosphoinositide and cholesterol is an essential component of the plasma membrane (PM). Both lipids play key roles in a variety of cellular functions including as signaling molecules and major regulators of protein function. This chapter provides an overview of these two important lipids. Starting from a brief description of their structure, synthesis, and regulation, the chapter continues to describe the primary functions and signaling processes in which PI(4,5)P2 and cholesterol are involved. While PI(4,5)P2 and cholesterol can act independently, they often act in concert or affect each other's impact. The chapters in this volume on "Cholesterol and PI(4,5)P2 in Vital Biological Functions: From Coexistence to Crosstalk" focus on the emerging relationship between cholesterol and PI(4,5)P2 in a variety of biological systems and processes. In this chapter, the next section provides examples from the ion channel field demonstrating that PI(4,5)P2 and cholesterol can act via common mechanisms. The chapter ends with a discussion of future directions.
Collapse
Affiliation(s)
| | - Dimitris Gazgalis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| | - Diomedes E Logothetis
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Bouvé College of Health Sciences, Northeastern University, Boston, MA, USA
| |
Collapse
|
7
|
Shishioh N, Kiryu-Seo S, Abe-Dohmae S, Yokoyama S, Kiyama H. Expression of ATP-binding cassette transporter A1 is induced by nerve injury and its deficiency affects neurite tip morphology and elongation in cultured neurons. J Chem Neuroanat 2022; 125:102164. [PMID: 36122678 DOI: 10.1016/j.jchemneu.2022.102164] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/12/2022] [Accepted: 09/15/2022] [Indexed: 11/28/2022]
Abstract
Axonal regeneration requires changes in the lipid dynamics of the axon membrane for growth and extension. Here, we examined the expression of genes associated with lipid transport after nerve injury. The expression of ATP-binding cassette transporter-A1 (ABCA1), which participates in the transport of cholesterol from the plasma membrane, was markedly upregulated in motor and sensory neurons after nerve injury. Stimulation of PC12 cells with the nerve growth factor induced neurite extension and ABCA1 expression predominantly in regions proximal to the neurite tip. To clarify the functional role of ABCA1 in neurite elongation, we examined the morphology of neurons cultured from conditionally-injured dorsal root ganglia from ABCA1-deficient mice. We found a significant increase in neurite branch formation in these neurons. In addition, the neurite tips of ABCA1-deficient neurons appeared excessively ruffled, and the direction of neurite elongation was unsteady. In contrast, the neurite tips of wild-type neurons were not excessively ruffled, and the neurites elongated rapidly in a stable directionally-oriented manner. Together, these findings suggest that ABCA1 plays an important role in regulating the membrane lipid composition of injured neurons and in axonal regeneration following nerve injury.
Collapse
Affiliation(s)
- Nobue Shishioh
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan; Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga 526-0829, Japan
| | - Sumiko Kiryu-Seo
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Sumiko Abe-Dohmae
- Food and Nutritional Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Shinji Yokoyama
- Food and Nutritional Sciences, Chubu University, 1200 Matsumoto-cho, Kasugai, Aichi 487-8501, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy & Neuroscience, Nagoya University, Graduate School of Medicine, Nagoya 466-8550, Japan.
| |
Collapse
|
8
|
Li YJ, Chen CY, Yang JH, Chiu YF. Modulating cholesterol-rich lipid rafts to disrupt influenza A virus infection. Front Immunol 2022; 13:982264. [PMID: 36177026 PMCID: PMC9513517 DOI: 10.3389/fimmu.2022.982264] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/15/2022] [Indexed: 11/13/2022] Open
Abstract
Influenza A virus (IAV) is widely disseminated across different species and can cause recurrent epidemics and severe pandemics in humans. During infection, IAV attaches to receptors that are predominantly located in cell membrane regions known as lipid rafts, which are highly enriched in cholesterol and sphingolipids. Following IAV entry into the host cell, uncoating, transcription, and replication of the viral genome occur, after which newly synthesized viral proteins and genomes are delivered to lipid rafts for assembly prior to viral budding from the cell. Moreover, during budding, IAV acquires an envelope with embedded cholesterol from the host cell membrane, and it is known that decreased cholesterol levels on IAV virions reduce infectivity. Statins are commonly used to inhibit cholesterol synthesis for preventing cardiovascular diseases, and several studies have investigated whether such inhibition can block IAV infection and propagation, as well as modulate the host immune response to IAV. Taken together, current research suggests that there may be a role for statins in countering IAV infections and modulating the host immune response to prevent or mitigate cytokine storms, and further investigation into this is warranted.
Collapse
Affiliation(s)
- Yu-Jyun Li
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Yuan Chen
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Jeng-How Yang
- Division of Infectious Diseases, Department of Medicine, Chang Gung Memorial Hospital, New Taipei, Taiwan
| | - Ya-Fang Chiu
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Research Center for Emerging Viral Infections, Chang Gung University, Taoyuan, Taiwan
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan
| |
Collapse
|
9
|
Hydroxypropyl-beta-cyclodextrin (HP-BCD) inhibits SARS-CoV-2 replication and virus-induced inflammatory cytokines. Antiviral Res 2022; 205:105373. [PMID: 35798224 PMCID: PMC9250893 DOI: 10.1016/j.antiviral.2022.105373] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 11/22/2022]
Abstract
COVID-19 is marked by extensive damage to the respiratory system, often accompanied by systemic manifestations, due to both viral cytopathic effects and hyperinflammatory syndrome. Therefore, the development of new therapeutic strategies or drug repurposing aiming to control virus replication and inflammation are required to mitigate the impact of the disease. Hydroxypropyl-beta-cyclodextrin (HP-BCD) is a cholesterol-sequestering agent with antiviral activity that has been demonstrated against enveloped viruses in in vitro and in vivo experimental models. We also demonstrated that HP-BCD has an immunomodulatory effect, inhibiting the production of selected proinflammatory cytokines induced by microbial products. Importantly, this drug has been used in humans for decades as an excipient in drug delivery systems and as a therapeutic agent in the treatment of Niemann pick C disease. The safety profile for this compound is well established. Here, we investigated whether HP-BCD would affect SARS-CoV-2 replication and virus-induced inflammatory response, using established cell lines and primary human cells. Treating virus or cells with HP-BCD significantly inhibited SARS-CoV-2 replication with a high selective index. A broad activity against distinct SARS-CoV-2 variants was evidenced by a remarkable reduction in the release of infectious particles. The drug did not alter ACE2 surface expression, but affected cholesterol accumulation into intracellular replication complexes, lowering virus RNA and protein levels, and reducing virus-induced cytopathic effects. Virus replication was also impaired by HP-BCD in Calu-3 pulmonary cell line and human primary monocytes, in which not only the virus, but also the production of proinflammatory cytokines were significantly inhibited. Given the pathophysiology of COVID-19 disease, these data indicate that the use HP-BCD, which inhibits both SARS-CoV2 replication and production of proinflammatory cytokines, as a potential COVID-19 therapeutic warrants further investigation.
Collapse
|
10
|
Rudiansyah M, Jasim SA, Mohammad Pour ZG, Athar SS, Jeda AS, Doewes RI, Jalil AT, Bokov DO, Mustafa YF, Noroozbeygi M, Karampoor S, Mirzaei R. Coronavirus disease 2019 (COVID-19) update: From metabolic reprogramming to immunometabolism. J Med Virol 2022; 94:4611-4627. [PMID: 35689351 PMCID: PMC9350347 DOI: 10.1002/jmv.27929] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 04/22/2022] [Accepted: 06/09/2022] [Indexed: 12/15/2022]
Abstract
The field of immunometabolism investigates and describes the effects of metabolic rewiring in immune cells throughout activation and the fates of these cells. Recently, it has been appreciated that immunometabolism plays an essential role in the progression of viral infections, cancer, and autoimmune diseases. Regarding COVID‐19, the aberrant immune response underlying the progression of diseases establishes two major respiratory pathologies, including acute respiratory distress syndrome (ARDS) or pneumonia‐induced acute lung injury (ALI). Both innate and adaptive immunity (T cell‐based) were impaired in the course of severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) infection. Current findings have deciphered that macrophages (innate immune cells) are involved in the inflammatory response seen in COVID‐19. It has been demonstrated that immune system cells can change metabolic reprogramming in some conditions, including autoimmune diseases, cancer, and infectious disease, including COVID‐19. The growing findings on metabolic reprogramming in COVID‐19 allow an exploration of metabolites with immunomodulatory properties as future therapies to combat this hyperinflammatory response. The elucidation of the exact role and mechanism underlying this metabolic reprograming in immune cells could help apply more precise approaches to initial diagnosis, prognosis, and in‐hospital therapy. This report discusses the latest findings from COVID‐19 on host metabolic reprogramming and immunometabolic responses.
Collapse
Affiliation(s)
- Mohammad Rudiansyah
- Division of Nephrology & Hypertension, Department of Internal Medicine, Faculty of Medicine, Universitas Lambung Mangkurat/Ulin Hospital, Banjarmasin, Indonesia
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, Al-Maarif University College, Al-Anbar-Ramadi, Iraq
| | | | - Sara Sohrabi Athar
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran.,Department of Human Nutrition, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Salimi Jeda
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rumi Iqbal Doewes
- Faculty of Sport, Universitas Sebelas Maret, Kentingan, Surakarta, Indonesia
| | - Abduladheem Turki Jalil
- Medical Laboratories Techniques Department, Al-Mustaqbal University College, Babylon, Hilla, Iraq
| | - D O Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Mina Noroozbeygi
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sajad Karampoor
- Department of Virology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.,Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Mirzaei
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.,Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
11
|
The PKD-Dependent Biogenesis of TGN-to-Plasma Membrane Transport Carriers. Cells 2021; 10:cells10071618. [PMID: 34203456 PMCID: PMC8303525 DOI: 10.3390/cells10071618] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/14/2021] [Accepted: 06/24/2021] [Indexed: 01/30/2023] Open
Abstract
Membrane trafficking is essential for processing and transport of proteins and lipids and to establish cell compartmentation and tissue organization. Cells respond to their needs and control the quantity and quality of protein secretion accordingly. In this review, we focus on a particular membrane trafficking route from the trans-Golgi network (TGN) to the cell surface: protein kinase D (PKD)-dependent pathway for constitutive secretion mediated by carriers of the TGN to the cell surface (CARTS). Recent findings highlight the importance of lipid signaling by organelle membrane contact sites (MCSs) in this pathway. Finally, we discuss our current understanding of multiple signaling pathways for membrane trafficking regulation mediated by PKD, G protein-coupled receptors (GPCRs), growth factors, metabolites, and mechanosensors.
Collapse
|
12
|
Ganesh GV, Mohanram RK. Metabolic reprogramming and immune regulation in viral diseases. Rev Med Virol 2021; 32:e2268. [PMID: 34176174 DOI: 10.1002/rmv.2268] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 06/02/2021] [Accepted: 06/10/2021] [Indexed: 12/11/2022]
Abstract
The recent outbreak and transmission of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) worldwide and the ensuing coronavirus disease 2019 (COVID-19) pandemic has left us scrambling for ways to contain the disease and develop vaccines that are safe and effective. Equally important, understanding the impact of the virus on the host system in convalescent patients, healthy otherwise or with co-morbidities, is expected to aid in developing effective strategies in the management of patients afflicted with the disease. Viruses possess the uncanny ability to redirect host metabolism to serve their needs and also limit host immune response to ensure their survival. An ever-increasingly powerful approach uses metabolomics to uncover diverse molecular signatures that influence a wide array of host signalling networks in different viral infections. This would also help integrate experimental findings from individual studies to yield robust evidence. In addition, unravelling the molecular mechanisms harnessed by both viruses and tumours in their host metabolism will help broaden the repertoire of therapeutic tools available to combat viral disease.
Collapse
Affiliation(s)
- Goutham V Ganesh
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| | - Ramkumar K Mohanram
- Life Science Division, SRM Research Institute and Department of Biotechnology, School of Bioengineering, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
13
|
Wakana Y, Hayashi K, Nemoto T, Watanabe C, Taoka M, Angulo-Capel J, Garcia-Parajo MF, Kumata H, Umemura T, Inoue H, Arasaki K, Campelo F, Tagaya M. The ER cholesterol sensor SCAP promotes CARTS biogenesis at ER-Golgi membrane contact sites. J Cell Biol 2021; 220:211521. [PMID: 33156328 PMCID: PMC7654440 DOI: 10.1083/jcb.202002150] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/15/2020] [Accepted: 10/12/2020] [Indexed: 12/13/2022] Open
Abstract
In response to cholesterol deprivation, SCAP escorts SREBP transcription factors from the endoplasmic reticulum to the Golgi complex for their proteolytic activation, leading to gene expression for cholesterol synthesis and uptake. Here, we show that in cholesterol-fed cells, ER-localized SCAP interacts through Sac1 phosphatidylinositol 4-phosphate (PI4P) phosphatase with a VAP-OSBP complex, which mediates counter-transport of ER cholesterol and Golgi PI4P at ER-Golgi membrane contact sites (MCSs). SCAP knockdown inhibited the turnover of PI4P, perhaps due to a cholesterol transport defect, and altered the subcellular distribution of the VAP-OSBP complex. As in the case of perturbation of lipid transfer complexes at ER-Golgi MCSs, SCAP knockdown inhibited the biogenesis of the trans-Golgi network-derived transport carriers CARTS, which was reversed by expression of wild-type SCAP or a Golgi transport-defective mutant, but not of cholesterol sensing-defective mutants. Altogether, our findings reveal a new role for SCAP under cholesterol-fed conditions in the facilitation of CARTS biogenesis via ER-Golgi MCSs, depending on the ER cholesterol.
Collapse
Affiliation(s)
- Yuichi Wakana
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Kaito Hayashi
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Takumi Nemoto
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Chiaki Watanabe
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Masato Taoka
- Faculty of Science, Department of Chemistry, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Jessica Angulo-Capel
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria F Garcia-Parajo
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona, Spain.,Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Hidetoshi Kumata
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Tomonari Umemura
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Hiroki Inoue
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Kohei Arasaki
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Felix Campelo
- Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Mitsuo Tagaya
- School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| |
Collapse
|
14
|
Cortada E, Serradesanferm R, Brugada R, Verges M. The voltage-gated sodium channel β2 subunit associates with lipid rafts by S-palmitoylation. J Cell Sci 2021; 134:jcs.252189. [PMID: 33602743 DOI: 10.1242/jcs.252189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/08/2021] [Indexed: 12/19/2022] Open
Abstract
The voltage-gated sodium channel is critical for cardiomyocyte function. It consists of a protein complex comprising a pore-forming α subunit and associated β subunits. In polarized Madin-Darby canine kidney cells, we show evidence by acyl-biotin exchange that β2 is S-acylated at Cys-182. Interestingly, we found that palmitoylation increases β2 association with detergent-resistant membranes. β2 localizes exclusively to the apical surface. However, depletion of plasma membrane cholesterol, or blocking intracellular cholesterol transport, caused mislocalization of β2, as well as of the non-palmitoylable C182S mutant, to the basolateral domain. Apical β2 did not undergo endocytosis and displayed limited diffusion within the plane of the membrane; such behavior suggests that, at least in part, it is cytoskeleton anchored. Upon acute cholesterol depletion, its mobility was greatly reduced, and a slight reduction was also measured as a result of lack of palmitoylation, supporting β2 association with cholesterol-rich lipid rafts. Indeed, lipid raft labeling confirmed a partial overlap with apical β2. Although β2 palmitoylation was not required to promote surface localization of the α subunit, our data suggest that it is likely implicated in lipid raft association and the polarized localization of β2.
Collapse
Affiliation(s)
- Eric Cortada
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Prov. Girona, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 17190 Salt, Prov. Girona, Spain
| | - Robert Serradesanferm
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Prov. Girona, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 17190 Salt, Prov. Girona, Spain
| | - Ramon Brugada
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Prov. Girona, Spain.,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 17190 Salt, Prov. Girona, Spain.,Medical Sciences Department, University of Girona Medical School, 17071 Girona, Spain.,Cardiology Department, Hospital Josep Trueta - University of Girona Medical School, 17007 Girona, Spain
| | - Marcel Verges
- Cardiovascular Genetics Group, Girona Biomedical Research Institute (IDIBGI), 17190 Salt, Prov. Girona, Spain .,Biomedical Research Networking Center on Cardiovascular Diseases (CIBERCV), 17190 Salt, Prov. Girona, Spain.,Medical Sciences Department, University of Girona Medical School, 17071 Girona, Spain
| |
Collapse
|
15
|
Live-cell monitoring of protein localization to membrane rafts using protein-fragment complementation. Biosci Rep 2021; 40:221616. [PMID: 31850494 PMCID: PMC6944658 DOI: 10.1042/bsr20191290] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
The plasma membrane consists of a variety of discrete domains differing from the surrounding membrane in composition and properties. Selective partitioning of protein to these microdomains is essential for membrane functioning and integrity. Studying the nanoscale size and dynamic nature of the membrane microdomains requires advanced imaging approaches with a high spatiotemporal resolution and, consequently, expensive and specialized equipment, unavailable for most researchers and unsuited for large-scale studies. Thus, understanding of protein partitioning to the membrane microdomains in health and disease is still hampered by the lack of inexpensive live-cell approaches with an appropriate spatial resolution. Here, we have developed a novel approach based on Gaussia princeps luciferase protein-fragment complementation assay to quantitively investigate protein partitioning to cholesterol and sphingomyelin-rich domains, sometimes called ‘lipid rafts’, in intact living cells with a high-spatial resolution. In the assay, the reporter construct, carrying one half of the luciferase protein, is targeted to lipid microdomains through the fused acetylation motif from Src-family kinase Fyn. A protein of interest carries the second half of the luciferase protein. Together, this serves as a reversible real-time sensor of raft recruitment for the studied protein. We demonstrated that the assay can efficiently detect the dynamic alterations in raft localization of two disease-associated proteins: Akt and APP. Importantly, this method can be used in high-throughput screenings and other large-scale studies in living cells. This inexpensive, and easy to implement raft localization assay will benefit all researchers interested in protein partitioning in rafts.
Collapse
|
16
|
Omi J, Watanabe-Takahashi M, Igai K, Shimizu E, Tseng CY, Miyasaka T, Waku T, Hama S, Nakanishi R, Goto Y, Nishino Y, Miyazawa A, Natori Y, Yamashita M, Nishikawa K. The inducible amphisome isolates viral hemagglutinin and defends against influenza A virus infection. Nat Commun 2020; 11:162. [PMID: 31919357 PMCID: PMC6952414 DOI: 10.1038/s41467-019-13974-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 12/10/2019] [Indexed: 12/31/2022] Open
Abstract
The emergence of drug-resistant influenza type A viruses (IAVs) necessitates the development of novel anti-IAV agents. Here, we target the IAV hemagglutinin (HA) protein using multivalent peptide library screens and identify PVF-tet, a peptide-based HA inhibitor. PVF-tet inhibits IAV cytopathicity and propagation in cells by binding to newly synthesized HA, rather than to the HA of the parental virus, thus inducing the accumulation of HA within a unique structure, the inducible amphisome, whose production from the autophagosome is accelerated by PVF-tet. The amphisome is also produced in response to IAV infection in the absence of PVF-tet by cells overexpressing ABC transporter subfamily A3, which plays an essential role in the maturation of multivesicular endosomes into the lamellar body, a lipid-sorting organelle. Our results show that the inducible amphisomes can function as a type of organelle-based anti-viral machinery by sequestering HA. PVF-tet efficiently rescues mice from the lethality of IAV infection.
Collapse
Affiliation(s)
- Jumpei Omi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Miho Watanabe-Takahashi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Katsura Igai
- Department of International Health, Institute of Tropical Medicine, Nagasaki University, Nagasaki, 8528523, Japan
| | - Eiko Shimizu
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Ching-Yi Tseng
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Tomohiro Miyasaka
- Department of Neuropathology, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Tsuyoshi Waku
- Department of Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Shinichiro Hama
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Rieka Nakanishi
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Yuki Goto
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan
| | - Yuri Nishino
- Graduate School of Life Science, University of Hyogo, Hyogo, 6781297, Japan
| | - Atsuo Miyazawa
- Graduate School of Life Science, University of Hyogo, Hyogo, 6781297, Japan
| | - Yasuhiro Natori
- Department of Health Chemistry, School of Pharmacy, Iwate Medical University, Iwate, 0208505, Japan
| | - Makoto Yamashita
- Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, 1088639, Japan
| | - Kiyotaka Nishikawa
- Department of Molecular Life Sciences, Graduate School of Life and Medical Sciences, Doshisha University, Kyoto, 6100394, Japan.
| |
Collapse
|
17
|
Vidal M. Exosomes: Revisiting their role as "garbage bags". Traffic 2019; 20:815-828. [PMID: 31418976 DOI: 10.1111/tra.12687] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 08/14/2019] [Indexed: 12/17/2022]
Abstract
In recent years, the term "extracellular vesicle" (EV) has been used to define different types of vesicles released by various cells. It includes plasma membrane-derived vesicles (ectosomes/microvesicles) and endosome-derived vesicles (exosomes). Although it remains difficult to evaluate the compartment of origin of the two kinds of vesicles once released, it is critical to discriminate these vesicles because their mode of biogenesis is probably directly related to their physiologic function and/or to the physio-pathologic state of the producing cell. The purpose of this review is to specifically consider exosome secretion and its consequences in terms of a material loss for producing cells, rather than on the effects of exosomes once they are taken up by recipient cells. I especially describe one putative basic function of exosomes, that is, to convey material out of cells for off-site degradation by recipient cells. As illustrated by some examples, these components could be evacuated from cells for various reasons, for example, to promote "differentiation" or enhance homeostatic responses. This basic function might explain why so many diseases have made use of the exosomal pathway during pathogenesis.
Collapse
Affiliation(s)
- Michel Vidal
- LPHI - Université de Montpellier, CNRS, Montpellier, France
| |
Collapse
|
18
|
Lebreton S, Paladino S, Zurzolo C. Clustering in the Golgi apparatus governs sorting and function of GPI‐APs in polarized epithelial cells. FEBS Lett 2019; 593:2351-2365. [DOI: 10.1002/1873-3468.13573] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 01/25/2023]
Affiliation(s)
- Stéphanie Lebreton
- Unité de Trafic Membranaire et Pathogénèse Institut Pasteur Paris France
| | - Simona Paladino
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche Università degli Studi di Napoli Federico II Naples Italy
| | - Chiara Zurzolo
- Unité de Trafic Membranaire et Pathogénèse Institut Pasteur Paris France
| |
Collapse
|
19
|
Sato R, Okura T, Kawahara M, Takizawa N, Momose F, Morikawa Y. Apical Trafficking Pathways of Influenza A Virus HA and NA via Rab17- and Rab23-Positive Compartments. Front Microbiol 2019; 10:1857. [PMID: 31456775 PMCID: PMC6700264 DOI: 10.3389/fmicb.2019.01857] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 07/29/2019] [Indexed: 11/13/2022] Open
Abstract
The envelope proteins of influenza A virus, hemagglutinin (HA) and neuraminidase (NA), play critical roles in viral entry to host cells and release from the cells, respectively. After protein synthesis, they are transported from the trans-Golgi network (TGN) to the apical plasma membrane (PM) and assembled into virus particles. However, the post-TGN transport pathways of HA and NA have not been clarified. Temporal study by confocal microscopy revealed that HA and NA colocalized soon after their synthesis, and relocated together from the TGN to the upper side of the cell. Using the Rab family protein, we investigated the post-TGN transport pathways of HA and NA. HA partially colocalized with AcGFP-Rab15, Rab17, and Rab23, but rarely with AcGFP-Rab11. When analyzed in cells stably expressing AcGFP-Rab, HA/NA colocalized with Rab15 and Rab17, markers of apical sorting and recycling endosomes, and later colocalized with Rab23, which distributes to the apical PM and endocytic vesicles. Overexpression of the dominant-negative (DN) mutants of Rab15 and Rab17, but not Rab23, significantly delayed HA transport to the PM. However, Rab23DN impaired cell surface expression of HA. Live-cell imaging revealed that NA moved rapidly with Rab17 but not with Rab15. NA also moved with Rab23 in the cytoplasm, but this motion was confined at the upper side of the cell. A fraction of HA was localized to Rab17 and Rab23 double-positive vesicles in the cytoplasm. Coimmunoprecipitation indicated that HA was associated with Rab17 and Rab23 in lipid raft fractions. When cholesterol was depleted by methyl-β-cyclodextrin treatment, the motion of NA and Rab17 signals ceased. These results suggest that HA and NA are incorporated into lipid raft microdomains and are cotransported to the PM by Rab17-positive and followed by Rab23-positive vesicles.
Collapse
Affiliation(s)
- Ryota Sato
- Graduate School for Infection Control, Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| | - Takashi Okura
- Graduate School for Infection Control, Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| | - Madoka Kawahara
- Graduate School for Infection Control, Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| | - Naoki Takizawa
- Laboratory of Basic Biology, Institute of Microbial Chemistry, Tokyo, Japan
| | - Fumitaka Momose
- Graduate School for Infection Control, Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| | - Yuko Morikawa
- Graduate School for Infection Control, Kitasato Institute for Life Sciences, Kitasato University, Tokyo, Japan
| |
Collapse
|
20
|
Goñi FM. "Rafts": A nickname for putative transient nanodomains. Chem Phys Lipids 2018; 218:34-39. [PMID: 30476484 DOI: 10.1016/j.chemphyslip.2018.11.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/29/2018] [Accepted: 11/21/2018] [Indexed: 01/24/2023]
Abstract
The membrane raft hypothesis, proposed in 1997 by Simons and Ikonen, has played a paradoxical role in the history of biomembrane research. While it has generated a large amount of investigations, thus helping to increase our understanding of membranes, the object that gives name to the hypothesis, i.e. the raft itself, has been and still is an object of controversy, in which its very reality is often questioned. In this contribution I review the history of the hypothesis and its reception by membrane biologists, and summarize some of the valuable physico-chemical results that have been obtained while testing the raft hypothesis. To save a useful concept from its many misuses I propose that the expression "(transient) nanodomains" be employed instead of "rafts".
Collapse
Affiliation(s)
- Félix M Goñi
- Instituto Biofisika (CSIC, UPV/EHU), Departamento de Bioquímica, Universidad del País Vasco, 48940 Leioa, Spain.
| |
Collapse
|
21
|
Bieberich E. Sphingolipids and lipid rafts: Novel concepts and methods of analysis. Chem Phys Lipids 2018; 216:114-131. [PMID: 30194926 PMCID: PMC6196108 DOI: 10.1016/j.chemphyslip.2018.08.003] [Citation(s) in RCA: 146] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/20/2018] [Accepted: 08/25/2018] [Indexed: 12/12/2022]
Abstract
About twenty years ago, the functional lipid raft model of the plasma membrane was published. It took into account decades of research showing that cellular membranes are not just homogenous mixtures of lipids and proteins. Lateral anisotropy leads to assembly of membrane domains with specific lipid and protein composition regulating vesicular traffic, cell polarity, and cell signaling pathways in a plethora of biological processes. However, what appeared to be a clearly defined entity of clustered raft lipids and proteins became increasingly fluid over the years, and many of the fundamental questions about biogenesis and structure of lipid rafts remained unanswered. Experimental obstacles in visualizing lipids and their interactions hampered progress in understanding just how big rafts are, where and when they are formed, and with which proteins raft lipids interact. In recent years, we have begun to answer some of these questions and sphingolipids may take center stage in re-defining the meaning and functional significance of lipid rafts. In addition to the archetypical cholesterol-sphingomyelin raft with liquid ordered (Lo) phase and the liquid-disordered (Ld) non-raft regions of cellular membranes, a third type of microdomains termed ceramide-rich platforms (CRPs) with gel-like structure has been identified. CRPs are "ceramide rafts" that may offer some fresh view on the membrane mesostructure and answer several critical questions for our understanding of lipid rafts.
Collapse
Affiliation(s)
- Erhard Bieberich
- Department of Physiology at the University of Kentucky, Lexington, KY, United States.
| |
Collapse
|
22
|
Goto A, Charman M, Ridgway ND. Protein kinase D1 and oxysterol-binding protein form a regulatory complex independent of phosphorylation. Traffic 2018; 19:854-866. [DOI: 10.1111/tra.12609] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/08/2018] [Accepted: 08/08/2018] [Indexed: 12/25/2022]
Affiliation(s)
- Asako Goto
- Department of Pediatrics; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
- Department of Molecular Biology and Molecular Biology; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
| | - Mark Charman
- Department of Pediatrics; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
- Department of Molecular Biology and Molecular Biology; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
| | - Neale D. Ridgway
- Department of Pediatrics; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
- Department of Molecular Biology and Molecular Biology; Atlantic Research Centre, Dalhousie University; Halifax Nova Scotia Canada
| |
Collapse
|
23
|
Pombo JP, Sanyal S. Perturbation of Intracellular Cholesterol and Fatty Acid Homeostasis During Flavivirus Infections. Front Immunol 2018; 9:1276. [PMID: 29915602 PMCID: PMC5994796 DOI: 10.3389/fimmu.2018.01276] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 05/22/2018] [Indexed: 12/11/2022] Open
Abstract
Cellular lipid homeostasis is maintained through an intricately linked array of anabolic and catabolic pathways. Upon flavivirus infections, these are significantly altered: on the one hand, these viruses can co-opt lipid metabolic pathways to generate ATP to facilitate replication, or to synthesize membrane components to generate replication sites; on the other hand, more recent evidence suggests counter strategies employed by host cells, which actively modulate several of these networks in response to infection, enhancing interferon signaling by doing so, and thus creating an antiviral environment. In this review, we discuss recent data on mechanisms of alteration of lipid metabolic pathways during infection by flaviviruses, with a focus on cholesterol and fatty acid biosynthesis, which can be manipulated by the invading viruses to support replication, but can also be modulated by the host immune system itself, as a means to fight infection.
Collapse
Affiliation(s)
- Joao Palma Pombo
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong
| | - Sumana Sanyal
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong, Hong Kong.,School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
24
|
Gélinas JF, Davies LA, Gill DR, Hyde SC. Assessment of selected media supplements to improve F/HN lentiviral vector production yields. Sci Rep 2017; 7:10198. [PMID: 28860488 PMCID: PMC5579034 DOI: 10.1038/s41598-017-07893-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/31/2017] [Indexed: 12/01/2022] Open
Abstract
The development of lentiviral-based therapeutics is challenged by the high cost of current Good Manufacturing Practices (cGMP) production. Lentiviruses are enveloped viruses that capture a portion of the host cell membrane during budding, which then constitutes part of the virus particle. This process might lead to lipid and protein depletion in the cell membrane and affect cell viability. Furthermore, growth in suspension also causes stresses that can affect virus production yields. To assess the impact of these issues, selected supplements (Cholesterol Lipid Concentrate, Chemically Defined Lipid Concentrate, Lipid Mixture 1, Gelatin Peptone N3, N-Acetyl-L-Cysteine and Pluronic F-68) were assayed in order to improve production yields in a transient transfection production of a Sendai virus F/HN-pseudotyped HIV-1-based third generation lentiviral vector in FreeStyle 293 (serum-free media) in suspension. None of the supplements tested had a significant positive impact on lentiviral vector yields, but small non-significant improvements could be combined to increase vector production in a cell line where other conditions have been optimised.
Collapse
Affiliation(s)
- Jean-François Gélinas
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK
| | - Lee A Davies
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK.,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK
| | - Deborah R Gill
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK.,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK
| | - Stephen C Hyde
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK. .,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK.
| |
Collapse
|
25
|
Wudiri GA, Nicola AV. Cellular Cholesterol Facilitates the Postentry Replication Cycle of Herpes Simplex Virus 1. J Virol 2017; 91:e00445-17. [PMID: 28446672 PMCID: PMC5487575 DOI: 10.1128/jvi.00445-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 04/19/2017] [Indexed: 12/24/2022] Open
Abstract
Cholesterol is an essential component of cell membranes and is required for herpes simplex virus 1 (HSV-1) entry (1-3). Treatment of HSV-1-infected Vero cells with methyl beta-cyclodextrin from 2 to 9 h postentry reduced plaque numbers. Transport of incoming viral capsids to the nuclear periphery was unaffected by the cholesterol reduction, suggesting that cell cholesterol is important for the HSV-1 replicative cycle at a stage(s) beyond entry, after the arrival of capsids at the nucleus. The synthesis and release of infectious HSV-1 and cell-to-cell spread of infection were all impaired in cholesterol-reduced cells. Propagation of HSV-1 on DHCR24-/- fibroblasts, which lack the desmosterol-to-cholesterol conversion enzyme, resulted in the generation of infectious extracellular virions (HSVdes) that lack cholesterol and likely contain desmosterol. The specific infectivities (PFU per viral genome) of HSVchol and HSVdes were similar, suggesting cholesterol and desmosterol in the HSV envelope support similar levels of infectivity. However, infected DHCR24-/- fibroblasts released ∼1 log less infectious HSVdes and ∼1.5 log fewer particles than release of cholesterol-containing particles (HSVchol) from parental fibroblasts, suggesting that the hydrocarbon tail of cholesterol facilitates viral synthesis. Together, the results suggest multiple roles for cholesterol in the HSV-1 replicative cycle.IMPORTANCE HSV-1 infections are associated with a wide range of clinical manifestations that are of public health importance. Cholesterol is a key player in the complex interaction between viral and cellular factors that allows HSV-1 to enter host cells and establish infection. Previous reports have demonstrated a role for cellular cholesterol in the entry of HSV-1 into target cells. Here, we employed both chemical treatment and cells that were genetically defined to synthesize only desmosterol to demonstrate that cholesterol is important at stages following the initial entry and transport of viral capsids to the nucleus. Viral protein expression, encapsidation of the viral genome, and the release of mature virions were impacted by the reduction of cellular cholesterol. Cholesterol was also critical for cell-to-cell spread of infection. These findings provide new insights into the cholesterol dependence of HSV-1 replication.
Collapse
Affiliation(s)
- George A Wudiri
- Department of Veterinary Microbiology and Pathology and Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| | - Anthony V Nicola
- Department of Veterinary Microbiology and Pathology and Paul G. Allen School for Global Animal Health, College of Veterinary Medicine, Washington State University, Pullman, Washington, USA
| |
Collapse
|
26
|
Kawano K, Yagi T, Fukada N, Yano Y, Matsuzaki K. Stoichiometric analysis of oligomeric states of three class-A GPCRs, chemokine-CXCR4, dopamine-D2, and prostaglandin-EP1 receptors, on living cells. J Pept Sci 2017. [PMID: 28626925 DOI: 10.1002/psc.3020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
G-protein-coupled receptors (GPCRs) form the largest family of transmembrane receptors, and their oligomerization has been suggested to be related to their functions. Despite extensive studies, their oligomeric states are highly controversial. One of the reasons is the overestimation of oligomerization by conventional methods. We recently established a stoichiometric analysis method for precisely determining the oligomeric state of membrane proteins on living cells with the combined use of the coiled-coil labeling method and a spectral imaging technique and showed that the prototypical class-A GPCR β2 -adrenergic receptor (β2 AR) did not form functional oligomers. In this study, we expanded our study to three well-studied class-A GPCRs: C-X-C chemokine receptor of stromal cell-derived factor-1α (CXCR4), dopamine receptor D2 short isotype (D2R), and prostaglandin E receptor subtype 1 (EP1R). We found that these receptors did not form constitutive homooligomers. The receptors exhibited calcium signaling upon agonist stimulation as monomers, although CXCR4 and EP1R gradually clustered after fast signaling. We conclude that homooligomerization is not necessary for the signal transductions of these four class-A GPCRs. Copyright © 2017 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Kenichi Kawano
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Tetsuya Yagi
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Nozomu Fukada
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yoshiaki Yano
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Katsumi Matsuzaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshida-Shimoadachicho, Sakyo-ku, Kyoto, 606-8501, Japan
| |
Collapse
|
27
|
Abstract
Shadoo (Sho), a member of prion protein family, has been shown to prevent embryonic lethality in Prnp0/0 mice and to be reduced in the brains of rodents with terminal prion diseases. Sho can also affect PrP structural dynamics and can increase the prion conversion into its misfolded isoform (PrPSc), which is amyloidogenic and strictly related to expression, intracellular localization and association of PrPC to lipid rafts. We reasoned that if Sho possesses a natural tendency to convert to amyloid-like forms in vitro, it should be able to exhibit “prion-like” properties, such as PK-resistance and aggregation state, also in live cells. We tested this hypothesis, by different approaches in neuronal cells, finding that Sho shows folding properties partially dependent on lipid rafts integrity whose alteration, as well as proteasomal block, regulated generation of intermediate Sho isoforms and exacerbated its misfolding. Moreover, a 18 kDa isoform of Sho, likely bearing the signal peptide, was targeted to mitochondria by interacting with the molecular chaperone TRAP1 which, in turn controlled Sho dual targeting to ER or mitochondria. Our studies contribute to understand the role of molecular chaperones and of PrP-related folding intermediates in “prion-like” conversion.
Collapse
|
28
|
Charman M, Goto A, Ridgway ND. Oxysterol-binding protein recruitment and activity at the endoplasmic reticulum-Golgi interface are independent of Sac1. Traffic 2017; 18:519-529. [DOI: 10.1111/tra.12491] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 05/01/2017] [Accepted: 05/01/2017] [Indexed: 12/26/2022]
Affiliation(s)
- Mark Charman
- The Atlantic Research Centre, Department of Pediatrics, and Biochemistry & Molecular Biology; Dalhousie University; Halifax Canada
| | - Asako Goto
- The Atlantic Research Centre, Department of Pediatrics, and Biochemistry & Molecular Biology; Dalhousie University; Halifax Canada
| | - Neale D. Ridgway
- The Atlantic Research Centre, Department of Pediatrics, and Biochemistry & Molecular Biology; Dalhousie University; Halifax Canada
| |
Collapse
|
29
|
Smith WS, Baker EJ, Holmes SE, Koster G, Hunt AN, Johnston DA, Flavell SU, Flavell DJ. Membrane cholesterol is essential for triterpenoid saponin augmentation of a saporin-based immunotoxin directed against CD19 on human lymphoma cells. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:993-1007. [PMID: 28235471 DOI: 10.1016/j.bbamem.2017.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/16/2017] [Accepted: 02/20/2017] [Indexed: 01/06/2023]
Abstract
Triterpenoid saponins from Saponinum Album (SA) exert potent lytic effects on eukaryotic cell plasma membranes and, when used at sub-lytic concentrations, significantly augment the cytotoxicity of saporin-based immunotoxins (IT). To help elucidate the mechanism(s) behind these two phenomena we investigated the role of cholesterol to both. Human Daudi lymphoma cells were lipid deprived using a combination of three different approaches. Following treatment, the total cellular lipid content was analyzed by electrospray ionization mass spectrometry (ESI-MS) and plasma membrane (PM) cholesterol content measured using the lipophilic fluorescent probe NR12S. Maximal lipid deprivation of cells resulted in a complete loss of sensitivity to lysis by SA. Similarly augmentation of the anti-CD19 immunotoxin (IT) BU12-SAPORIN by SA was lost but without a concomitant loss of intrinsic IT cytotoxicity. The lytic activity of SA was restored following incubation of lipid deprived Daudi cells with Synthecol or LDL. The augmentative effect of SA on IT cytotoxicity for Daudi cells was restored following repletion of PM cholesterol levels with LDL. NR12S fluorescence and ESI-MS analysis of cellular lipids demonstrated that restoration of SA lytic activity by Synthecol was entirely due to increased PM cholesterol levels. Restoration of cellular and PM cholesterol levels by LDL also restored the augmentative effect of SA for IT, an effect associated with repletion of PM cholesterol with minor changes in some phospholipid species. These results indicate that the lytic and IT augmentative properties of SA are cholesterol-dependent in contrast to intrinsic IT cytotoxicity that is at least partially cholesterol independent.
Collapse
Affiliation(s)
- Wendy S Smith
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - Ella J Baker
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom; Human Development and Health Academic Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Suzanne E Holmes
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - Grielof Koster
- NIHR Respiratory Biomedical Research Unit, UHS, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom; Clinical and Experimental Sciences, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Alan N Hunt
- Clinical and Experimental Sciences, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - David A Johnston
- Biomedical Imaging Unit, University of Southampton School of Medicine, Southampton General Hospital, Southampton SO16 6YD, United Kingdom
| | - Sopsamorn U Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom
| | - David J Flavell
- The Simon Flavell Leukaemia Research Laboratory, Southampton General Hospital, Southampton, Hampshire SO16 6YD, United Kingdom.
| |
Collapse
|
30
|
Hu X, Zhu M, Liang Z, Kumar D, Chen F, Zhu L, Kuang S, Xue R, Cao G, Gong C. Proteomic analysis of BmN cell lipid rafts reveals roles in Bombyx mori nucleopolyhedrovirus infection. Mol Genet Genomics 2017; 292:465-474. [DOI: 10.1007/s00438-016-1284-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 12/26/2016] [Indexed: 11/25/2022]
|
31
|
Woods PS, Doolittle LM, Rosas LE, Joseph LM, Calomeni EP, Davis IC. Lethal H1N1 influenza A virus infection alters the murine alveolar type II cell surfactant lipidome. Am J Physiol Lung Cell Mol Physiol 2016; 311:L1160-L1169. [PMID: 27836900 DOI: 10.1152/ajplung.00339.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 11/08/2016] [Indexed: 11/22/2022] Open
Abstract
Alveolar type II (ATII) epithelial cells are the primary site of influenza virus replication in the distal lung. Development of acute respiratory distress syndrome in influenza-infected mice correlates with significant alterations in ATII cell function. However, the impact of infection on ATII cell surfactant lipid metabolism has not been explored. C57BL/6 mice were inoculated intranasally with influenza A/WSN/33 (H1N1) virus (10,000 plaque-forming units/mouse) or mock-infected with virus diluent. ATII cells were isolated by a standard lung digestion protocol at 2 and 6 days postinfection. Levels of 77 surfactant lipid-related compounds of known identity in each ATII cell sample were measured by ultra-high-performance liquid chromatography-mass spectrometry. In other mice, bronchoalveolar lavage fluid was collected to measure lipid and protein content using commercial assay kits. Relative to mock-infected animals, ATII cells from influenza-infected mice contained reduced levels of major surfactant phospholipids (phosphatidylcholine, phosphatidylglycerol, and phosphatidylethanolamine) but increased levels of minor phospholipids (phosphatidylserine, phosphatidylinositol, and sphingomyelin), cholesterol, and diacylglycerol. These changes were accompanied by reductions in cytidine 5'-diphosphocholine and 5'-diphosphoethanolamine (liponucleotide precursors for ATII cell phosphatidylcholine and phosphatidylethanolamine synthesis, respectively). ATII cell lamellar bodies were ultrastructurally abnormal after infection. Changes in ATII cell phospholipids were reflected in the composition of bronchoalveolar lavage fluid, which contained reduced amounts of phosphatidylcholine and phosphatidylglycerol but increased amounts of sphingomyelin, cholesterol, and protein. Influenza infection significantly alters ATII cell surfactant lipid metabolism, which may contribute to surfactant dysfunction and development of acute respiratory distress syndrome in influenza-infected mice.
Collapse
Affiliation(s)
- Parker S Woods
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio; and
| | - Lauren M Doolittle
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio; and
| | - Lucia E Rosas
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio; and
| | - Lisa M Joseph
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio; and
| | - Edward P Calomeni
- Department of Pathology, College of Medicine, The Ohio State University, Columbus, Ohio
| | - Ian C Davis
- Department of Veterinary Biosciences, College of Veterinary Medicine, The Ohio State University, Columbus, Ohio; and
| |
Collapse
|
32
|
Van Blerkom J, Zimmermann S. Ganglioside-enriched microdomains define an oolemma that is functionally polarized with respect to fertilizability in the mouse. Reprod Biomed Online 2016; 33:458-475. [DOI: 10.1016/j.rbmo.2016.06.029] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 10/21/2022]
|
33
|
Söderholm S, Fu Y, Gaelings L, Belanov S, Yetukuri L, Berlinkov M, Cheltsov AV, Anders S, Aittokallio T, Nyman TA, Matikainen S, Kainov DE. Multi-Omics Studies towards Novel Modulators of Influenza A Virus-Host Interaction. Viruses 2016; 8:v8100269. [PMID: 27690086 PMCID: PMC5086605 DOI: 10.3390/v8100269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Revised: 09/13/2016] [Accepted: 09/22/2016] [Indexed: 12/20/2022] Open
Abstract
Human influenza A viruses (IAVs) cause global pandemics and epidemics. These viruses evolve rapidly, making current treatment options ineffective. To identify novel modulators of IAV–host interactions, we re-analyzed our recent transcriptomics, metabolomics, proteomics, phosphoproteomics, and genomics/virtual ligand screening data. We identified 713 potential modulators targeting 199 cellular and two viral proteins. Anti-influenza activity for 48 of them has been reported previously, whereas the antiviral efficacy of the 665 remains unknown. Studying anti-influenza efficacy and immuno/neuro-modulating properties of these compounds and their combinations as well as potential viral and host resistance to them may lead to the discovery of novel modulators of IAV–host interactions, which might be more effective than the currently available anti-influenza therapeutics.
Collapse
Affiliation(s)
- Sandra Söderholm
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
- Finnish Institute of Occupational Health, Helsinki 00250, Finland.
| | - Yu Fu
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Lana Gaelings
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Sergey Belanov
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Laxman Yetukuri
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Mikhail Berlinkov
- Institute of Mathematics and Computer Science, Ural Federal University, Yekaterinburg 620083, Russia.
| | - Anton V Cheltsov
- Q-Mol L.L.C. in Silico Pharmaceuticals, San Diego, CA 92037, USA.
| | - Simon Anders
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
- Department of Mathematics and Statistics, University of Turku, Turku 20014, Finland.
| | | | - Sampsa Matikainen
- Finnish Institute of Occupational Health, Helsinki 00250, Finland.
- Department of Rheumatology, Helsinki University Hospital, University of Helsinki, Helsinki 00015, Finland.
| | - Denis E Kainov
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
34
|
Nagaraj V, Kazim AS, Helgeson J, Lewold C, Barik S, Buda P, Reinbothe TM, Wennmalm S, Zhang E, Renström E. Elevated Basal Insulin Secretion in Type 2 Diabetes Caused by Reduced Plasma Membrane Cholesterol. Mol Endocrinol 2016; 30:1059-1069. [PMID: 27533789 PMCID: PMC5045496 DOI: 10.1210/me.2016-1023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Elevated basal insulin secretion under fasting conditions together with insufficient stimulated insulin release is an important hallmark of type 2 diabetes, but the mechanisms controlling basal insulin secretion remain unclear. Membrane rafts exist in pancreatic islet cells and spatially organize membrane ion channels and proteins controlling exocytosis, which may contribute to the regulation of insulin secretion. Membrane rafts (cholesterol and sphingolipid containing microdomains) were dramatically reduced in human type 2 diabetic and diabetic Goto-Kakizaki (GK) rat islets when compared with healthy islets. Oxidation of membrane cholesterol markedly reduced microdomain staining intensity in healthy human islets, but was without effect in type 2 diabetic islets. Intriguingly, oxidation of cholesterol affected glucose-stimulated insulin secretion only modestly, whereas basal insulin release was elevated. This was accompanied by increased intracellular Ca2+ spike frequency and Ca2+ influx and explained by enhanced single Ca2+ channel activity. These results suggest that the reduced presence of membrane rafts could contribute to the elevated basal insulin secretion seen in type 2 diabetes.
Collapse
Affiliation(s)
- Vini Nagaraj
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Abdulla S Kazim
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Johan Helgeson
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Clemens Lewold
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Satadal Barik
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Pawel Buda
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Thomas M Reinbothe
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Stefan Wennmalm
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Enming Zhang
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| | - Erik Renström
- Department of Clinical Sciences Malmö (V.N., A.S.K., J.H., C.L., S.B., P.B., T.M.R., E.Z., E.R.), Lund University Diabetes Centre, Lund University, SE-20502 Malmö, Sweden; and Science for Life Laboratory (S.W.), KTH Royal Institute of Technology, SE-171 77 Stockholm, Sweden
| |
Collapse
|
35
|
African horse sickness virus infects BSR cells through macropinocytosis. Virology 2016; 497:217-232. [PMID: 27497184 DOI: 10.1016/j.virol.2016.07.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 07/11/2016] [Accepted: 07/12/2016] [Indexed: 11/23/2022]
Abstract
Cellular pathways involved in cell entry by African horse sickness virus (AHSV), a member of the Orbivirus genus within the Reoviridae family, have not yet been determined. Here, we show that acidic pH is required for productive infection of BSR cells by AHSV-4, suggesting that the virus is likely internalized by an endocytic pathway. We subsequently analyzed the major endocytic routes using specific inhibitors and determined the consequences for AHSV-4 entry into BSR cells. The results indicated that virus entry is dynamin dependent, but clathrin- and lipid raft/caveolae-mediated endocytic pathways were not used by AHSV-4 to enter and infect BSR cells. Instead, binding of AHSV-4 to BSR cells stimulated uptake of a macropinocytosis-specific cargo and inhibition of Na(+)/H(+) exchangers, actin polymerization and cellular GTPases and kinases involved in macropinocytosis significantly inhibited AHSV-4 infection. Altogether, the data suggest that AHSV-4 infects BSR cells by utilizing macropinocytosis as the primary entry pathway.
Collapse
|
36
|
Pohl MO, Lanz C, Stertz S. Late stages of the influenza A virus replication cycle-a tight interplay between virus and host. J Gen Virol 2016; 97:2058-2072. [PMID: 27449792 DOI: 10.1099/jgv.0.000562] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
After successful infection and replication of its genome in the nucleus of the host cell, influenza A virus faces several challenges before newly assembled viral particles can bud off from the plasma membrane, giving rise to a new infectious virus. The viral ribonucleoprotein (vRNP) complexes need to exit from the nucleus and be transported to the virus assembly sites at the plasma membrane. Moreover, they need to be bundled to ensure the incorporation of precisely one of each of the eight viral genome segments into newly formed viral particles. Similarly, viral envelope glycoproteins and other viral structural proteins need to be targeted to virus assembly sites for viral particles to form and bud off from the plasma membrane. During all these steps influenza A virus heavily relies on a tight interplay with its host, exploiting host-cell proteins for its own purposes. In this review, we summarize current knowledge on late stages of the influenza virus replication cycle, focusing on the role of host-cell proteins involved in this process.
Collapse
Affiliation(s)
- Marie O Pohl
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Caroline Lanz
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Silke Stertz
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
37
|
Chlanda P, Zimmerberg J. Protein-lipid interactions critical to replication of the influenza A virus. FEBS Lett 2016; 590:1940-54. [PMID: 26921878 DOI: 10.1002/1873-3468.12118] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/08/2016] [Accepted: 02/21/2016] [Indexed: 12/12/2022]
Abstract
Influenza A virus (IAV) assembles on the plasma membrane where viral proteins localize to form a bud encompassing the viral genome, which ultimately pinches off to give rise to newly formed infectious virions. Upon entry, the virus faces the opposite task-fusion with the endosomal membrane and disassembly to deliver the viral genome to the cytoplasm. There are at least four influenza proteins-hemagglutinin (HA), neuraminidase (NA), matrix 1 protein (M1), and the M2 ion channel-that are known to directly interact with the cellular membrane and modify membrane curvature in order to both assemble and disassemble membrane-enveloped virions. Here, we summarize and discuss current knowledge of the interactions of lipids and membrane proteins involved in the IAV replication cycle.
Collapse
Affiliation(s)
- Petr Chlanda
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
38
|
Wilson RL, Frisz JF, Klitzing HA, Zimmerberg J, Weber PK, Kraft ML. Hemagglutinin clusters in the plasma membrane are not enriched with cholesterol and sphingolipids. Biophys J 2016; 108:1652-1659. [PMID: 25863057 DOI: 10.1016/j.bpj.2015.02.026] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 02/11/2015] [Accepted: 02/18/2015] [Indexed: 01/28/2023] Open
Abstract
The clusters of the influenza envelope protein, hemagglutinin, within the plasma membrane are hypothesized to be enriched with cholesterol and sphingolipids. Here, we directly tested this hypothesis by using high-resolution secondary ion mass spectrometry to image the distributions of antibody-labeled hemagglutinin and isotope-labeled cholesterol and sphingolipids in the plasma membranes of fibroblast cells that stably express hemagglutinin. We found that the hemagglutinin clusters were neither enriched with cholesterol nor colocalized with sphingolipid domains. Thus, hemagglutinin clustering and localization in the plasma membrane is not controlled by cohesive interactions between hemagglutinin and liquid-ordered domains enriched with cholesterol and sphingolipids, or from specific binding interactions between hemagglutinin, cholesterol, and/or the majority of sphingolipid species in the plasma membrane.
Collapse
Affiliation(s)
- Robert L Wilson
- Department of Chemistry, University of Illinois, Urbana, Illinois
| | - Jessica F Frisz
- Department of Chemistry, University of Illinois, Urbana, Illinois
| | - Haley A Klitzing
- Department of Chemistry, University of Illinois, Urbana, Illinois
| | - Joshua Zimmerberg
- Section on Cellular and Membrane Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | - Peter K Weber
- Glenn T. Seaborg Institute, Lawrence Livermore National Laboratory, Livermore, California
| | - Mary L Kraft
- Department of Chemistry, University of Illinois, Urbana, Illinois; Department of Chemical and Biomolecular Engineering, University of Illinois, Urbana, Illnois.
| |
Collapse
|
39
|
Abu-Siniyeh A, Owen DM, Benzing C, Rinkwitz S, Becker TS, Majumdar A, Gaus K. The aPKC/Par3/Par6 Polarity Complex and Membrane Order Are Functionally Interdependent in Epithelia During Vertebrate Organogenesis. Traffic 2015; 17:66-79. [PMID: 26456025 DOI: 10.1111/tra.12339] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 10/06/2015] [Accepted: 10/06/2015] [Indexed: 12/17/2022]
Abstract
The differential distribution of lipids between apical and basolateral membranes is necessary for many epithelial cell functions, but how this characteristic membrane organization is integrated within the polarity network during ductal organ development is poorly understood. Here we quantified membrane order in the gut, kidney and liver ductal epithelia in zebrafish larvae at 3-11 days post fertilization (dpf) with Laurdan 2-photon microscopy. We then applied a combination of Laurdan imaging, antisense knock-down and analysis of polarity markers to understand the relationship between membrane order and apical-basal polarity. We found a reciprocal relationship between membrane order and the cell polarity network. Reducing membrane condensation by exogenously added oxysterol or depletion of cholesterol reduced apical targeting of the polarity protein, aPKC. Conversely, using morpholino knock down in zebrafish, we found that membrane order was dependent upon the Crb3 and Par3 polarity protein expression in ductal epithelia. Hence our data suggest that the biophysical property of membrane lipid packing is a regulatory element in apical basal polarity.
Collapse
Affiliation(s)
- Ahmed Abu-Siniyeh
- School of Medical Sciences, ARC Centre for Advanced Molecular Imaging and Australian Centre for NanoMedicine, The University of New South Wales, Australia.,Present address: Department of Chemistry and Medical Analysis, Faculty of Science, Al-Balqa' Applied University, Al-Salt, 19117, Jordan
| | - Dylan M Owen
- Department of Physics and Randall Division of Cell and Molecular Biophysics, King's College London, UK
| | - Carola Benzing
- School of Medical Sciences, ARC Centre for Advanced Molecular Imaging and Australian Centre for NanoMedicine, The University of New South Wales, Australia
| | - Silke Rinkwitz
- Brain and Mind Research Institute, Sydney Medical School and Department of Health Sciences, University of Sydney, Australia
| | - Thomas S Becker
- Brain and Mind Research Institute, Sydney Medical School and Department of Health Sciences, University of Sydney, Australia
| | - Arindam Majumdar
- Department of Immunology, Genetics, and Pathology, Uppsala University, Sweden
| | - Katharina Gaus
- School of Medical Sciences, ARC Centre for Advanced Molecular Imaging and Australian Centre for NanoMedicine, The University of New South Wales, Australia
| |
Collapse
|
40
|
Huang SS, Chen CL, Huang FW, Johnson FE, Huang JS. Ethanol Enhances TGF-β Activity by Recruiting TGF-β Receptors From Intracellular Vesicles/Lipid Rafts/Caveolae to Non-Lipid Raft Microdomains. J Cell Biochem 2015; 117:860-71. [PMID: 26419316 DOI: 10.1002/jcb.25389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022]
Abstract
Regular consumption of moderate amounts of ethanol has important health benefits on atherosclerotic cardiovascular disease (ASCVD). Overindulgence can cause many diseases, particularly alcoholic liver disease (ALD). The mechanisms by which ethanol causes both beneficial and harmful effects on human health are poorly understood. Here we demonstrate that ethanol enhances TGF-β-stimulated luciferase activity with a maximum of 0.5-1% (v/v) in Mv1Lu cells stably expressing a luciferase reporter gene containing Smad2-dependent elements. In Mv1Lu cells, 0.5% ethanol increases the level of P-Smad2, a canonical TGF-β signaling sensor, by ∼ 2-3-fold. Ethanol (0.5%) increases cell-surface expression of the type II TGF-β receptor (TβR-II) by ∼ 2-3-fold from its intracellular pool, as determined by I(125) -TGF-β-cross-linking/Western blot analysis. Sucrose density gradient ultracentrifugation and indirect immunofluorescence staining analyses reveal that ethanol (0.5% and 1%) also displaces cell-surface TβR-I and TβR-II from lipid rafts/caveolae and facilitates translocation of these receptors to non-lipid raft microdomains where canonical signaling occurs. These results suggest that ethanol enhances canonical TGF-β signaling by increasing non-lipid raft microdomain localization of the TGF-β receptors. Since TGF-β plays a protective role in ASCVD but can also cause ALD, the TGF-β enhancer activity of ethanol at low and high doses appears to be responsible for both beneficial and harmful effects. Ethanol also disrupts the location of lipid raft/caveolae of other membrane proteins (e.g., neurotransmitter, growth factor/cytokine, and G protein-coupled receptors) which utilize lipid rafts/caveolae as signaling platforms. Displacement of these membrane proteins induced by ethanol may result in a variety of pathologies in nerve, heart and other tissues.
Collapse
Affiliation(s)
| | - Chun-Lin Chen
- Department of Biological Science, National Sun Yat-sen University, Kaohsiung, 804, Taiwan.,Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University and Academia Sinica, Kaohsiung, 804, Taiwan
| | - Franklin W Huang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, 02115.,Harvard Medical School, Boston, Massachusetts, 02115
| | - Frank E Johnson
- Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri, 63104
| | - Jung San Huang
- Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, Doisy Research Center, St. Louis, Missouri, 63104
| |
Collapse
|
41
|
Rigon RB, Oyafuso MH, Fujimura AT, Gonçalez ML, do Prado AH, Gremião MPD, Chorilli M. Nanotechnology-Based Drug Delivery Systems for Melanoma Antitumoral Therapy: A Review. BIOMED RESEARCH INTERNATIONAL 2015; 2015:841817. [PMID: 26078967 PMCID: PMC4442269 DOI: 10.1155/2015/841817] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 04/06/2015] [Accepted: 04/07/2015] [Indexed: 12/11/2022]
Abstract
Melanoma (MEL) is a less common type of skin cancer, but it is more aggressive with a high mortality rate. The World Cancer Research Fund International (GLOBOCAN 2012) estimates that there were 230,000 new cases of MEL in the world in 2012. Conventional MEL treatment includes surgery and chemotherapy, but many of the chemotherapeutic agents used present undesirable properties. Drug delivery systems are an alternative strategy by which to carry antineoplastic agents. Encapsulated drugs are advantageous due to such properties as high stability, better bioavailability, controlled drug release, a long blood circulation time, selective organ or tissue distribution, a lower total required dose, and minimal toxic side effects. This review of scientific research supports applying a nanotechnology-based drug delivery system for MEL therapy.
Collapse
Affiliation(s)
- Roberta Balansin Rigon
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Márcia Helena Oyafuso
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Andressa Terumi Fujimura
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Maíra Lima Gonçalez
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Alice Haddad do Prado
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Maria Palmira Daflon Gremião
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Sciences, Department of Drug and Medicines, São Paulo State University, 14801-902 Araraquara, SP, Brazil
| |
Collapse
|
42
|
Albulescu L, Wubbolts R, van Kuppeveld FJM, Strating JRPM. Cholesterol shuttling is important for RNA replication of coxsackievirus B3 and encephalomyocarditis virus. Cell Microbiol 2015; 17:1144-56. [PMID: 25645595 DOI: 10.1111/cmi.12425] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 01/21/2015] [Accepted: 01/26/2015] [Indexed: 11/29/2022]
Abstract
Picornaviruses are a family of positive-strand RNA viruses that includes important human and animal pathogens. Upon infection, picornaviruses induce an extensive remodelling of host cell membranes into replication organelles (ROs), which is critical for replication. Membrane lipids and lipid remodelling processes are at the base of RO formation, yet their involvement remains largely obscure. Recently, phosphatidylinositol-4-phosphate was the first lipid discovered to be important for the replication of a number of picornaviruses. Here, we investigate the role of the lipid cholesterol in picornavirus replication. We show that two picornaviruses from distinct genera that rely on different host factors for replication, namely the enterovirus coxsackievirus B3 (CVB3) and the cardiovirus encephalomyocarditis virus (EMCV), both recruited cholesterol to their ROs. Although CVB3 and EMCV both required cholesterol for efficient genome replication, the viruses appeared to rely on different cellular cholesterol pools. Treatments that altered the distribution of endosomal cholesterol inhibited replication of both CVB3 and EMCV, showing the importance of endosomal cholesterol shuttling for the replication of these viruses. Summarizing, we here demonstrate the importance of cholesterol homeostasis for efficient replication of CVB3 and EMCV.
Collapse
Affiliation(s)
- Lucian Albulescu
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Richard Wubbolts
- Department of Biochemistry and Cell Biology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Frank J M van Kuppeveld
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Jeroen R P M Strating
- Virology Division, Department of Infectious Diseases and Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
43
|
Moscatelli A, Gagliardi A, Maneta-Peyret L, Bini L, Stroppa N, Onelli E, Landi C, Scali M, Idilli AI, Moreau P. Characterisation of detergent-insoluble membranes in pollen tubes of Nicotiana tabacum (L.). Biol Open 2015; 4:378-99. [PMID: 25701665 PMCID: PMC4359744 DOI: 10.1242/bio.201410249] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Pollen tubes are the vehicle for sperm cell delivery to the embryo sac during fertilisation of Angiosperms. They provide an intriguing model for unravelling mechanisms of growing to extremes. The asymmetric distribution of lipids and proteins in the pollen tube plasma membrane modulates ion fluxes and actin dynamics and is maintained by a delicate equilibrium between exocytosis and endocytosis. The structural constraints regulating polarised secretion and asymmetric protein distribution on the plasma membrane are mostly unknown. To address this problem, we investigated whether ordered membrane microdomains, namely membrane rafts, might contribute to sperm cell delivery. Detergent insoluble membranes, rich in sterols and sphingolipids, were isolated from tobacco pollen tubes. MALDI TOF/MS analysis revealed that actin, prohibitins and proteins involved in methylation reactions and in phosphoinositide pattern regulation are specifically present in pollen tube detergent insoluble membranes. Tubulins, voltage-dependent anion channels and proteins involved in membrane trafficking and signalling were also present. This paper reports the first evidence of membrane rafts in Angiosperm pollen tubes, opening new perspectives on the coordination of signal transduction, cytoskeleton dynamics and polarised secretion.
Collapse
Affiliation(s)
- Alessandra Moscatelli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Assunta Gagliardi
- Laboratorio di Proteomica Funzionale, Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Lilly Maneta-Peyret
- Laboratoire de Biogenèse Membranaire, Université Bordeaux Segalen, 71 Avenue Edouard Bourlaux, 33883 Villenave d'Ornon, France
| | - Luca Bini
- Laboratorio di Proteomica Funzionale, Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Nadia Stroppa
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Elisabetta Onelli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Claudia Landi
- Laboratorio di Proteomica Funzionale, Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Monica Scali
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via P. A. Mattioli 4, 53100 Siena, Italy
| | - Aurora Irene Idilli
- Dipartimento di Bioscienze, Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy Present address: Institute of Biophysics, National Research Council and FBK, 38123 Trento, Italy
| | - Patrick Moreau
- Laboratoire de Biogenèse Membranaire, Université Bordeaux Segalen, 71 Avenue Edouard Bourlaux, 33883 Villenave d'Ornon, France
| |
Collapse
|
44
|
De Vuyst É, Giltaire S, Lambert de Rouvroit C, Chrétien A, Salmon M, Poumay Y. The activation of cultured keratinocytes by cholesterol depletion during reconstruction of a human epidermis is reminiscent of monolayer cultures. Arch Dermatol Res 2015; 307:309-18. [DOI: 10.1007/s00403-015-1537-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 12/05/2014] [Accepted: 01/06/2015] [Indexed: 10/24/2022]
|
45
|
A cholesterol consensus motif is required for efficient intracellular transport and raft association of a group 2 HA from influenza virus. Biochem J 2015; 465:305-14. [DOI: 10.1042/bj20141114] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The external part of the transmembrane region of HA (haemagglutinin) of influenza virus contains a cholesterol consensus motif originally identified in G-protein-coupled receptors. Various mutations in this motif retard transport of HA through the Golgi and reduce raft association.
Collapse
|
46
|
Anderson HA, Roche PA. MHC class II association with lipid rafts on the antigen presenting cell surface. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2014; 1853:775-80. [PMID: 25261705 DOI: 10.1016/j.bbamcr.2014.09.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 09/16/2014] [Accepted: 09/17/2014] [Indexed: 12/29/2022]
Abstract
MHC class II (MHC-II) molecules function by binding peptides derived from either self or foreign proteins and expressing these peptides on the surface of antigen presenting cells (APCs) for recognition by CD4 T cells. MHC-II is known to exist on clusters on the surface of APCs, and a variety of biochemical and functional studies have suggested that these clusters represent lipid raft microdomain-associated MHC-II. This review will summarize data exploring the biosynthesis of raft-associated MHC-II and the role that lipid raft association plays in regulating T cell activation by APCs. This article is part of a Special Issue entitled: Nanoscale membrane organisation and signalling.
Collapse
Affiliation(s)
- Howard A Anderson
- Division of Therapeutic Proteins, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Paul A Roche
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
47
|
Diaz-Rohrer B, Levental KR, Levental I. Rafting through traffic: Membrane domains in cellular logistics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:3003-3013. [PMID: 25130318 DOI: 10.1016/j.bbamem.2014.07.029] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/28/2014] [Accepted: 07/31/2014] [Indexed: 01/03/2023]
Abstract
The intricate and tightly regulated organization of eukaryotic cells into spatially and functionally distinct membrane-bound compartments is a defining feature of complex organisms. These compartments are defined by their lipid and protein compositions, with their limiting membrane as the functional interface to the rest of the cell. Thus, proper segregation of membrane proteins and lipids is necessary for the maintenance of organelle identity, and this segregation must be maintained despite extensive, rapid membrane exchange between compartments. Sorting processes of high efficiency and fidelity are required to avoid potentially deleterious mis-targeting and maintain cellular function. Although much molecular machinery associated with membrane traffic (i.e. membrane budding/fusion/fission) has been characterized both structurally and biochemically, the mechanistic details underlying the tightly regulated distribution of membranes between subcellular locations remain to be elucidated. This review presents evidence for the role of ordered lateral membrane domains known as lipid rafts in both biosynthetic sorting in the late secretory pathway, as well as endocytosis and recycling to/from the plasma membrane. Although such evidence is extensive and the involvement of membrane domains in sorting is definitive, specific mechanistic details for raft-dependent sorting processes remain elusive.
Collapse
Affiliation(s)
- Blanca Diaz-Rohrer
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA
| | - Kandice R Levental
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA
| | - Ilya Levental
- University of Texas Health Science Center at Houston, 6431 Fannin St, Houston, TX 77030, USA; Cancer Prevention and Research Institute of Texas, USA.
| |
Collapse
|
48
|
Influenza A virus hemagglutinin and neuraminidase mutually accelerate their apical targeting through clustering of lipid rafts. J Virol 2014; 88:10039-55. [PMID: 24965459 DOI: 10.1128/jvi.00586-14] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED In polarized epithelial cells, influenza A virus hemagglutinin (HA) and neuraminidase (NA) are intrinsically associated with lipid rafts and target the apical plasma membrane for viral assembly and budding. Previous studies have indicated that the transmembrane domain (TMD) and cytoplasmic tail (CT) of HA and NA are required for association with lipid rafts, but the raft dependencies of their apical targeting are controversial. Here, we show that coexpression of HA with NA accelerated their apical targeting through accumulation in lipid rafts. HA was targeted to the apical plasma membrane even when expressed alone, but the kinetics was much slower than that of HA in infected cells. Coexpression experiments revealed that apical targeting of HA and NA was accelerated by their coexpression. The apical targeting of HA was also accelerated by coexpression with M1 but not M2. The mutations in the outer leaflet of the TMD and the deletion of the CT in HA and NA that reduced their association with lipid rafts abolished the acceleration of their apical transport, indicating that the lipid raft association is essential for efficient apical trafficking of HA and NA. An in situ proximity ligation assay (PLA) revealed that HA and NA were accumulated and clustered in the cytoplasmic compartments only when both were associated with lipid rafts. Analysis with mutant viruses containing nonraft HA/NA confirmed these findings. We further analyzed lipid raft markers by in situ PLA and suggest a possible mechanism of the accelerated apical transport of HA and NA via clustering of lipid rafts. IMPORTANCE Lipid rafts serve as sites for viral entry, particle assembly, and budding, leading to efficient viral replication. The influenza A virus utilizes lipid rafts for apical plasma membrane targeting and particle budding. The hemagglutinin (HA) and neuraminidase (NA) of influenza virus, key players for particle assembly, contain determinants for apical sorting and lipid raft association. However, it remains to be elucidated how lipid rafts contribute to the apical trafficking and budding. We investigated the relation of lipid raft association of HA and NA to the efficiency of apical trafficking. We show that coexpression of HA and NA induces their accumulation in lipid rafts and accelerates their apical targeting, and we suggest that the accelerated apical transport likely occurs by clustering of lipid rafts at the TGN. This finding provides the first evidence that two different raft-associated viral proteins induce lipid raft clustering, thereby accelerating apical trafficking of the viral proteins.
Collapse
|
49
|
Tokumasu F, Crivat G, Ackerman H, Hwang J, Wellems TE. Inward cholesterol gradient of the membrane system in P. falciparum-infected erythrocytes involves a dilution effect from parasite-produced lipids. Biol Open 2014; 3:529-41. [PMID: 24876390 PMCID: PMC4058088 DOI: 10.1242/bio.20147732] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Plasmodium falciparum (Pf) infection remodels the human erythrocyte with new membrane systems, including a modified host erythrocyte membrane (EM), a parasitophorous vacuole membrane (PVM), a tubulovesicular network (TVN), and Maurer's clefts (MC). Here we report on the relative cholesterol contents of these membranes in parasitized normal (HbAA) and hemoglobin S-containing (HbAS, HbAS) erythrocytes. Results from fluorescence lifetime imaging microscopy (FLIM) experiments with a cholesterol-sensitive fluorophore show that membrane cholesterol levels in parasitized erythrocytes (pRBC) decrease inwardly from the EM, to the MC/TVN, to the PVM, and finally to the parasite membrane (PM). Cholesterol depletion of pRBC by methyl-β-cyclodextrin treatment caused a collapse of this gradient. Lipid and cholesterol exchange data suggest that the cholesterol gradient involves a dilution effect from non-sterol lipids produced by the parasite. FLIM signals from the PVM or PM showed little or no difference between parasitized HbAA vs HbS-containing erythrocytes that differed in lipid content, suggesting that malaria parasites may regulate the cholesterol contents of the PVM and PM independently of levels in the host cell membrane. Cholesterol levels may affect raft structures and the membrane trafficking and sorting functions that support Pf survival in HbAA, HbAS and HbSS erythrocytes.
Collapse
Affiliation(s)
- Fuyuki Tokumasu
- Malaria Genetics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-8132, USA Present address: Department of Lipidomics, Faculty of Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Georgeta Crivat
- Malaria Genetics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-8132, USA Quantum Electronics and Photonics Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Boulder, CO 80305, USA
| | - Hans Ackerman
- Malaria Genetics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-8132, USA
| | - Jeeseong Hwang
- Quantum Electronics and Photonics Division, Physical Measurement Laboratory, National Institute of Standards and Technology, Boulder, CO 80305, USA
| | - Thomas E Wellems
- Malaria Genetics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-8132, USA
| |
Collapse
|
50
|
Abstract
The lipid raft hypothesis proposes lateral domains driven by preferential interactions between sterols, sphingolipids, and specific proteins as a central mechanism for the regulation of membrane structure and function; however, experimental limitations in defining raft composition and properties have prevented unequivocal demonstration of their functional relevance. Here, we establish a quantitative, functional relationship between raft association and subcellular protein sorting. By systematic mutation of the transmembrane and juxtamembrane domains of a model transmembrane protein, linker for activation of T-cells (LAT), we generated a panel of variants possessing a range of raft affinities. These mutations revealed palmitoylation, transmembrane domain length, and transmembrane sequence to be critical determinants of membrane raft association. Moreover, plasma membrane (PM) localization was strictly dependent on raft partitioning across the entire panel of unrelated mutants, suggesting that raft association is necessary and sufficient for PM sorting of LAT. Abrogation of raft partitioning led to mistargeting to late endosomes/lysosomes because of a failure to recycle from early endosomes. These findings identify structural determinants of raft association and validate lipid-driven domain formation as a mechanism for endosomal protein sorting.
Collapse
|