1
|
Dell'Annunziata F, Mosidze E, Folliero V, Lamparelli EP, Lopardo V, Pagliano P, Porta GD, Galdiero M, Bakuridze AD, Franci G. Eco-friendly synthesis of silver nanoparticles from peel and juice C. limon and their antiviral efficacy against HSV-1 and SARS-CoV-2. Virus Res 2024; 349:199455. [PMID: 39181453 PMCID: PMC11387364 DOI: 10.1016/j.virusres.2024.199455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/05/2024] [Accepted: 08/18/2024] [Indexed: 08/27/2024]
Abstract
The growing threat of viral infections requires innovative therapeutic approaches to safeguard human health. Nanomaterials emerge as a promising solution to overcome the limitations associated with conventional therapies. The eco-friendly synthesis of silver nanoparticles (AgNPs) currently represents a method that guarantees antimicrobial efficacy, safety, and cost-effectiveness. This study explores the use of AgNPs derived from the peel (Lp-AgNPs) and juice (Lj-AgNPs) Citrus limon "Ovale di Sorrento", cultivars of the Campania region. The antiviral potential was tested against viruses belonging to the Coronaviridae and Herpesviridae. AgNPs were synthesized by reduction method using silver nitrate solution mixed with aqueous extract of C. limon peel and juice. The formation of Lp-AgNPs and Lj-AgNPs was assessed using a UV-Vis spectrophotometer. The size, ζ-potential, concentration, and morphology of AgNPs were evaluated by dynamic light scattering (DLS), nanoparticle tracking analysis (NTA), and field emission-scanning electron microscopy (FE-SEM). Cytotoxicity was evaluated in a concentration range between 500 and 7.8 µg/mL on VERO-76 and HaCaT cells, with the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium test bromide (MTT). Antiviral activity consisted of virus pre-treatment, co-treatment, cellular pre-treatment, and post-infection tests versus HSV-1 and SARS-CoV-2 at a multiplicity of infections (MOI) of 0.01. Plaque reduction assays and real-time PCR provided data on the antiviral potential of tested compounds. Lp-AgNPs and Lj-AgNPs exhibited spherical morphology with respective diameters of 60 and 92 nm with concentrations of 4.22 and 4.84 × 1010 particles/mL, respectively. The MTT data demonstrated minimal cytotoxicity, with 50 % cytotoxic concentrations (CC50) of Lp-AgNPs and Lj-AgNPs against VERO cells of 754.6 and 486.7 µg/mL. Similarly, CC50 values against HaCaT were 457.3 µg/mL for Lp-AgNPs and 339.6 µg/mL for Lj-AgNPs, respectively. In the virus pre-treatment assay, 90 % inhibitory concentrations of HSV-1 and SARS-CoV-2 were 8.54-135.04 µg/mL for Lp-AgNPs and 6.13-186.77 µg/mL for Lj-AgNPs, respectively. The molecular investigation confirmed the antiviral data, recording a reduction in the UL54 and UL27 genes for HSV-1 and in the Spike (S) gene for SARS-CoV-2, following AgNP exposure. The results of this study suggest that Lp-AgNPs and Lj-AgNPs derived from C. Limon could offer a valid ecological, natural, local and safe strategy against viral infections.
Collapse
Affiliation(s)
- Federica Dell'Annunziata
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy; Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Ekaterine Mosidze
- Department of Pharmaceutical Technology, Tbilisi State Medical University, 33 Vazha-Pshavela Ave, Tbilisi, 0178, Georgia
| | - Veronica Folliero
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Erwin P Lamparelli
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Valentina Lopardo
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", 80138 Naples, Italy
| | - Aliosha Dzh Bakuridze
- Department of Pharmaceutical Technology, Tbilisi State Medical University, 33 Vazha-Pshavela Ave, Tbilisi, 0178, Georgia.
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, 84081 Baronissi, Italy; UOC Patologia e Microbiologia, San Giovanni di Dio e Ruggi D'Aragona University Hospital, 84126 Salerno, Italy.
| |
Collapse
|
2
|
Desai N, Rana D, Salave S, Benival D, Khunt D, Prajapati BG. Achieving Endo/Lysosomal Escape Using Smart Nanosystems for Efficient Cellular Delivery. Molecules 2024; 29:3131. [PMID: 38999083 PMCID: PMC11243486 DOI: 10.3390/molecules29133131] [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: 06/06/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
The delivery of therapeutic agents faces significant hurdles posed by the endo-lysosomal pathway, a bottleneck that hampers clinical effectiveness. This comprehensive review addresses the urgent need to enhance cellular delivery mechanisms to overcome these obstacles. It focuses on the potential of smart nanomaterials, delving into their unique characteristics and mechanisms in detail. Special attention is given to their ability to strategically evade endosomal entrapment, thereby enhancing therapeutic efficacy. The manuscript thoroughly examines assays crucial for understanding endosomal escape and cellular uptake dynamics. By analyzing various assessment methods, we offer nuanced insights into these investigative approaches' multifaceted aspects. We meticulously analyze the use of smart nanocarriers, exploring diverse mechanisms such as pore formation, proton sponge effects, membrane destabilization, photochemical disruption, and the strategic use of endosomal escape agents. Each mechanism's effectiveness and potential application in mitigating endosomal entrapment are scrutinized. This paper provides a critical overview of the current landscape, emphasizing the need for advanced delivery systems to navigate the complexities of cellular uptake. Importantly, it underscores the transformative role of smart nanomaterials in revolutionizing cellular delivery strategies, leading to a paradigm shift towards improved therapeutic outcomes.
Collapse
Affiliation(s)
- Nimeet Desai
- Indian Institute of Technology Hyderabad, Kandi 502285, Telangana, India;
| | - Dhwani Rana
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Sagar Salave
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Derajram Benival
- National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad 382355, Gujarat, India; (D.R.); (S.S.); (D.B.)
| | - Dignesh Khunt
- School of Pharmacy, Gujarat Technological University, Gandhinagar 382027, Gujarat, India
| | - Bhupendra G. Prajapati
- Shree S. K. Patel College of Pharmaceutical Education and Research, Ganpat University, Kherva 384012, Gujarat, India
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand
| |
Collapse
|
3
|
Ramirez JM, Calderon-Zavala AC, Balaram A, Heldwein EE. In vitro reconstitution of herpes simplex virus 1 fusion identifies low pH as a fusion co-trigger. mBio 2023; 14:e0208723. [PMID: 37874146 PMCID: PMC10746285 DOI: 10.1128/mbio.02087-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/25/2023] Open
Abstract
IMPORTANCE HSV-1 causes lifelong, incurable infections and diseases ranging from mucocutaneous lesions to fatal encephalitis. Fusion of viral and host membranes is a critical step in HSV-1 infection of target cells that requires multiple factors on both the viral and host sides. Due to this complexity, many fundamental questions remain unanswered, such as the identity of the viral and host factors that are necessary and sufficient for HSV-1-mediated membrane fusion and the nature of the fusion trigger. Here, we developed a simplified in vitro fusion assay to examine the fusion requirements and identified low pH as a co-trigger for virus-mediated fusion in vitro. We hypothesize that low pH has a critical role in cell entry and, potentially, pathogenesis.
Collapse
Affiliation(s)
- J. Martin Ramirez
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Medical Scientist Training Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ariana C. Calderon-Zavala
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ariane Balaram
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Ekaterina E. Heldwein
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, Massachusetts, USA
- Graduate Program in Molecular Microbiology, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
- Medical Scientist Training Program, Graduate School of Biomedical Sciences, Tufts University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Fenizia S, Gaggini M, Vassalle C. The Sphingolipid-Signaling Pathway as a Modulator of Infection by SARS-CoV-2. Curr Issues Mol Biol 2023; 45:7956-7973. [PMID: 37886946 PMCID: PMC10605018 DOI: 10.3390/cimb45100503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 10/28/2023] Open
Abstract
Ceramides and other related sphingolipids, important cellular components linked to metabolic homeostasis and cardiometabolic diseases, have been found to be involved in different steps of the SARS-CoV-2 life cycle. Hence, changes in their physiological levels are identified as predictors of COVID-19 severity and prognosis, as well as potential therapeutic targets. In this review, an overview of the SARS-CoV-2 life cycle is given, followed by a description of the sphingolipid metabolism and its role in viral infection, with a particular focus on those steps required to finalize the viral life cycle. Furthermore, the use and development of pharmaceutical strategies to target sphingolipids to prevent and treat severe and long-term symptoms of infectious diseases, particularly COVID-19, are reviewed herein. Finally, research perspectives and current challenges in this research field are highlighted. Although many aspects of sphingolipid metabolism are not fully known, this review aims to highlight how the discovery and use of molecules targeting sphingolipids with reliable and selective properties may offer new therapeutic alternatives to infectious and other diseases, including COVID-19.
Collapse
Affiliation(s)
- Simona Fenizia
- Istituto di Fisiologia Clinica, Italian National Research Council, Via Moruzzi 1, 56124 Pisa, Italy
| | - Melania Gaggini
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| | - Cristina Vassalle
- Fondazione CNR-Regione Toscana G. Monasterio, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
5
|
Ahmed N, Francis ME, Ahmed N, Kelvin AA, Pezacki JP. microRNA-185 Inhibits SARS-CoV-2 Infection through the Modulation of the Host's Lipid Microenvironment. Viruses 2023; 15:1921. [PMID: 37766327 PMCID: PMC10536008 DOI: 10.3390/v15091921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/01/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
With the emergence of the novel betacoronavirus Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), there has been an urgent need for the development of fast-acting antivirals, particularly in dealing with different variants of concern (VOC). SARS-CoV-2, like other RNA viruses, depends on host cell machinery to propagate and misregulate metabolic pathways to its advantage. Herein, we discovered that the immunometabolic microRNA-185 (miR-185) restricts SARS-CoV-2 propagation by affecting its entry and infectivity. The antiviral effects of miR-185 were studied in SARS-CoV-2 Spike protein pseudotyped virus, surrogate virus (HCoV-229E), as well as live SARS-CoV-2 virus in Huh7, A549, and Calu-3 cells. In each model, we consistently observed microRNA-induced reduction in lipid metabolism pathways-associated genes including SREBP2, SQLE, PPARG, AGPAT3, and SCARB1. Interestingly, we also observed changes in angiotensin-converting enzyme 2 (ACE2) levels, the entry receptor for SARS-CoV-2. Taken together, these data show that miR-185 significantly restricts host metabolic and other pathways that appear to be essential to SAR-CoV-2 replication and propagation. Overall, this study highlights an important link between non-coding RNAs, immunometabolic pathways, and viral infection. miR-185 mimics alone or in combination with other antiviral therapeutics represent possible future fast-acting antiviral strategies that are likely to be broadly antiviral against multiple variants as well as different virus types of potential pandemics.
Collapse
Affiliation(s)
- Nadine Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Magen E. Francis
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - Noreen Ahmed
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Alyson A. Kelvin
- Department of Biochemistry, Microbiology, and Immunology, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Vaccine and Infectious Disease Organization-International Vaccine Centre (VIDO-InterVac), University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
6
|
Tate P, Mastrodomenico V, Cunha C, McClure J, Barron AE, Diamond G, Mounce BC, Kirshenbaum K. Peptidomimetic Oligomers Targeting Membrane Phosphatidylserine Exhibit Broad Antiviral Activity. ACS Infect Dis 2023; 9:1508-1522. [PMID: 37530426 PMCID: PMC10425984 DOI: 10.1021/acsinfecdis.3c00063] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Indexed: 08/03/2023]
Abstract
The development of durable new antiviral therapies is challenging, as viruses can evolve rapidly to establish resistance and attenuate therapeutic efficacy. New compounds that selectively target conserved viral features are attractive therapeutic candidates, particularly for combating newly emergent viral threats. The innate immune system features a sustained capability to combat pathogens through production of antimicrobial peptides (AMPs); however, these AMPs have shortcomings that can preclude clinical use. The essential functional features of AMPs have been recapitulated by peptidomimetic oligomers, yielding effective antibacterial and antifungal agents. Here, we show that a family of AMP mimetics, called peptoids, exhibit direct antiviral activity against an array of enveloped viruses, including the key human pathogens Zika, Rift Valley fever, and chikungunya viruses. These data suggest that the activities of peptoids include engagement and disruption of viral membrane constituents. To investigate how these peptoids target lipid membranes, we used liposome leakage assays to measure membrane disruption. We found that liposomes containing phosphatidylserine (PS) were markedly sensitive to peptoid treatment; in contrast, liposomes formed exclusively with phosphatidylcholine (PC) showed no sensitivity. In addition, chikungunya virus containing elevated envelope PS was more susceptible to peptoid-mediated inactivation. These results indicate that peptoids mimicking the physicochemical characteristics of AMPs act through a membrane-specific mechanism, most likely through preferential interactions with PS. We provide the first evidence for the engagement of distinct viral envelope lipid constituents, establishing an avenue for specificity that may enable the development of a new family of therapeutics capable of averting the rapid development of resistance.
Collapse
Affiliation(s)
- Patrick
M. Tate
- Department
of Chemistry, New York University, New York, New York 10003, United States
| | - Vincent Mastrodomenico
- Department
of Microbiology and Immunology, Loyola University
Chicago Medical Center, Maywood, Illinois 60130, United States
| | - Christina Cunha
- Department
of Microbiology and Immunology, Loyola University
Chicago Medical Center, Maywood, Illinois 60130, United States
| | | | - Annelise E. Barron
- Maxwell
Biosciences, Austin, Texas 78738, United States
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| | - Gill Diamond
- Department
of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, Kentucky 40292, United States
| | - Bryan C. Mounce
- Department
of Microbiology and Immunology, Loyola University
Chicago Medical Center, Maywood, Illinois 60130, United States
| | - Kent Kirshenbaum
- Department
of Chemistry, New York University, New York, New York 10003, United States
- Maxwell
Biosciences, Austin, Texas 78738, United States
| |
Collapse
|
7
|
Polita A, Stancikaitė M, Žvirblis R, Maleckaitė K, Dodonova-Vaitkūnienė J, Tumkevičius S, Shivabalan AP, Valinčius G. Designing a green-emitting viscosity-sensitive 4,4-difluoro-4-bora-3a,4a-diaza- s-indacene (BODIPY) probe for plasma membrane viscosity imaging. RSC Adv 2023; 13:19257-19264. [PMID: 37377877 PMCID: PMC10291278 DOI: 10.1039/d3ra04126c] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/29/2023] Open
Abstract
Viscosity is a key characteristic of lipid membranes - it governs the passive diffusion of solutes and affects the lipid raft formation and membrane fluidity. Precise determination of viscosity values in biological systems is of great interest and viscosity-sensitive fluorescent probes offer a convenient solution for this task. In this work we present a novel membrane-targeting and water-soluble viscosity probe BODIPY-PM, which is based on one of the most frequently used probes BODIPY-C10. Despite its regular use, BODIPY-C10 suffers from poor integration into liquid-ordered lipid phases and lack of water solubility. Here, we investigate the photophysical characteristics of BODIPY-PM and demonstrate that solvent polarity only slightly affects the viscosity-sensing qualities of BODIPY-PM. In addition, with fluorescence lifetime imaging microscopy (FLIM), we imaged microviscosity in complex biological systems - large unilamellar vesicles (LUVs), tethered bilayer membranes (tBLMs) and live lung cancer cells. Our study showcases that BODIPY-PM preferentially stains the plasma membranes of live cells, equally well partitions into both liquid-ordered and liquid-disordered phases and reliably distinguishes lipid phase separation in tBLMs and LUVs.
Collapse
Affiliation(s)
- Artūras Polita
- Institute of Biochemistry, Life Sciences Center, Vilnius University Saulėtekio Av. 7 Vilnius LT-10257 Lithuania
| | - Milda Stancikaitė
- Center of Physical Sciences and Technology Saulėtekio Av. 3 Vilnius LT-10257 Lithuania
| | - Rokas Žvirblis
- Life Sciences Center, Institute of Biotechnology, Vilnius University Saulėtekio Av. 7 Vilnius LT-10257 Lithuania
| | - Karolina Maleckaitė
- Center of Physical Sciences and Technology Saulėtekio Av. 3 Vilnius LT-10257 Lithuania
| | - Jelena Dodonova-Vaitkūnienė
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University Naugarduko St. 24 Vilnius LT-03225 Lithuania
| | - Sigitas Tumkevičius
- Institute of Chemistry, Faculty of Chemistry and Geosciences, Vilnius University Naugarduko St. 24 Vilnius LT-03225 Lithuania
| | - Arun Prabha Shivabalan
- Institute of Biochemistry, Life Sciences Center, Vilnius University Saulėtekio Av. 7 Vilnius LT-10257 Lithuania
| | - Gintaras Valinčius
- Institute of Biochemistry, Life Sciences Center, Vilnius University Saulėtekio Av. 7 Vilnius LT-10257 Lithuania
| |
Collapse
|
8
|
Frawley AT, Leslie KG, Wycisk V, Galiani S, Shrestha D, Eggeling C, Anderson HL. A Photoswitchable Solvatochromic Dye for Probing Membrane Ordering by RESOLFT Super-resolution Microscopy. Chemphyschem 2023; 24:e202300125. [PMID: 36946252 DOI: 10.1002/cphc.202300125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 03/23/2023]
Abstract
A switchable solvatochromic fluorescent dyad can be used to map ordering of lipids in vesicle membranes at a resolution better than the diffraction limit. Combining a Nile Red fluorophore with a photochromic spironaphthoxazine quencher allows the fluorescence to be controlled using visible light, via photoswitching and FRET quenching. Synthetic lipid vesicles of varying composition were imaged with an average 2.5-fold resolution enhancement, compared to the confocal images. Ratiometric detection was used to probe the membrane polarity, and domains of different lipid ordering were distinguished within the same membrane.
Collapse
Affiliation(s)
- Andrew T Frawley
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Kathryn G Leslie
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Virginia Wycisk
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| | - Silvia Galiani
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Dilip Shrestha
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
- Institute of Applied Optics and Biophysics, Friedrich-Schiller-University Jena, Max-Wien-Platz 4, 07743, Jena, Germany
- Leibniz Institute of Photonic Technology e.V., Albert-Einstein-Strasse 9, 07745, Jena, Germany
- Jena Center for Soft Matter (JCSM), Philosophenweg 7, 07743, Jena, Germany
| | - Harry L Anderson
- Department of Chemistry, University of Oxford, Chemistry Research Laboratory, Mansfield Road, Oxford, OX1 3TA, UK
| |
Collapse
|
9
|
Qiu C, Xia F, Zhang J, Shi Q, Meng Y, Wang C, Pang H, Gu L, Xu C, Guo Q, Wang J. Advanced Strategies for Overcoming Endosomal/Lysosomal Barrier in Nanodrug Delivery. RESEARCH (WASHINGTON, D.C.) 2023; 6:0148. [PMID: 37250954 PMCID: PMC10208951 DOI: 10.34133/research.0148] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 04/27/2023] [Indexed: 05/31/2023]
Abstract
Nanocarriers have therapeutic potential to facilitate drug delivery, including biological agents, small-molecule drugs, and nucleic acids. However, their efficiency is limited by several factors; among which, endosomal/lysosomal degradation after endocytosis is the most important. This review summarizes advanced strategies for overcoming endosomal/lysosomal barriers to efficient nanodrug delivery based on the perspective of cellular uptake and intracellular transport mechanisms. These strategies include promoting endosomal/lysosomal escape, using non-endocytic methods of delivery to directly cross the cell membrane to evade endosomes/lysosomes and making a detour pathway to evade endosomes/lysosomes. On the basis of the findings of this review, we proposed several promising strategies for overcoming endosomal/lysosomal barriers through the smarter and more efficient design of nanodrug delivery systems for future clinical applications.
Collapse
Affiliation(s)
- Chong Qiu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Fei Xia
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Junzhe Zhang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiaoli Shi
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Yuqing Meng
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chen Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Huanhuan Pang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Liwei Gu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Chengchao Xu
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Qiuyan Guo
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Jigang Wang
- Artemisinin Research Center, and Institute of Chinese Materia Medica,
China Academy of Chinese Medical Sciences, Beijing 100700, China
- Department of Nephrology, and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital,
Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| |
Collapse
|
10
|
Lau NCH, Yam JWP. From Exosome Biogenesis to Absorption: Key Takeaways for Cancer Research. Cancers (Basel) 2023; 15:cancers15071992. [PMID: 37046653 PMCID: PMC10093369 DOI: 10.3390/cancers15071992] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 03/29/2023] Open
Abstract
Exosomes are mediators of intercellular communication in normal physiology and diseases. While many studies have emerged on the function of exosomal cargoes, questions remain regarding the origin of these exosomes. The packaging and secretion of exosomes in different contexts modify exosomal composition, which may in turn impact delivery, uptake and cargo function in recipient cells. A mechanistic understanding of exosome biology is therefore crucial to investigating exosomal function in complex biological systems and to the development of novel therapeutic approaches. Here, we outline the steps in exosome biogenesis, including endosome formation, MVB formation, cargo sorting and extracellular release, as well as exosome absorption, including targeting, interaction with recipient cells and the fate of internalized exosomes. In addition to providing a framework of exosome dynamics, we summarize current evidence on major pathways and regulatory mechanisms. We also highlight the various mechanisms observed in cancer and point out directions to improve study design in exosome biology. Further research is needed to illuminate the relationship between exosome biogenesis and function, which will aid the development of translational applications.
Collapse
Affiliation(s)
- Nicolas Cheuk Hang Lau
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Judy Wai Ping Yam
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
- State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
- Correspondence: ; Tel.: +852-22552681
| |
Collapse
|
11
|
Shekunov EV, Zlodeeva PD, Efimova SS, Muryleva AA, Zarubaev VV, Slita AV, Ostroumova OS. Cyclic lipopeptides as membrane fusion inhibitors against SARS-CoV-2: New tricks for old dogs. Antiviral Res 2023; 212:105575. [PMID: 36868316 PMCID: PMC9977712 DOI: 10.1016/j.antiviral.2023.105575] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
With the resurgence of the coronavirus pandemic, the repositioning of FDA-approved drugs against coronovirus and finding alternative strategies for antiviral therapy are both important. We previously identified the viral lipid envelope as a potential target for the prevention and treatment of SARS-CoV-2 infection with plant alkaloids (Shekunov et al., 2021). Here, we investigated the effects of eleven cyclic lipopeptides (CLPs), including well-known antifungal and antibacterial compounds, on the liposome fusion triggered by calcium, polyethylene glycol 8000, and a fragment of SARS-CoV-2 fusion peptide (816-827) by calcein release assays. Differential scanning microcalorimetry of the gel-to-liquid-crystalline and lamellar-to-inverted hexagonal phase transitions and confocal fluorescence microscopy demonstrated the relation of the fusion inhibitory effects of CLPs to alterations in lipid packing, membrane curvature stress and domain organization. The antiviral effects of CLPs were evaluated in an in vitro Vero-based cell model, and aculeacin A, anidulafugin, iturin A, and mycosubtilin attenuated the cytopathogenicity of SARS-CoV-2 without specific toxicity.
Collapse
Affiliation(s)
- Egor V Shekunov
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064, Saint Petersburg, Russia
| | - Polina D Zlodeeva
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064, Saint Petersburg, Russia
| | - Svetlana S Efimova
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064, Saint Petersburg, Russia
| | - Anna A Muryleva
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064, Saint Petersburg, Russia; Saint-Petersburg Pasteur Institute of Epidemiology and Microbiology, Mira 14, 197101, Saint Petersburg, Russia
| | - Vladimir V Zarubaev
- Saint-Petersburg Pasteur Institute of Epidemiology and Microbiology, Mira 14, 197101, Saint Petersburg, Russia
| | - Alexander V Slita
- Saint-Petersburg Pasteur Institute of Epidemiology and Microbiology, Mira 14, 197101, Saint Petersburg, Russia
| | - Olga S Ostroumova
- Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064, Saint Petersburg, Russia.
| |
Collapse
|
12
|
Systematic Identification and Comparison of the Expressed Profiles of Exosomal MiRNAs in Pigs Infected with NADC30-like PRRSV Strain. Animals (Basel) 2023; 13:ani13050876. [PMID: 36899733 PMCID: PMC10000162 DOI: 10.3390/ani13050876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Exosomes are biological vesicles secreted and released by cells that act as mediators of intercellular communication and play a unique role in virus infection, antigen presentation, and suppression/promotion of body immunity. Porcine reproductive and respiratory syndrome virus (PRRSV) is one of the most damaging pathogens in the pig industry and can cause reproductive disorders in sows, respiratory diseases in pigs, reduced growth performance, and other diseases leading to pig mortality. In this study, we used the PRRSV NADC30-like CHsx1401 strain to artificially infect 42-day-old pigs and isolate serum exosomes. Based on high-throughput sequencing technology, 305 miRNAs were identified in serum exosomes before and after infection, among which 33 miRNAs were significantly differentially expressed between groups (13 relatively upregulated and 20 relatively downregulated). Sequence conservation analysis of the CHsx1401 genome identified 8 conserved regions, of which a total of 16 differentially expressed (DE) miRNAs were predicted to bind to the conserved region closest to the 3' UTR of the CHsx1401 genome, including 5 DE miRNAs capable of binding to the CHsx1401 3' UTR (ssc-miR-34c, ssc-miR-375, ssc-miR-378, ssc-miR-486, ssc-miR-6529). Further analysis revealed that the target genes of differentially expressed miRNAs were widely involved in exosomal function-related and innate immunity-related signaling pathways, and 18 DE miRNAs (ssc-miR-4331-3p, ssc-miR-744, ssc-miR-320, ssc-miR-10b, ssc-miR-124a, ssc-miR-128, etc.) associated with PRRSV infection and immunity were screened as potential functional molecules involved in the regulation of PRRSV virus infection by exosomes.
Collapse
|
13
|
Maja M, Tyteca D. Alteration of cholesterol distribution at the plasma membrane of cancer cells: From evidence to pathophysiological implication and promising therapy strategy. Front Physiol 2022; 13:999883. [PMID: 36439249 PMCID: PMC9682260 DOI: 10.3389/fphys.2022.999883] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/10/2022] [Indexed: 11/11/2022] Open
Abstract
Cholesterol-enriched domains are nowadays proposed to contribute to cancer cell proliferation, survival, death and invasion, with important implications in tumor progression. They could therefore represent promising targets for new anticancer treatment. However, although diverse strategies have been developed over the years from directly targeting cholesterol membrane content/distribution to adjusting sterol intake, all approaches present more or less substantial limitations. Those data emphasize the need to optimize current strategies, to develop new specific cholesterol-targeting anticancer drugs and/or to combine them with additional strategies targeting other lipids than cholesterol. Those objectives can only be achieved if we first decipher (i) the mechanisms that govern the formation and deformation of the different types of cholesterol-enriched domains and their interplay in healthy cells; (ii) the mechanisms behind domain deregulation in cancer; (iii) the potential generalization of observations in different types of cancer; and (iv) the specificity of some alterations in cancer vs. non-cancer cells as promising strategy for anticancer therapy. In this review, we will discuss the current knowledge on the homeostasis, roles and membrane distribution of cholesterol in non-tumorigenic cells. We will then integrate documented alterations of cholesterol distribution in domains at the surface of cancer cells and the mechanisms behind their contribution in cancer processes. We shall finally provide an overview on the potential strategies developed to target those cholesterol-enriched domains in cancer therapy.
Collapse
|
14
|
Rahman K, Datta SA, Beaven AH, Jolley AA, Sodt AJ, Compton AA. Cholesterol Binds the Amphipathic Helix of IFITM3 and Regulates Antiviral Activity. J Mol Biol 2022; 434:167759. [PMID: 35872070 PMCID: PMC9342930 DOI: 10.1016/j.jmb.2022.167759] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 07/19/2022] [Accepted: 07/19/2022] [Indexed: 02/05/2023]
Abstract
The interferon-induced transmembrane (IFITM) proteins broadly inhibit the entry of diverse pathogenic viruses, including Influenza A virus (IAV), Zika virus, HIV-1, and SARS coronaviruses by inhibiting virus-cell membrane fusion. IFITM3 was previously shown to disrupt cholesterol trafficking, but the functional relationship between IFITM3 and cholesterol remains unclear. We previously showed that inhibition of IAV entry by IFITM3 is associated with its ability to promote cellular membrane rigidity, and these activities are functionally linked by a shared requirement for the amphipathic helix (AH) found in the intramembrane domain (IMD) of IFITM3. Furthermore, it has been shown that the AH of IFITM3 alters lipid membranes in vitro in a cholesterol-dependent manner. Therefore, we aimed to elucidate the relationship between IFITM3 and cholesterol in more detail. Using a fluorescence-based in vitro binding assay, we found that a peptide derived from the AH of IFITM3 directly interacted with the cholesterol analog, NBD-cholesterol, while other regions of the IFITM3 IMD did not, and native cholesterol competed with this interaction. In addition, recombinant full-length IFITM3 protein also exhibited NBD-cholesterol binding activity. Importantly, previously characterized mutations within the AH of IFITM3 that strongly inhibit antiviral function (F63Q and F67Q) disrupted AH structure in solution, inhibited cholesterol binding in vitro, and restricted bilayer insertion in silico. Our data suggest that direct interactions with cholesterol may contribute to the inhibition of membrane fusion pore formation by IFITM3. These findings may facilitate the design of therapeutic peptides for use in broad-spectrum antiviral therapy.
Collapse
Affiliation(s)
- Kazi Rahman
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Siddhartha A.K. Datta
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Andrew H. Beaven
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States,National Institute of General Medical Sciences, Bethesda, MD 20892, United States
| | - Abigail A. Jolley
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States
| | - Alexander J. Sodt
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, United States
| | - Alex A. Compton
- HIV Dynamics and Replication Program, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, United States,Corresponding author
| |
Collapse
|
15
|
Das T, Yang X, Lee H, Garst EH, Valencia E, Chandran K, Im W, Hang HC. S-Palmitoylation and Sterol Interactions Mediate Antiviral Specificity of IFITMs. ACS Chem Biol 2022; 17:2109-2120. [PMID: 35861660 PMCID: PMC10597057 DOI: 10.1021/acschembio.2c00176] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Interferon-induced transmembrane proteins (IFITM1, 2, and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV), and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITM proteins exhibit specificity in activity, but their distinct mechanisms of action and regulation are unclear. Since S-palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by photoaffinity cross-linking in mammalian cells along with molecular dynamic simulations and nuclear magnetic resonance analysis in vitro. These studies suggest that cholesterol can directly interact with S-palmitoylated IFITMs in cells and alter the conformation of IFITMs in membrane bilayers. Notably, we discovered that the S-palmitoylation levels regulate differential IFITM protein interactions with cholesterol in mammalian cells and specificity of antiviral activity toward IAV, SARS-CoV-2, and EBOV. Our studies suggest that modulation of IFITM S-palmitoylation levels and cholesterol interaction influence host susceptibility to different viruses.
Collapse
Affiliation(s)
- Tandrila Das
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, NY 10065, United States
- Tri-Institutional Ph.D. Program in Chemical Biology, New York, NY 10065, United States
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - Xinglin Yang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
| | - Hwayoung Lee
- Department of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA 18015, United States
| | - Emma H. Garst
- Laboratory of Chemical Biology and Microbial Pathogenesis, The Rockefeller University, New York, NY 10065, United States
- Tri-Institutional Ph.D. Program in Chemical Biology, New York, NY 10065, United States
| | - Estefania Valencia
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY 10461, United States
| | - Wonpil Im
- Department of Biological Sciences, Chemistry, and Bioengineering, Lehigh University, Bethlehem, PA 18015, United States
| | - Howard C. Hang
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA 92037, United States
- Department of Chemistry, Scripps Research, La Jolla, CA 92037, United States
| |
Collapse
|
16
|
Jiang XC, Zhang T, Gao JQ. The in vivo fate and targeting engineering of crossover vesicle-based gene delivery system. Adv Drug Deliv Rev 2022; 187:114324. [PMID: 35640803 DOI: 10.1016/j.addr.2022.114324] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 04/22/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022]
Abstract
Exosomes and biomimetic vesicles are widely used for gene delivery because of their excellent gene loading capacity and stability and their natural targeting delivery potential. These vesicles take advantages of both cell-based bioactive delivery system and synthetical lipid-derived nanovectors to form crossover characteristics. To further optimize the specific targeting properties of crossover vesicles, studies of their in vivo fate and various engineering approaches including nanobiotechnology are required. This review describes the preparation process of exosomes and biomimetic vesicles, and summarizes the mechanism of loading and delivery of nucleic acids or gene editing systems. We provide a comprehensive overview of the techniques employed for preparing the targeting crossover vesicles based on their cellular uptake and targeting mechanism. To delineate the future prospects of crossover vesicle gene delivery systems, various challenges and clinical applications of vesicles have also been discussed.
Collapse
|
17
|
Fonnesu R, Thunuguntla VBSC, Veeramachaneni GK, Bondili JS, La Rocca V, Filipponi C, Spezia PG, Sidoti M, Plicanti E, Quaranta P, Freer G, Pistello M, Mathai ML, Lai M. Palmitoylethanolamide (PEA) Inhibits SARS-CoV-2 Entry by Interacting with S Protein and ACE-2 Receptor. Viruses 2022; 14:1080. [PMID: 35632821 PMCID: PMC9146540 DOI: 10.3390/v14051080] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 05/16/2022] [Accepted: 05/16/2022] [Indexed: 01/08/2023] Open
Abstract
Lipids play a crucial role in the entry and egress of viruses, regardless of whether they are naked or enveloped. Recent evidence shows that lipid involvement in viral infection goes much further. During replication, many viruses rearrange internal lipid membranes to create niches where they replicate and assemble. Because of the close connection between lipids and inflammation, the derangement of lipid metabolism also results in the production of inflammatory stimuli. Due to its pivotal function in the viral life cycle, lipid metabolism has become an area of intense research to understand how viruses seize lipids and to design antiviral drugs targeting lipid pathways. Palmitoylethanolamide (PEA) is a lipid-derived peroxisome proliferator-activated receptor-α (PPAR-α) agonist that also counteracts SARS-CoV-2 entry and its replication. Our work highlights for the first time the antiviral potency of PEA against SARS-CoV-2, exerting its activity by two different mechanisms. First, its binding to the SARS-CoV-2 S protein causes a drop in viral infection of ~70%. We show that this activity is specific for SARS-CoV-2, as it does not prevent infection by VSV or HSV-2, other enveloped viruses that use different glycoproteins and entry receptors to mediate their entry. Second, we show that in infected Huh-7 cells, treatment with PEA dismantles lipid droplets, preventing the usage of these vesicular bodies by SARS-CoV-2 as a source of energy and protection against innate cellular defenses. This is not surprising since PEA activates PPAR-α, a transcription factor that, once activated, generates a cascade of events that leads to the disruption of fatty acid droplets, thereby bringing about lipid droplet degradation through β-oxidation. In conclusion, the present work demonstrates a novel mechanism of action for PEA as a direct and indirect antiviral agent against SARS-CoV-2. This evidence reinforces the notion that treatment with this compound might significantly impact the course of COVID-19. Indeed, considering that the protective effects of PEA in COVID-19 are the current objectives of two clinical trials (NCT04619706 and NCT04568876) and given the relative lack of toxicity of PEA in humans, further preclinical and clinical tests will be needed to fully consider PEA as a promising adjuvant therapy in the current COVID-19 pandemic or against emerging RNA viruses that share the same route of replication as coronaviruses.
Collapse
Affiliation(s)
- Rossella Fonnesu
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | | | - Ganesh Kumar Veeramachaneni
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522502, India; (G.K.V.); (J.S.B.)
| | - Jayakumar Singh Bondili
- Department of Biotechnology, Koneru Lakshmaiah Education Foundation, Vaddeswaram 522502, India; (G.K.V.); (J.S.B.)
| | - Veronica La Rocca
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Carolina Filipponi
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Pietro Giorgio Spezia
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Maria Sidoti
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Erika Plicanti
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Paola Quaranta
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Giulia Freer
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Mauro Pistello
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| | - Michael Lee Mathai
- Institute of Health and Sport, Victoria University, Melbourne, VIC 8001, Australia; (V.B.S.C.T.); (M.L.M.)
| | - Michele Lai
- Retrovirus Center, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56100 Pisa, Italy; (R.F.); (V.L.R.); (C.F.); (P.G.S.); (M.S.); (E.P.); (P.Q.); (G.F.); (M.P.)
| |
Collapse
|
18
|
LoPresti ST, Arral ML, Chaudhary N, Whitehead KA. The replacement of helper lipids with charged alternatives in lipid nanoparticles facilities targeted mRNA delivery to the spleen and lungs. J Control Release 2022; 345:819-831. [PMID: 35346768 PMCID: PMC9447088 DOI: 10.1016/j.jconrel.2022.03.046] [Citation(s) in RCA: 131] [Impact Index Per Article: 43.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 03/21/2022] [Accepted: 03/23/2022] [Indexed: 12/30/2022]
Abstract
The broad clinical application of mRNA therapeutics has been hampered by a lack of delivery vehicles that induce protein expression in extrahepatic organs and tissues. Recently, it was shown that mRNA delivery to the spleen or lungs is possible upon the addition of a charged lipid to a standard four-component lipid nanoparticle formulation. This approach, while effective, further complicates an already complex drug formulation and has the potential to slow regulatory approval and adversely impact manufacturing processes. We were thus motivated to maintain a four-component nanoparticle system while achieving shifts in tropism. To that end, we replaced the standard helper lipid in lipidoid nanoparticles, DOPE, with one of eight alternatives. These lipids included the neutral lipids, DOPC, sphingomyelin, and ceramide; the anionic lipids, phosphatidylserine (PS), phosphatidylglycerol, and phosphatidic acid; and the cationic lipids, DOTAP and ethyl phosphatidylcholine. While neutral helper lipids maintained protein expression in the liver, anionic and cationic lipids shifted protein expression to the spleen and lungs, respectively. For example, replacing DOPE with DOTAP increased positive LNP surface charge at pH 7 by 5-fold and altered the ratio of liver to lung protein expression from 36:1 to 1:56. Similarly, replacing DOPE with PS reduced positive charge by half and altered the ratio of liver to spleen protein expression from 8:1 to 1:3. Effects were consistent across ionizable lipidoid chemistries. Regarding mechanism, nanoparticles formulated with neutral and anionic helper lipids best transfected epithelial and immune cells, respectively. Further, the lung-tropic effect of DOTAP was linked to reduced immune cell infiltration of the lungs compared to neutral or anionic lipids. Together, these data show that intravenous non-hepatocellular mRNA delivery is readily achievable while maintaining a four-component formulation with modified helper lipid chemistry.
Collapse
Affiliation(s)
- Samuel T LoPresti
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Mariah L Arral
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Namit Chaudhary
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America
| | - Kathryn A Whitehead
- Department of Chemical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America; Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States of America.
| |
Collapse
|
19
|
Pizzato M, Baraldi C, Boscato Sopetto G, Finozzi D, Gentile C, Gentile MD, Marconi R, Paladino D, Raoss A, Riedmiller I, Ur Rehman H, Santini A, Succetti V, Volpini L. SARS-CoV-2 and the Host Cell: A Tale of Interactions. FRONTIERS IN VIROLOGY 2022. [DOI: 10.3389/fviro.2021.815388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The ability of a virus to spread between individuals, its replication capacity and the clinical course of the infection are macroscopic consequences of a multifaceted molecular interaction of viral components with the host cell. The heavy impact of COVID-19 on the world population, economics and sanitary systems calls for therapeutic and prophylactic solutions that require a deep characterization of the interactions occurring between virus and host cells. Unveiling how SARS-CoV-2 engages with host factors throughout its life cycle is therefore fundamental to understand the pathogenic mechanisms underlying the viral infection and to design antiviral therapies and prophylactic strategies. Two years into the SARS-CoV-2 pandemic, this review provides an overview of the interplay between SARS-CoV-2 and the host cell, with focus on the machinery and compartments pivotal for virus replication and the antiviral cellular response. Starting with the interaction with the cell surface, following the virus replicative cycle through the characterization of the entry pathways, the survival and replication in the cytoplasm, to the mechanisms of egress from the infected cell, this review unravels the complex network of interactions between SARS-CoV-2 and the host cell, highlighting the knowledge that has the potential to set the basis for the development of innovative antiviral strategies.
Collapse
|
20
|
Das T, Yang X, Lee H, Garst E, Valencia E, Chandran K, Im W, Hang H. S-palmitoylation and sterol interactions mediate antiviral specificity of IFITM isoforms. RESEARCH SQUARE 2021:rs.3.rs-1179000. [PMID: 34981045 PMCID: PMC8722608 DOI: 10.21203/rs.3.rs-1179000/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Interferon-induced transmembrane proteins (IFITM1, 2 and 3) are important antiviral proteins that are active against many viruses, including influenza A virus (IAV), dengue virus (DENV), Ebola virus (EBOV), Zika virus (ZIKV) and severe acute respiratory syndrome coronavirus (SARS-CoV). IFITMs exhibit isoform-specific activity, but their distinct mechanisms of action and regulation are unclear. Since S -palmitoylation and cholesterol homeostasis are crucial for viral infections, we investigated IFITM interactions with cholesterol by molecular dynamic stimulations, nuclear magnetic resonance analysis in vitro and photoaffinity crosslinking in mammalian cells. These studies suggest that cholesterol can alter the conformation of IFITMs in membrane bilayers and directly interact with S -palmitoylated IFITMs in cells. Notably, we discovered that the S -palmitoylation levels regulate differential IFITM isoform interactions with cholesterol in mammalian cells and specificity of antiviral activity towards IAV, SARS-CoV-2 and EBOV. Our studies suggest that modulation of IFITM S -palmitoylation levels and cholesterol interaction may influence host susceptibility to different viruses.
Collapse
Affiliation(s)
| | | | | | | | | | - Kartik Chandran
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York
| | | | | |
Collapse
|
21
|
Theken KN, Tang SY, Sengupta S, FitzGerald GA. The roles of lipids in SARS-CoV-2 viral replication and the host immune response. J Lipid Res 2021; 62:100129. [PMID: 34599996 PMCID: PMC8480132 DOI: 10.1016/j.jlr.2021.100129] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 09/01/2021] [Accepted: 09/02/2021] [Indexed: 02/06/2023] Open
Abstract
The significant morbidity and mortality associated with severe acute respiratory syndrome coronavirus 2 infection has underscored the need for novel antiviral strategies. Lipids play essential roles in the viral life cycle. The lipid composition of cell membranes can influence viral entry by mediating fusion or affecting receptor conformation. Upon infection, viruses can reprogram cellular metabolism to remodel lipid membranes and fuel the production of new virions. Furthermore, several classes of lipid mediators, including eicosanoids and sphingolipids, can regulate the host immune response to viral infection. Here, we summarize the existing literature on the mechanisms through which these lipid mediators may regulate viral burden in COVID-19. Furthermore, we define the gaps in knowledge and identify the core areas in which lipids offer therapeutic promise for severe acute respiratory syndrome coronavirus 2.
Collapse
Affiliation(s)
- Katherine N Theken
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Oral Surgery and Pharmacology, University of Pennsylvania School of Dental Medicine, Philadelphia, PA, USA
| | - Soon Yew Tang
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Shaon Sengupta
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Garret A FitzGerald
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA; Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
22
|
Tran A, Monreal IA, Moskovets E, Aguilar HC, Jones JW. Rapid Detection of Viral Envelope Lipids Using Lithium Adducts and AP-MALDI High-Resolution Mass Spectrometry. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:2322-2333. [PMID: 33886294 PMCID: PMC8995026 DOI: 10.1021/jasms.1c00058] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
There is an unmet need to develop analytical strategies that not only characterize the lipid composition of the viral envelope but also do so on a time scale that would allow for high-throughput analysis. With that in mind, we report the use of atmospheric pressure (AP) matrix-assisted laser desorption/ionization (MALDI) high-resolution mass spectrometry (HRMS) combined with lithium adduct consolidation to profile total lipid extracts rapidly and confidently from enveloped viruses. The use of AP-MALDI reduced the dependency of using a dedicated MALDI mass spectrometer and allowed for interfacing the MALDI source to a mass spectrometer with the desired features, which included high mass resolving power (>100000) and tandem mass spectrometry. AP-MALDI combined with an optimized MALDI matrix system, featuring 2',4',6'-trihydroxyacetophenone spiked with lithium salt, resulted in a robust and high-throughput lipid detection platform, specifically geared to sphingolipid detection. Application of the developed workflow included the structural characterization of prominent sphingolipids and detection of over 130 lipid structures from Influenza A virions. Overall, we demonstrate a high-throughput workflow for the detection and structural characterization of total lipid extracts from enveloped viruses using AP-MALDI HRMS and lithium adduct consolidation.
Collapse
Affiliation(s)
- Anh Tran
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - I Abrrey Monreal
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | | | - Hector C Aguilar
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York 14853, United States
| | - Jace W Jones
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
23
|
Moreira P, Sequeira AM, Pereira S, Rodrigues R, Rocha M, Lousa D. ViralFP: A Web Application of Viral Fusion Proteins. FRONTIERS IN MEDICAL TECHNOLOGY 2021; 3:722392. [PMID: 35047951 PMCID: PMC8757699 DOI: 10.3389/fmedt.2021.722392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/19/2021] [Indexed: 12/03/2022] Open
Abstract
Viral fusion proteins are attached to the membrane of enveloped viruses (a group that includes Coronaviruses, Dengue, HIV and Influenza) and catalyze fusion between the viral and host membranes, enabling the virus to insert its genetic material into the host cell. Given the importance of these biomolecules, this work presents a centralized database containing the most relevant information on viral fusion proteins, available through a free-to-use web server accessible through the URL https://viralfp.bio.di.uminho.pt/. This web application contains several bioinformatic tools, such as Clustal sequence alignment and Weblogo, including as well a machine learning-based tool capable of predicting the location of fusion peptides (the component of fusion proteins that inserts into the host's cell membrane) within the fusion protein sequence. Given the crucial role of these proteins in viral infection, their importance as natural targets of our immune system and their potential as therapeutic targets, this web application aims to foster our ability to fight pathogenic viruses.
Collapse
Affiliation(s)
- Pedro Moreira
- Centro de Engenharia Biológica, Escola de Engenharia da Universidade do Minho, Braga, Portugal
| | - Ana Marta Sequeira
- Centro de Engenharia Biológica, Escola de Engenharia da Universidade do Minho, Braga, Portugal
| | - Sara Pereira
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Rúben Rodrigues
- Centro de Engenharia Biológica, Escola de Engenharia da Universidade do Minho, Braga, Portugal
| | - Miguel Rocha
- Centro de Engenharia Biológica, Escola de Engenharia da Universidade do Minho, Braga, Portugal
| | - Diana Lousa
- ITQB NOVA, Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
24
|
Fu PF, Cheng X, Su BQ, Duan LF, Wang CR, Niu XR, Wang J, Yang GY, Chu BB. CRISPR/Cas9-based generation of a recombinant double-reporter pseudorabies virus and its characterization in vitro and in vivo. Vet Res 2021; 52:95. [PMID: 34174954 PMCID: PMC8233574 DOI: 10.1186/s13567-021-00964-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 05/27/2021] [Indexed: 11/24/2022] Open
Abstract
Pseudorabies, caused by pseudorabies virus (PRV) variants, has broken out among commercial PRV vaccine-immunized swine herds and resulted in major economic losses to the pig industry in China since late 2011. However, the mechanism of virulence enhancement of variant PRV is currently unclear. Here, a recombinant PRV (rPRV HN1201-EGFP-Luc) with stable expression of enhanced green fluorescent protein (EGFP) and firefly luciferase as a double reporter virus was constructed on the basis of the PRV variant HN1201 through CRISPR/Cas9 gene-editing technology coupled with two sgRNAs. The biological characteristics of the recombinant virus and its lethality to mice were similar to those of the parental strain and displayed a stable viral titre and luciferase activity through 20 passages. Moreover, bioluminescence signals were detected in mice at 12 h after rPRV HN1201-EGFP-Luc infection. Using the double reporter PRV, we also found that 25-hydroxycholesterol had a significant inhibitory effect on PRV both in vivo and in vitro. These results suggested that the double reporter PRV based on PRV variant HN1201 should be an excellent tool for basic virology studies and evaluating antiviral agents.
Collapse
Affiliation(s)
- Peng-Fei Fu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Xuan Cheng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Bing-Qian Su
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Li-Fang Duan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Cong-Rong Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Xin-Rui Niu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China
| | - Guo-Yu Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China.
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, 450046, Henan, China.
| |
Collapse
|
25
|
Current Understanding of the Role of Cholesterol in the Life Cycle of Alphaviruses. Viruses 2020; 13:v13010035. [PMID: 33383613 PMCID: PMC7823518 DOI: 10.3390/v13010035] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 12/28/2022] Open
Abstract
Enveloped viruses rely on different lipid classes present in cell membranes to accomplish several steps of their life cycle in the host. Particularly for alphaviruses, a medically important group of arboviruses, which are part of the Togaviridae family, cholesterol seems to be a critical lipid exploited during infection, although its relevance may vary depending on which stage of the virus life cycle is under consideration and whether infection takes place in vertebrate or invertebrate hosts. In this review, the role of cholesterol in both early and late events of alphavirus infection and how viral replication may affect cholesterol metabolism are summarized, taking into account studies on Old World and New World alphaviruses in different cell lines. Moreover, the importance of cholesterol for the structural stability of alphavirus particles is also discussed, shedding light on the role played by this lipid when they leave the host cell.
Collapse
|
26
|
Mavri M, Spiess K, Rosenkilde MM, Rutland CS, Vrecl M, Kubale V. Methods for Studying Endocytotic Pathways of Herpesvirus Encoded G Protein-Coupled Receptors. Molecules 2020; 25:E5710. [PMID: 33287269 PMCID: PMC7730005 DOI: 10.3390/molecules25235710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Endocytosis is a fundamental process involved in trafficking of various extracellular and transmembrane molecules from the cell surface to its interior. This enables cells to communicate and respond to external environments, maintain cellular homeostasis, and transduce signals. G protein-coupled receptors (GPCRs) constitute a family of receptors with seven transmembrane alpha-helical domains (7TM receptors) expressed at the cell surface, where they regulate physiological and pathological cellular processes. Several herpesviruses encode receptors (vGPCRs) which benefits the virus by avoiding host immune surveillance, supporting viral dissemination, and thereby establishing widespread and lifelong infection, processes where receptor signaling and/or endocytosis seem central. vGPCRs are rising as potential drug targets as exemplified by the cytomegalovirus-encoded receptor US28, where its constitutive internalization has been exploited for selective drug delivery in virus infected cells. Therefore, studying GPCR trafficking is of great importance. This review provides an overview of the current knowledge of endocytic and cell localization properties of vGPCRs and methodological approaches used for studying receptor internalization. Using such novel approaches, we show constitutive internalization of the BILF1 receptor from human and porcine γ-1 herpesviruses and present motifs from the eukaryotic linear motif (ELM) resources with importance for vGPCR endocytosis.
Collapse
Affiliation(s)
- Maša Mavri
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Catrin Sian Rutland
- School of Veterinary Medicine and Science, Medical Faculty, Sutton, Bonington Campus, University of Nottingham, Sutton Bonington LE12 5RD, UK;
| | - Milka Vrecl
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
| | - Valentina Kubale
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
| |
Collapse
|
27
|
Zhu F, Song D, Chen H, Tang Q, Huo S, Liu X, Chen K. A Lipidome Map of the Silkworm Bombyx mori: Influences of Viral Infection. J Proteome Res 2020; 20:695-703. [PMID: 33175548 DOI: 10.1021/acs.jproteome.0c00608] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Lipids have been recently proposed as key molecules for virus entry and egress, and lipid biosynthesis and signaling were reported necessary for some viruses during replication and infection. The silkworm Bombyx mori is an important economic insect and a model organism, but its lipid profiles have not been systematically investigated. Most silkworm strains are susceptible to the B. mori nuclear polyhedrovirus (BmNPV), a baculovirus that causes serious loss to the sericulture industry. Previously, our lab has screened a natural mutant of B. mori that is highly resistant to BmNPV. In this study, a comprehensive lipidomic analysis by ultrahigh pressure liquid chromatography-mass spectrometry (UPLC-MS) was carried out on the BmNPV-susceptible strain 306 and resistant strain NB (data deposited in MetaboLight MTBLS2142). Comparisons of the lipid profiles between the two strains reveal that phosphosphingolipids, diacylglycerolipids, ceramides, and quinones were present at notably higher levels in the susceptible strain, while lysophosphocholines were found at a higher level in the resistant strain. BmNPV administration changed the lipid profiles in both strains, revealing key lipids involved in virus infection and immune response. Some key enzymes in the lipid biosynthesis pathway were analyzed for their activities in the two silkworm strains and their virus-administered counterparts, underlining the relation among lipid biosynthesis, viral resistance, and immune response in the host.
Collapse
|
28
|
A Broad-Spectrum Antiviral Peptide Blocks Infection of Viruses by Binding to Phosphatidylserine in the Viral Envelope. Cells 2020; 9:cells9091989. [PMID: 32872420 PMCID: PMC7563927 DOI: 10.3390/cells9091989] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 08/26/2020] [Accepted: 08/27/2020] [Indexed: 01/04/2023] Open
Abstract
The ongoing threat of viral infections and the emergence of antiviral drug resistance warrants a ceaseless search for new antiviral compounds. Broadly-inhibiting compounds that act on elements shared by many viruses are promising antiviral candidates. Here, we identify a peptide derived from the cowpox virus protein CPXV012 as a broad-spectrum antiviral peptide. We found that CPXV012 peptide hampers infection by a multitude of clinically and economically important enveloped viruses, including poxviruses, herpes simplex virus-1, hepatitis B virus, HIV-1, and Rift Valley fever virus. Infections with non-enveloped viruses such as Coxsackie B3 virus and adenovirus are not affected. The results furthermore suggest that viral particles are neutralized by direct interactions with CPXV012 peptide and that this cationic peptide may specifically bind to and disrupt membranes composed of the anionic phospholipid phosphatidylserine, an important component of many viral membranes. The combined results strongly suggest that CPXV012 peptide inhibits virus infections by direct interactions with phosphatidylserine in the viral envelope. These results reiterate the potential of cationic peptides as broadly-acting virus inhibitors.
Collapse
|
29
|
Multifaceted Functions of CH25H and 25HC to Modulate the Lipid Metabolism, Immune Responses, and Broadly Antiviral Activities. Viruses 2020; 12:v12070727. [PMID: 32640529 PMCID: PMC7411728 DOI: 10.3390/v12070727] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 07/03/2020] [Indexed: 12/12/2022] Open
Abstract
With the frequent outbreaks of emerging infectious diseases in recent years, an effective broad-spectrum antiviral drug is becoming an urgent need for global public health. Cholesterol-25-hydroxylase (CH25H) and its enzymatic products 25-hydroxycholesterol (25HC), a well-known oxysterol that regulates lipid metabolism, have been reported to play multiple functions in modulating cholesterol homeostasis, inflammation, and immune responses. CH25H and 25HC were recently identified as exerting broadly antiviral activities, including upon a variety of highly pathogenic viruses such as human immunodeficiency virus (HIV), Ebola virus (EBOV), Nipah virus (NiV), Rift Valley fever virus (RVFV), and Zika virus (ZIKV). The underlying mechanisms for its antiviral activities are being extensively investigated but have not yet been fully clarified. In this study, we summarized the current findings on how CH25H and 25HC play multiple roles to modulate cholesterol metabolism, inflammation, immunity, and antiviral infections. Overall, 25HC should be further studied as a potential therapeutic agent to control emerging infectious diseases in the future.
Collapse
|
30
|
Milasan A, Farhat M, Martel C. Extracellular Vesicles as Potential Prognostic Markers of Lymphatic Dysfunction. Front Physiol 2020; 11:476. [PMID: 32523544 PMCID: PMC7261898 DOI: 10.3389/fphys.2020.00476] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/20/2020] [Indexed: 12/21/2022] Open
Abstract
Despite significant efforts made to treat cardiovascular disease (CVD), more than half of cardiovascular events still occur in asymptomatic subjects devoid of traditional risk factors. These observations underscore the need for the identification of new biomarkers for the prevention of atherosclerosis, the main underlying cause of CVD. Extracellular vesicles (EVs) and lymphatic vessel function are emerging targets in this context. EVs are small vesicles released by cells upon activation or death that are present in several biological tissues and fluids, including blood and lymph. They interact with surrounding cells to transfer their cargo, and the complexity of their biological content makes these EVs potential key players in several chronic inflammatory settings. Many studies focused on the interaction of EVs with the most well-known players of atherosclerosis such as the vascular endothelium, smooth muscle cells and monocytes. However, the fate of EVs within the lymphatic network, a crucial route in the mobilization of cholesterol out the artery wall, is not known. In this review, we aim to bring forward evidence that EVs could be at the interplay between lymphatic function and atherosclerosis by summarizing the recent findings on the characterization of EVs in this setting.
Collapse
Affiliation(s)
- Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Montreal Heart Institute, Montreal, QC, Canada
| | - Maya Farhat
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Montreal Heart Institute, Montreal, QC, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Montreal Heart Institute, Montreal, QC, Canada
| |
Collapse
|
31
|
Labyrinthopeptins Exert Broad-Spectrum Antiviral Activity through Lipid-Binding-Mediated Virolysis. J Virol 2020; 94:JVI.01471-19. [PMID: 31666384 DOI: 10.1128/jvi.01471-19] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/23/2019] [Indexed: 12/31/2022] Open
Abstract
To counteract the serious health threat posed by known and novel viral pathogens, drugs that target a variety of viruses through a common mechanism have attracted recent attention due to their potential in treating (re)emerging infections, for which direct-acting antivirals are not available. We found that labyrinthopeptins A1 and A2, the prototype congeners of carbacyclic lanthipeptides, inhibit the proliferation of diverse enveloped viruses, including dengue virus, Zika virus, West Nile virus, hepatitis C virus, chikungunya virus, Kaposi's sarcoma-associated herpesvirus, cytomegalovirus, and herpes simplex virus, in the low micromolar to nanomolar range. Mechanistic studies on viral particles revealed that labyrinthopeptins induce a virolytic effect through binding to the viral membrane lipid phosphatidylethanolamine (PE). These effects are enhanced by a combined equimolar application of both labyrinthopeptins, and a clear synergism was observed across a concentration range corresponding to 10% to 90% inhibitory concentrations of the compounds. Time-resolved experiments with large unilamellar vesicles (LUVs) reveal that membrane lipid raft compositions (phosphatidylcholine [PC]/PE/cholesterol/sphingomyelin at 17:10:33:40) are particularly sensitive to labyrinthopeptins in comparison to PC/PE (90:10) LUVs, even though the overall PE amount remains constant. Labyrinthopeptins exhibited low cytotoxicity and had favorable pharmacokinetic properties in mice (half-life [t 1/2] = 10.0 h), which designates them promising antiviral compounds acting by an unusual viral lipid targeting mechanism.IMPORTANCE For many viral infections, current treatment options are insufficient. Because the development of each antiviral drug is time-consuming and expensive, the prospect of finding broad-spectrum antivirals that can fight multiple, diverse viruses-well-known viruses as well as (re)emerging species-has gained attention, especially for the treatment of viral coinfections. While most known broad-spectrum agents address processes in the host cell, we found that targeting lipids of the free virus outside the host cell with the natural products labyrinthopeptin A1 and A2 is a viable strategy to inhibit the proliferation of a broad range of viruses from different families, including chikungunya virus, dengue virus, Zika virus, Kaposi's sarcoma-associated herpesvirus, and cytomegalovirus. Labyrinthopeptins bind to viral phosphatidylethanolamine and induce virolysis without exerting cytotoxicity on host cells. This represents a novel and unusual mechanism to tackle medically relevant viral infections.
Collapse
|
32
|
Dong H, Zhou L, Ge X, Guo X, Han J, Yang H. Antiviral effect of 25-hydroxycholesterol against porcine reproductive and respiratory syndrome virus in vitro. Antivir Ther 2019; 23:395-404. [PMID: 29561734 DOI: 10.3851/imp3232] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/14/2018] [Indexed: 10/17/2022]
Abstract
BACKGROUND Porcine reproductive and respiratory syndrome virus (PRRSV) is an important pathogen that causes economically huge losses to the pig industry worldwide. Current control of PRRSV infection remains inadequate although various means have been implemented. Thus, investigating novel antiviral therapeutics to combat PRRSV infection is essential. In the present study, the antiviral effect in vitro of 25-hydroxycholesterol (25HC) against PRRSV was investigated. METHODS Cell viability assay was performed to examine the impact of 25HC on the cell viability. Indirect immunofluorescence assay and virus titration were utilized to evaluate the levels of PRRSV growth. Viral attachment assay, penetration assay and release assay were conducted to investigate the antiviral mechanism of 25HC against PRRSV. Real-time RT-PCR assay was used to analyse the effect of 25HC on the genome synthesis of PRRSV. RESULTS We demonstrated that the growth of PRRSV was significantly inhibited in 25HC-pretreated cells and PRRSV-infected cells by 25HC. Moreover, 25HC could impair the attachment and entry of PRRSV in vitro, but not affect viral genome synthesis and virion release. CONCLUSIONS Our findings clearly indicate that 25HC can exert antiviral effect against PRRSV infection in vitro, suggesting that 25HC might be a novel potential agent to control PRRSV infection.
Collapse
Affiliation(s)
- Hong Dong
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Lei Zhou
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Xinna Ge
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Xin Guo
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Jun Han
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| | - Hanchun Yang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing, People's Republic of China.,State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Corradi V, Sejdiu BI, Mesa-Galloso H, Abdizadeh H, Noskov SY, Marrink SJ, Tieleman DP. Emerging Diversity in Lipid-Protein Interactions. Chem Rev 2019; 119:5775-5848. [PMID: 30758191 PMCID: PMC6509647 DOI: 10.1021/acs.chemrev.8b00451] [Citation(s) in RCA: 291] [Impact Index Per Article: 48.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Indexed: 02/07/2023]
Abstract
Membrane lipids interact with proteins in a variety of ways, ranging from providing a stable membrane environment for proteins to being embedded in to detailed roles in complicated and well-regulated protein functions. Experimental and computational advances are converging in a rapidly expanding research area of lipid-protein interactions. Experimentally, the database of high-resolution membrane protein structures is growing, as are capabilities to identify the complex lipid composition of different membranes, to probe the challenging time and length scales of lipid-protein interactions, and to link lipid-protein interactions to protein function in a variety of proteins. Computationally, more accurate membrane models and more powerful computers now enable a detailed look at lipid-protein interactions and increasing overlap with experimental observations for validation and joint interpretation of simulation and experiment. Here we review papers that use computational approaches to study detailed lipid-protein interactions, together with brief experimental and physiological contexts, aiming at comprehensive coverage of simulation papers in the last five years. Overall, a complex picture of lipid-protein interactions emerges, through a range of mechanisms including modulation of the physical properties of the lipid environment, detailed chemical interactions between lipids and proteins, and key functional roles of very specific lipids binding to well-defined binding sites on proteins. Computationally, despite important limitations, molecular dynamics simulations with current computer power and theoretical models are now in an excellent position to answer detailed questions about lipid-protein interactions.
Collapse
Affiliation(s)
- Valentina Corradi
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Besian I. Sejdiu
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Haydee Mesa-Galloso
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Haleh Abdizadeh
- Groningen
Biomolecular Sciences and Biotechnology Institute and Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - Sergei Yu. Noskov
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| | - Siewert J. Marrink
- Groningen
Biomolecular Sciences and Biotechnology Institute and Zernike Institute
for Advanced Materials, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands
| | - D. Peter Tieleman
- Centre
for Molecular Simulation and Department of Biological Sciences, University of Calgary, 2500 University Drive NW, Calgary, Alberta T2N 1N4, Canada
| |
Collapse
|
34
|
3D visualization of extracellular vesicle uptake by endothelial cells. Cell Mol Biol Lett 2018; 23:57. [PMID: 30574165 PMCID: PMC6296015 DOI: 10.1186/s11658-018-0123-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 11/28/2018] [Indexed: 12/13/2022] Open
Abstract
Background Extracellular vesicles are small vesicles that contain cytoplasmic and membrane components from their paternal cells. They enter target cells through uptake to transfer their biological cargo. In this study, we investigated the process of endothelial EV internalization and created a 3D visualization of their intracellular distribution. Methods and results Two immortalized endothelial cell lines that express h-TERT (human telomerase) were used for EV release: microvascular TIME and macrovascular HUVEC. EVs were isolated from the cell culture medium via differential centrifugation and used for the uptake experiments. The size distribution of the EVs was measured using TRPS technology on a qNano instrument. Internalization of EVs was observed using a Zeiss LSM 710 confocal laser microscope after staining of the EVs with PKH26. EVs were observed intracellularly and distributed in the perinuclear region of the target cells. The distribution patterns were similar in both cell lines. Conclusion The perinuclear localization of the internalized EVs shows their biological stability after their uptake to the endothelial cells. The 3D visualization allows the determination of a more accurate location of EVs relative to the donor cell nucleus. Electronic supplementary material The online version of this article (10.1186/s11658-018-0123-z) contains supplementary material, which is available to authorized users.
Collapse
|
35
|
25-Hydroxycholesterol Production by the Cholesterol-25-Hydroxylase Interferon-Stimulated Gene Restricts Mammalian Reovirus Infection. J Virol 2018; 92:JVI.01047-18. [PMID: 29950420 DOI: 10.1128/jvi.01047-18] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 06/23/2018] [Indexed: 11/20/2022] Open
Abstract
Following the initial detection of viral infection, innate immune responses trigger the induction of numerous interferon-stimulated genes (ISGs) to inhibit virus replication and dissemination. One such ISG encodes cholesterol-25-hydroxylase (CH25H), an enzyme that catalyzes the oxidation of cholesterol to form a soluble product, 25-hydroxycholesterol (25HC). Recent studies have found that CH25H is broadly antiviral; it inhibits infection by several viruses. For enveloped viruses, 25HC inhibits membrane fusion, likely by altering membrane characteristics such as hydrophobicity or cholesterol aggregation. However, the mechanisms by which 25HC restricts infection of nonenveloped viruses are unknown. We examined whether 25HC restricts infection by mammalian reovirus. Treatment with 25HC restricted infection by reovirus prototype strains type 1 Lang and type 3 Dearing. In contrast to reovirus virions, 25HC did not restrict infection by reovirus infectious subvirion particles (ISVPs), which can penetrate either directly at the cell surface or in early endosomal membranes. Treatment with 25HC altered trafficking of reovirus particles to late endosomes and delayed the kinetics of reovirus uncoating. These results suggest that 25HC inhibits the efficiency of cellular entry of reovirus virions, which may require specific endosomal membrane dynamics for efficient membrane penetration.IMPORTANCE The innate immune system is crucial for effective responses to viral infection. Type I interferons, central components of innate immunity, induce expression of hundreds of ISGs; however, the mechanisms of action of these antiviral proteins are not well understood. CH25H, encoded by an ISG, represents a significant constituent of these cellular antiviral strategies, as its metabolic product, 25HC, can act in both an autocrine and a paracrine fashion to protect cells from infection and has been shown to limit viral infection in animal models. Further investigation into the mechanism of action of 25HC may inform novel antiviral therapies and influence the use of mammalian reovirus in clinical trials as an oncolytic agent.
Collapse
|
36
|
Mrowczynski OD, Zacharia BE, Connor JR. Exosomes and their implications in central nervous system tumor biology. Prog Neurobiol 2018; 172:71-83. [PMID: 30003942 DOI: 10.1016/j.pneurobio.2018.06.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 05/04/2018] [Accepted: 06/30/2018] [Indexed: 01/08/2023]
Abstract
Exosomes are 20-100 nm cellular derived vesicles that upon discovery, were thought to be a form of cellular recycling of intracellular contents. More recently, these vesicles are under investigation for their purported significant roles in intercellular communication in both healthy and diseased states. Herein, we focus on the secretion of exosomes associated with glioblastoma, as most exosome studies on brain tumors have been performed in this tumor type. However, we included exosomes secreted from other forms of brain tumors for comparison as available. Exosomes contain intracellular content that can be transferred to other cells in the tumor or to cells of the immune system and endothelial cells. These recipient cells may subsequently take on oncogenic properties, including therapeutic resistance, cancer progression, and angiogenesis. Genetic components (DNA, RNA and miRNA) of the cell of origin may be included in the secreted exosomes. The presence of genetic material in the exosomes could serve as a biomarker for mutations in tumors, potentially leading to novel treatment strategies. In the last decade, exosomes have been identified as having a major impact on multiple aspects of medicine and tumor biology, and appear to be primed for a critical position in cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Oliver D Mrowczynski
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Brad E Zacharia
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - James R Connor
- Department of Neurosurgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
37
|
Molotkovsky RJ, Alexandrova VV, Galimzyanov TR, Jiménez-Munguía I, Pavlov KV, Batishchev OV, Akimov SA. Lateral Membrane Heterogeneity Regulates Viral-Induced Membrane Fusion during HIV Entry. Int J Mol Sci 2018; 19:ijms19051483. [PMID: 29772704 PMCID: PMC5983600 DOI: 10.3390/ijms19051483] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 05/08/2018] [Accepted: 05/14/2018] [Indexed: 11/16/2022] Open
Abstract
Sphingomyelin- and cholesterol- enriched membrane domains, commonly referred to as “rafts” play a crucial role in a large number of intra- and intercellular processes. Recent experiments suggest that not only the volumetric inhomogeneity of lipid distribution in rafts, but also the arrangement of the 1D boundary between the raft and the surrounding membrane is important for the membrane-associated processes. The reason is that the boundary preferentially recruits different peptides, such as HIV (human immunodeficiency virus) fusion peptide. In the present work, we report a theoretical investigation of mechanisms of influence of the raft boundary arrangement upon virus-induced membrane fusion. We theoretically predict that the raft boundary can act as an attractor for viral fusion peptides, which preferentially distribute into the vicinity of the boundary, playing the role of ‘line active components’ of the membrane (‘linactants’). We have calculated the height of the fusion energy barrier and demonstrated that, in the case of fusion between HIV membrane and the target cell, presence of the raft boundary in the vicinity of the fusion site facilitates fusion. The results we obtained can be further generalized to be applicable to other enveloped viruses.
Collapse
Affiliation(s)
- Rodion J Molotkovsky
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia.
| | - Veronika V Alexandrova
- Faculty of Physics, M.V. Lomonosov Moscow State University, 1-2 Leninskie Gory, 119991 Moscow, Russia.
| | - Timur R Galimzyanov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia.
- Department of Theoretical Physics and Quantum Technologies, National University of Science and Technology "MISiS", 4 Leninskiy Prospekt, 119049 Moscow, Russia.
| | - Irene Jiménez-Munguía
- Department of Engineering of Technological Equipment, National University of Science and Technology "MISiS", 4 Leninskiy Prospekt, 119049 Moscow, Russia.
| | - Konstantin V Pavlov
- Laboratory of Electrophysiology, Federal Clinical Center of Physical-Chemical Medicine of FMBA, 1a Malaya Pirogovskaya Street, 119435 Moscow, Russia.
| | - Oleg V Batishchev
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia.
- Department of Physics of Living Systems, Moscow Institute of Physics and Technology (State University), 9 Institutskiy Lane, Dolgoprudniy, 141700 Moscow Region, Russia.
| | - Sergey A Akimov
- Laboratory of Bioelectrochemistry, A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy Prospekt, 119071 Moscow, Russia.
- Department of Theoretical Physics and Quantum Technologies, National University of Science and Technology "MISiS", 4 Leninskiy Prospekt, 119049 Moscow, Russia.
| |
Collapse
|
38
|
Cellular cholesterol is required for porcine nidovirus infection. Arch Virol 2017; 162:3753-3767. [PMID: 28884395 PMCID: PMC7086867 DOI: 10.1007/s00705-017-3545-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 08/12/2017] [Indexed: 12/14/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) and porcine epidemic diarrhea virus (PEDV) are porcine nidoviruses that are considered emerging and re-emerging viral pathogens of pigs that pose a significant economic threat to the global pork industry. Although cholesterol is known to affect the replication of a broad range of viruses in vitro, its significance and role in porcine nidovirus infection remains to be elucidated. Therefore, the present study was conducted to determine whether cellular or/and viral cholesterol levels play a role in porcine nidovirus infection. Our results showed that depletion of cellular cholesterol by treating cells with methyl-β-cyclodextrin (MβCD) dose-dependently suppressed the replication of both nidoviruses. Conversely, cholesterol depletion from the viral envelope had no inhibitory effect on porcine nidovirus production. The addition of exogenous cholesterol to MβCD-treated cells moderately restored the infectivity of porcine nidoviruses, indicating that the presence of cholesterol in the target cell membrane is critical for viral replication. The antiviral activity of MβCD on porcine nidovirus infection was found to be predominantly exerted when used as a treatment pre-infection or prior to the viral entry process. Furthermore, pharmacological sequestration of cellular cholesterol efficiently blocked both virus attachment and internalization and, accordingly, markedly affected subsequent post-entry steps of the replication cycle, including viral RNA and protein biosynthesis and progeny virus production. Taken together, our data indicate that cell membrane cholesterol is required for porcine nidovirus entry into cells, and pharmacological drugs that hamper cholesterol-dependent virus entry may have antiviral potential against porcine nidoviruses.
Collapse
|
39
|
Imipramine Inhibits Chikungunya Virus Replication in Human Skin Fibroblasts through Interference with Intracellular Cholesterol Trafficking. Sci Rep 2017; 7:3145. [PMID: 28600536 PMCID: PMC5466638 DOI: 10.1038/s41598-017-03316-5] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 04/26/2017] [Indexed: 02/06/2023] Open
Abstract
Chikungunya virus (CHIKV) is an emerging arbovirus of the Togaviridae family that poses a present worldwide threat to human in the absence of any licensed vaccine or antiviral treatment to control viral infection. Here, we show that compounds interfering with intracellular cholesterol transport have the capacity to inhibit CHIKV replication in human skin fibroblasts, a major viral entry site in the human host. Pretreatment of these cells with the class II cationic amphiphilic compound U18666A, or treatment with the FDA-approved antidepressant drug imipramine resulted in a near total inhibition of viral replication and production at the highest concentration used without any cytotoxic effects. Imipramine was found to affect both the fusion and replication steps of the viral life cycle. The key contribution of cholesterol availability to the CHIKV life cycle was validated further by the use of fibroblasts from Niemann-Pick type C (NPC) patients in which the virus was unable to replicate. Interestingly, imipramine also strongly inhibited the replication of several Flaviviridae family members, including Zika, West Nile and Dengue virus. Together, these data show that this compound is a potential drug candidate for anti-arboviral treatment.
Collapse
|
40
|
Caivano A, La Rocca F, Laurenzana I, Trino S, De Luca L, Lamorte D, Del Vecchio L, Musto P. Extracellular Vesicles in Hematological Malignancies: From Biology to Therapy. Int J Mol Sci 2017; 18:E1183. [PMID: 28574430 PMCID: PMC5486006 DOI: 10.3390/ijms18061183] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 12/22/2022] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous group of particles, between 15 nanometers and 10 microns in diameter, released by almost all cell types in physiological and pathological conditions, including tumors. EVs have recently emerged as particularly interesting informative vehicles, so that they could be considered a true "cell biopsy". Indeed, EV cargo, including proteins, lipids, and nucleic acids, generally reflects the nature and status of the origin cells. In some cases, EVs are enriched of peculiar molecular cargo, thus suggesting at least a degree of specific cellular packaging. EVs are identified as important and critical players in intercellular communications in short and long distance interplays. Here, we examine the physiological role of EVs and their activity in cross-talk between bone marrow microenvironment and neoplastic cells in hematological malignancies (HMs). In these diseases, HM EVs can modify tumor and bone marrow microenvironment, making the latter "stronger" in supporting malignancy, inducing drug resistance, and suppressing the immune system. Moreover, EVs are abundant in biologic fluids and protect their molecular cargo against degradation. For these and other "natural" characteristics, EVs could be potential biomarkers in a context of HM liquid biopsy and therapeutic tools. These aspects will be also analyzed in this review.
Collapse
Affiliation(s)
- Antonella Caivano
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Francesco La Rocca
- Laboratory of Clinical Research and Advanced Diagnostics, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| | - Ilaria Laurenzana
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Stefania Trino
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luciana De Luca
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Daniela Lamorte
- Laboratory of Preclinical and Translational Research, IRCCS-Referral Cancer Center of Basilicata (CROB), 858028 Rionero in Vulture, Italy.
| | - Luigi Del Vecchio
- CEINGE-Biotecnologie Avanzate scarl, Federico II University, 80138 Naples, Italy.
- Department of Molecular Medicine and Medical Biotechnologies, Federico II University, 80138 Naples, Italy.
| | - Pellegrino Musto
- Scientific Direction, IRCCS-Referral Cancer Center of Basilicata (CROB), 85028 Rionero in Vulture, Italy.
| |
Collapse
|
41
|
Sezgin E, Levental I, Mayor S, Eggeling C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol 2017; 18:361-374. [PMID: 28356571 PMCID: PMC5500228 DOI: 10.1038/nrm.2017.16] [Citation(s) in RCA: 1351] [Impact Index Per Article: 168.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Cellular plasma membranes are laterally heterogeneous, featuring a variety of distinct subcompartments that differ in their biophysical properties and composition. A large number of studies have focused on understanding the basis for this heterogeneity and its physiological relevance. The membrane raft hypothesis formalized a physicochemical principle for a subtype of such lateral membrane heterogeneity, in which the preferential associations between cholesterol and saturated lipids drive the formation of relatively packed (or ordered) membrane domains that selectively recruit certain lipids and proteins. Recent studies have yielded new insights into this mechanism and its relevance in vivo, owing primarily to the development of improved biochemical and biophysical technologies.
Collapse
Affiliation(s)
- Erdinc Sezgin
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| | - Ilya Levental
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, 6431 Fannin Street, Houston, Texas 77030, USA
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata Institute for Fundamental Research, Bellary Road, Bangalore 560065, India
| | - Christian Eggeling
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Headley Way, Oxford OX3 9DS, UK
| |
Collapse
|
42
|
Li L, Yu L, Hou X. Cholesterol-rich lipid rafts play a critical role in bovine parainfluenza virus type 3 (BPIV3) infection. Res Vet Sci 2017; 114:341-347. [PMID: 28654867 DOI: 10.1016/j.rvsc.2017.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/14/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022]
Abstract
Lipid rafts are specialized lipid domains enriched in cholesterol and sphingolipid, which can be utilized in the lifecycle of numerous enveloped viruses. Bovine parainfluenza virustype3 (BPIV3) entry to cell is mediated by receptor binding and membrane fusion, but how lipid rafts in host cell membrane and BPIV3 envelope affect virus infection remains unclear. In this study, we investigated the role of lipid rafts in the different stages of BPIV3 infection. The MDBK cells were treated by methyl-β-cyclodextrin (MβCD) to disrupt cellular lipid raft, and the virus infection was determined. The results showed that MβCD significantly inhibited BPIV3 infection in a dose-dependent manner, but didn't block the binding of virus to the cell membrane. Whereas, the MDBK cells treated by MβCD after virus-entry had no effects on the virus infection, to suggest that BPIV3 infection was associated with lipid rafts in cell membrane during viral entry stage. To further confirm lipid rafts in viral envelope also affected BPIV3 infection, we treated BPIV3 with MβCD to determine the virus titer. We found that disruption of the viral lipid raft caused a significant reduction of viral yield. Cholesterol reconstitution experiment showed that BPIV3 infection was successfully restored by cholesterol supplementation both in cellular membrane and viral envelope, which demonstrated that cholesterol-rich lipid rafts played a critical role in BPIV3 infection. These findings provide insights on our understanding of the mechanism of BPIV3 infection and imply that lipid raft might be a good potential therapeutic target to prevent virus infection.
Collapse
Affiliation(s)
- Liyang Li
- College of Animal Science and Veterinary Medicine, HeiLongJiang BaYi Agricultural University, Daqing 163319, China; College of Life Science and Technology, HeiLongJiang BaYi Agricultural University, Daqing 163319, China
| | - Liyun Yu
- College of Life Science and Technology, HeiLongJiang BaYi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- College of Animal Science and Veterinary Medicine, HeiLongJiang BaYi Agricultural University, Daqing 163319, China.
| |
Collapse
|
43
|
Klug YA, Rotem E, Schwarzer R, Shai Y. Mapping out the intricate relationship of the HIV envelope protein and the membrane environment. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2016; 1859:550-560. [PMID: 27793589 DOI: 10.1016/j.bbamem.2016.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/20/2016] [Accepted: 10/24/2016] [Indexed: 01/08/2023]
Abstract
The HIV gp160 envelope fusion protein is situated in the viral membrane and mediates virus entry into its host cell. Increasing evidence suggests that virtually all parts of the HIV envelope are structurally and functionally dependent on membranes. Protein-lipid interactions and membrane properties influence the dynamics of a manifold of gp160 biological activities such as membrane fusion, immune suppression and gp160 incorporation into virions during HIV budding and assembly. In the following we will summarize our current understanding of this interdependence between membrane interaction, structural conformation and functionality of the different gp160 domains. This article is part of a Special Issue entitled: Lipid order/lipid defects and lipid-control of protein activity edited by Dirk Schneider.
Collapse
Affiliation(s)
- Yoel A Klug
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Etai Rotem
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Roland Schwarzer
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Yechiel Shai
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
44
|
Martín-Acebes MA, Vázquez-Calvo Á, Saiz JC. Lipids and flaviviruses, present and future perspectives for the control of dengue, Zika, and West Nile viruses. Prog Lipid Res 2016; 64:123-137. [PMID: 27702593 DOI: 10.1016/j.plipres.2016.09.005] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 08/17/2016] [Accepted: 09/16/2016] [Indexed: 02/06/2023]
Abstract
Flaviviruses are emerging arthropod-borne pathogens that cause life-threatening diseases such as yellow fever, dengue, West Nile encephalitis, tick-borne encephalitis, Kyasanur Forest disease, tick-borne encephalitis, or Zika disease. This viral genus groups >50 viral species of small enveloped plus strand RNA virus that are phylogenetically closely related to hepatitis C virus. Importantly, the flavivirus life cycle is intimately associated to host cell lipids. Along this line, flaviviruses rearrange intracellular membranes from the endoplasmic-reticulum of the infected cells to develop adequate platforms for viral replication and particle biogenesis. Moreover, flaviviruses dramatically orchestrate a profound reorganization of the host cell lipid metabolism to create a favorable environment for viral multiplication. Consistently, recent work has shown the importance of specific lipid classes in flavivirus infections. For instances, fatty acid synthesis is linked to viral replication, phosphatidylserine and phosphatidylethanolamine are involved on the entry of flaviviruses, sphingolipids (ceramide and sphingomyelin) play a key role on virus assembly and pathogenesis, and cholesterol is essential for innate immunity evasion in flavivirus-infected cells. Here, we revise the current knowledge on the interactions of the flaviviruses with the cellular lipid metabolism to identify potential targets for future antiviral development aimed to combat these relevant health-threatening pathogens.
Collapse
Affiliation(s)
- Miguel A Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de A Coruña km 7.5, 28040 Madrid, Spain.
| | - Ángela Vázquez-Calvo
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de A Coruña km 7.5, 28040 Madrid, Spain
| | - Juan-Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Carretera de A Coruña km 7.5, 28040 Madrid, Spain
| |
Collapse
|
45
|
Stremersch S, De Smedt SC, Raemdonck K. Therapeutic and diagnostic applications of extracellular vesicles. J Control Release 2016; 244:167-183. [PMID: 27491882 DOI: 10.1016/j.jconrel.2016.07.054] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/24/2016] [Accepted: 07/29/2016] [Indexed: 02/07/2023]
Abstract
During the past two decades, extracellular vesicles (EVs) have been identified as important mediators of intercellular communication, enabling the functional transfer of bioactive molecules from one cell to another. Consequently, it is becoming increasingly clear that these vesicles are involved in many (patho)physiological processes, providing opportunities for therapeutic applications. Moreover, it is known that the molecular composition of EVs reflects the physiological status of the producing cell and tissue, rationalizing their exploitation as biomarkers in various diseases. In this review the composition, biogenesis and diversity of EVs is discussed in a therapeutic and diagnostic context. We describe emerging therapeutic applications, including the use of EVs as drug delivery vehicles and as cell-free vaccines, and reflect on future challenges for clinical translation. Finally, we discuss the use of EVs as a biomarker source and highlight recent studies and clinical successes.
Collapse
Affiliation(s)
- Stephan Stremersch
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Stefaan C De Smedt
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium
| | - Koen Raemdonck
- Laboratory of General Biochemistry and Physical Pharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium.
| |
Collapse
|
46
|
Singaravelu R, O'Hara S, Jones DM, Chen R, Taylor NG, Srinivasan P, Quan C, Roy DG, Steenbergen RH, Kumar A, Lyn RK, Özcelik D, Rouleau Y, Nguyen MA, Rayner KJ, Hobman TC, Tyrrell DL, Russell RS, Pezacki JP. MicroRNAs regulate the immunometabolic response to viral infection in the liver. Nat Chem Biol 2015; 11:988-93. [DOI: 10.1038/nchembio.1940] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 09/11/2015] [Indexed: 12/12/2022]
|
47
|
Ivanova PT, Myers DS, Milne SB, McClaren JL, Thomas PG, Brown HA. Lipid composition of viral envelope of three strains of influenza virus - not all viruses are created equal. ACS Infect Dis 2015; 1:399-452. [PMID: 26448476 DOI: 10.1021/acsinfecdis.5b00040] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
While differences in the rate of virus fusion and budding from the host cell membrane have been correlated with pathogenicity, no systematic study of the contribution of differences in viral envelope composition has previously been attempted. Using rigorous virus purification, marked differences between virions and host were observed. Over 125 phospholipid species have been quantitated for three strains of influenza (HKx31- H3N2, PR8- H1N1, and VN1203- H5N1) grown in eggs. The glycerophospholipid composition of purified virions differs from that of the host or that of typical mammalian cells. Phosphatidylcholine is the major component in most mammalian cell membranes, while in purified virions phosphatidylethanolamine dominates. Due to its effects on membrane curvature, it is likely that the variations in its content are important to viral processing during infection. This integrated method of virion isolation with systematic analysis of glycerophospholipids provides a tool for the assessment of species specific biomarkers of viral pathogenicity.
Collapse
Affiliation(s)
- Pavlina T. Ivanova
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States
| | - David S. Myers
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States
| | - Stephen B. Milne
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States
| | - Jennifer L. McClaren
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee 38105-3678, United States
| | - H. Alex Brown
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232-6600, United States
- Department of Biochemistry, The Vanderbilt
Institute of Chemical Biology, The Vanderbilt Ingram Cancer Center, Vanderbilt University, Nashville, Tennessee 37232-6600, United States
| |
Collapse
|
48
|
Abstract
Chikungunya virus (CHIKV) is a rapidly emerging mosquito-borne alphavirus causing millions of infections in the tropical and subtropical regions of the world. CHIKV infection often leads to an acute self-limited febrile illness with debilitating myalgia and arthralgia. A potential long-term complication of CHIKV infection is severe joint pain, which can last for months to years. There are no vaccines or specific therapeutics available to prevent or treat infection. This review describes the critical steps in CHIKV cell entry. We summarize the latest studies on the virus-cell tropism, virus-receptor binding, internalization, membrane fusion and review the molecules and compounds that have been described to interfere with virus cell entry. The aim of the review is to give the reader a state-of-the-art overview on CHIKV cell entry and to provide an outlook on potential new avenues in CHIKV research.
Collapse
|
49
|
Abstract
Effective antivirals have been developed against specific viruses, such as HIV, Hepatitis C virus and influenza virus. This 'one bug-one drug' approach to antiviral drug development can be successful, but it may be inadequate for responding to an increasing diversity of viruses that cause significant diseases in humans. The majority of viral pathogens that cause emerging and re-emerging infectious diseases are membrane-enveloped viruses, which require the fusion of viral and cell membranes for virus entry. Therefore, antivirals that target the membrane fusion process represent new paradigms for broad-spectrum antiviral discovery. In this Review, we discuss the mechanisms responsible for the fusion between virus and cell membranes and explore how broad-spectrum antivirals target this process to prevent virus entry.
Collapse
Affiliation(s)
- Frederic Vigant
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisbon, Portugal
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, #1124, New York, New York 10029, USA
| |
Collapse
|
50
|
Dolganiuc A. Alcohol and Viral Hepatitis: Role of Lipid Rafts. Alcohol Res 2015; 37:299-309. [PMID: 26695752 PMCID: PMC4590625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Both alcohol abuse and infection with hepatitis viruses can lead to liver disease, including chronic hepatitis. Alcohol and hepatitis viruses have synergistic effects in the development of liver disease. Some of these involve the cellular membranes and particularly their functionally active domains, termed lipid rafts, which contain many proteins with essential roles in signaling and other processes. These lipid rafts play a central role in the lifecycles of hepatitis viruses. Alcohol's actions at the lipid rafts may contribute to the synergistic harmful effects of alcohol and hepatitis viruses on the liver and the pathogenesis of liver disease.
Collapse
Affiliation(s)
- Angela Dolganiuc
- Department of Medicine/Gastroenterology, Hepatology, and Nutrition at the University of Florida, Gainesville, Florida
| |
Collapse
|