1
|
Guan X, Wang T, Gao Y, Zhai H, Jiang F, Hou Q, Yang X, Wu H, Li LF, Luo Y, Li S, Sun Y, Qiu HJ, Li Y. The CP123L protein of African swine fever virus is a membrane-associated, palmitoylated protein required for viral replication. J Virol 2025; 99:e0144524. [PMID: 39714165 PMCID: PMC11784412 DOI: 10.1128/jvi.01445-24] [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: 08/21/2024] [Accepted: 10/31/2024] [Indexed: 12/24/2024] Open
Abstract
African swine fever (ASF) is a highly contagious and often lethal disease caused by African swine fever virus (ASFV) in pigs. Protein palmitoylation is a prevalent posttranslational lipid modification that can modulate viral replication. In this study, we investigated the palmitoylation of ASFV proteins. The results revealed that the CP123L protein (pCP123L) of ASFV was palmitoylated at the cysteine residue at position 18 (C18). To further elucidate the functional significance of this posttranslational modification, abolishing palmitoylation through a cysteine-to-serine mutation at C18 (C18S) of pCP123L (pCP123L/C18S) or treatment with 2-bromopalmitate (2-BP), a palmitoylation inhibitor, led to altered cytomembrane localization and migration rate of pCP123L. Furthermore, depalmitoylation achieved through 2-BP treatment significantly suppressed ASFV replication and exerted a profound impact on virus budding. Remarkably, blocking pCP123L palmitoylation via the C18S mutation resulted in decreased replication of ASFV. Our study represents the first evidence for the presence of palmitoylation in ASFV proteins and underscores its crucial role in viral replication. IMPORTANCE African swine fever (ASF) poses a significant threat to the global pig industry. The causative agent of ASF is African swine fever virus (ASFV), which encodes more than 165 proteins. Protein palmitoylation, a common posttranslational lipid modification, can modulate viral infection. To date, the ASFV proteins that undergo palmitoylation and their impacts on viral replication remain elusive. In this study, the CP123L protein (pCP123L) of ASFV was identified as a palmitoylated protein, and the cysteine residue at position 18 of pCP123L is responsible for its palmitoylation. Notably, our findings demonstrate that palmitoylation plays significant roles in ASFV protein functions and facilitates viral replication.
Collapse
Affiliation(s)
- Xiangyu Guan
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Tao Wang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuxuan Gao
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Huanjie Zhai
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Fengwei Jiang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Qinghe Hou
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Xiaoke Yang
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Hongxia Wu
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Lian-Feng Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuzi Luo
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Su Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yuan Sun
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Hua-Ji Qiu
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| | - Yongfeng Li
- State Key Laboratory for Animal Disease Control and Prevention, CAAS Harbin Veterinary Research Institute, Harbin, Heilongjiang, China
| |
Collapse
|
2
|
Ngo VN, Winski DP, Aho B, Kamath PL, King BL, Waters H, Zimmerberg J, Sodt A, Hess ST. Conserved sequence features in intracellular domains of viral spike proteins. Virology 2024; 599:110198. [PMID: 39116647 PMCID: PMC11383743 DOI: 10.1016/j.virol.2024.110198] [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: 06/06/2024] [Revised: 07/23/2024] [Accepted: 07/31/2024] [Indexed: 08/10/2024]
Abstract
Viral spike proteins mutate frequently, but conserved features within these proteins often have functional importance and can inform development of anti-viral therapies which circumvent the effects of viral sequence mutations. Through analysis of large numbers of viral spike protein sequences from several viral families, we found highly (>99%) conserved patterns within their intracellular domains. The patterns generally consist of one or more basic amino acids (arginine or lysine) adjacent to a cysteine, many of which are known to undergo acylation. These patterns were not enriched in cellular proteins in general. Molecular dynamics simulations show direct electrostatic and hydrophobic interactions between these conserved residues in hemagglutinin (HA) from influenza A and B and the phosphoinositide PIP2. Super-resolution microscopy shows nanoscale colocalization of PIP2 and several of the same viral proteins. We propose the hypothesis that these conserved viral spike protein features can interact with phosphoinositides such as PIP2.
Collapse
Affiliation(s)
- Vinh-Nhan Ngo
- Department of Physics and Astronomy, 120 Bennett Hall, University of Maine, Orono, ME, 04469-5709, USA
| | - David P Winski
- Department of Physics and Astronomy, 120 Bennett Hall, University of Maine, Orono, ME, 04469-5709, USA
| | - Brandon Aho
- Department of Physics and Astronomy, 120 Bennett Hall, University of Maine, Orono, ME, 04469-5709, USA
| | - Pauline L Kamath
- School of Food and Agriculture, 342 Hitchner Hall, University of Maine, And Maine Center for Genetics in the Environment, Orono, ME, USA.
| | - Benjamin L King
- Department of Molecular and Biomedical Sciences, 5735 Hitchner Hall, University of Maine, Orono, ME, USA.
| | - Hang Waters
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Alexander Sodt
- Unit on Membrane Chemical Physics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Samuel T Hess
- Department of Physics and Astronomy, 120 Bennett Hall, University of Maine, Orono, ME, 04469-5709, USA.
| |
Collapse
|
3
|
Forrester MT, Egol JR, Tata A, Tata PR, Foster MW. Analysis of Protein Cysteine Acylation Using a Modified Suspension Trap (Acyl-Trap). J Proteome Res 2024; 23:3716-3725. [PMID: 39008777 PMCID: PMC11826151 DOI: 10.1021/acs.jproteome.4c00225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Proteins undergo reversible S-acylation via a thioester linkage in vivo. S-palmitoylation, modification by C16:0 fatty acid, is a common S-acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S-acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 μg of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised "Acyl-Trap", a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S-acyl enrichment. We show that the method is compatible with protein-level detection of S-acylated proteins (e.g., H-Ras) as well as S-acyl site identification and quantification using "on trap" isobaric labeling and LC-MS/MS from as little as 20 μg of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S-acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamline the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.
Collapse
Affiliation(s)
- Michael T. Forrester
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jacob R. Egol
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Purushothama Rao Tata
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
4
|
Forrester MT, Egol JR, Tata A, Tata PR, Foster MW. Analysis of Protein Cysteine Acylation Using a Modified Suspension Trap (Acyl-Trap). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.23.586403. [PMID: 38585928 PMCID: PMC10996552 DOI: 10.1101/2024.03.23.586403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Proteins undergo reversible S-acylation via a thioester linkage in vivo. S-palmitoylation, modification by C16:0 fatty acid, is a common S-acylation that mediates critical protein-membrane and protein-protein interactions. The most widely used S-acylation assays, including acyl-biotin exchange and acyl resin-assisted capture, utilize blocking of free Cys thiols, hydroxylamine-dependent cleavage of the thioester and subsequent labeling of nascent thiol. These assays generally require >500 micrograms of protein input material per sample and numerous reagent removal and washing steps, making them laborious and ill-suited for high throughput and low input applications. To overcome these limitations, we devised "Acyl-Trap", a suspension trap-based assay that utilizes a thiol-reactive quartz to enable buffer exchange and hydroxylamine-mediated S-acyl enrichment. We show that the method is compatible with protein-level detection of S-acylated proteins (e.g. H-Ras) as well as S-acyl site identification and quantification using "on trap" isobaric labeling and LC-MS/MS from as little as 20 micrograms of protein input. In mouse brain, Acyl-Trap identified 279 reported sites of S-acylation and 1298 previously unreported putative sites. Also described are conditions for long-term hydroxylamine storage, which streamlines the assay. More generally, Acyl-Trap serves as a proof-of-concept for PTM-tailored suspension traps suitable for both traditional protein detection and chemoproteomic workflows.
Collapse
Affiliation(s)
- Michael T. Forrester
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Jacob R. Egol
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Purushothama Rao Tata
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27710, USA
- Duke Regeneration Center, Duke University School of Medicine, Durham, NC, 27710, USA
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care Medicine, Duke University School of Medicine, Durham, NC, 27710, USA
- Proteomics and Metabolomics Core Facility, Duke University School of Medicine, Durham, NC, 27710, USA
| |
Collapse
|
5
|
Bai M, Gallen E, Memarzadeh S, Howie J, Gao X, Kuo CWS, Brown E, Swingler S, Wilson SJ, Shattock MJ, France DJ, Fuller W. Targeted degradation of zDHHC-PATs decreases substrate S-palmitoylation. PLoS One 2024; 19:e0299665. [PMID: 38512906 PMCID: PMC10956751 DOI: 10.1371/journal.pone.0299665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 02/14/2024] [Indexed: 03/23/2024] Open
Abstract
Reversible S-palmitoylation of protein cysteines, catalysed by a family of integral membrane zDHHC-motif containing palmitoyl acyl transferases (zDHHC-PATs), controls the localisation, activity, and interactions of numerous integral and peripheral membrane proteins. There are compelling reasons to want to inhibit the activity of individual zDHHC-PATs in both the laboratory and the clinic, but the specificity of existing tools is poor. Given the extensive conservation of the zDHHC-PAT active site, development of isoform-specific competitive inhibitors is highly challenging. We therefore hypothesised that proteolysis-targeting chimaeras (PROTACs) may offer greater specificity to target this class of enzymes. In proof-of-principle experiments we engineered cell lines expressing tetracycline-inducible Halo-tagged zDHHC5 or zDHHC20, and evaluated the impact of Halo-PROTACs on zDHHC-PAT expression and substrate palmitoylation. In HEK-derived FT-293 cells, Halo-zDHHC5 degradation significantly decreased palmitoylation of its substrate phospholemman, and Halo-zDHHC20 degradation significantly diminished palmitoylation of its substrate IFITM3, but not of the SARS-CoV-2 spike protein. In contrast, in a second kidney derived cell line, Vero E6, Halo-zDHHC20 degradation did not alter palmitoylation of either IFITM3 or SARS-CoV-2 spike. We conclude from these experiments that PROTAC-mediated targeting of zDHHC-PATs to decrease substrate palmitoylation is feasible. However, given the well-established degeneracy in the zDHHC-PAT family, in some settings the activity of non-targeted zDHHC-PATs may substitute and preserve substrate palmitoylation.
Collapse
Affiliation(s)
- Mingjie Bai
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Emily Gallen
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sarah Memarzadeh
- School of Chemistry, University of Glasgow, Glasgow, United Kingdom
| | - Jacqueline Howie
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Xing Gao
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Chien-Wen S. Kuo
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Elaine Brown
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Simon Swingler
- Medical Research Council–University of Glasgow Centre for Virus Research, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Sam J. Wilson
- Medical Research Council–University of Glasgow Centre for Virus Research, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michael J. Shattock
- School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, United Kingdom
| | - David J. France
- School of Chemistry, University of Glasgow, Glasgow, United Kingdom
| | - William Fuller
- School of Cardiovascular & Metabolic Health, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
6
|
Carter T, Iqbal M. The Influenza A Virus Replication Cycle: A Comprehensive Review. Viruses 2024; 16:316. [PMID: 38400091 PMCID: PMC10892522 DOI: 10.3390/v16020316] [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: 01/18/2024] [Revised: 02/15/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Influenza A virus (IAV) is the primary causative agent of influenza, colloquially called the flu. Each year, it infects up to a billion people, resulting in hundreds of thousands of human deaths, and causes devastating avian outbreaks with worldwide losses worth billions of dollars. Always present is the possibility that a highly pathogenic novel subtype capable of direct human-to-human transmission will spill over into humans, causing a pandemic as devastating if not more so than the 1918 influenza pandemic. While antiviral drugs for influenza do exist, they target very few aspects of IAV replication and risk becoming obsolete due to antiviral resistance. Antivirals targeting other areas of IAV replication are needed to overcome this resistance and combat the yearly epidemics, which exact a serious toll worldwide. This review aims to summarise the key steps in the IAV replication cycle, along with highlighting areas of research that need more focus.
Collapse
Affiliation(s)
- Toby Carter
- The Pirbright Institute, Ash Road, Pirbright, Woking GU24 0NF, UK;
| | | |
Collapse
|
7
|
Michalski M, Setny P. Molecular Mechanisms behind Conformational Transitions of the Influenza Virus Hemagglutinin Membrane Anchor. J Phys Chem B 2023; 127:9450-9460. [PMID: 37877534 PMCID: PMC10641832 DOI: 10.1021/acs.jpcb.3c05257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 10/26/2023]
Abstract
Membrane fusion is a fundamental process that is exploited by enveloped viruses to enter host cells. In the case of the influenza virus, fusion is facilitated by the trimeric viral hemagglutinin protein (HA). So far, major focus has been put on its N-terminal fusion peptides, which are directly responsible for fusion initiation. A growing body of evidence points also to a significant functional role of the HA C-terminal domain, which however remains incompletely understood. Our computational study aimed to elucidate the structural and functional interdependencies within the HA C-terminal region encompassing the transmembrane domain (TMD) and the cytoplasmic tail (CT). In particular, we were interested in the conformational shift of the TMD in response to varying cholesterol concentration in the viral membrane and in its modulation by the presence of CT. Using free-energy calculations based on atomistic molecular dynamics simulations, we characterized transitions between straight and tilted metastable TMD configurations under varying conditions. We found that the presence of CT is essential for achieving a stable, highly tilted TMD configuration. As we demonstrate, such a configuration of HA membrane anchor likely supports the tilting motion of its ectodomain, which needs to be executed during membrane fusion. This finding highlights the functional role of, so far, the relatively overlooked CT region.
Collapse
Affiliation(s)
- Michal Michalski
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| | - Piotr Setny
- Centre of New Technologies, University of Warsaw, 02-097 Warsaw, Poland
| |
Collapse
|
8
|
Meng X, Veit M. Palmitoylation of the hemagglutinin of influenza B virus by ER-localized DHHC enzymes 1, 2, 4, and 6 is required for efficient virus replication. J Virol 2023; 97:e0124523. [PMID: 37792001 PMCID: PMC10617437 DOI: 10.1128/jvi.01245-23] [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: 08/16/2023] [Accepted: 08/17/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Influenza viruses are a public health concern since they cause seasonal outbreaks and occasionally pandemics. Our study investigates the importance of a protein modification called "palmitoylation" in the replication of influenza B virus. Palmitoylation involves attaching fatty acids to the viral protein hemagglutinin and has previously been studied for influenza A virus. We found that this modification is important for the influenza B virus to replicate, as mutating the sites where palmitate is attached prevented the virus from generating viable particles. Our experiments also showed that this modification occurs in the endoplasmic reticulum. We identified the specific enzymes responsible for this modification, which are different from those involved in palmitoylation of HA of influenza A virus. Overall, our research illuminates the similarities and differences in fatty acid attachment to HA of influenza A and B viruses and identifies the responsible enzymes, which might be promising targets for anti-viral therapy.
Collapse
Affiliation(s)
- Xiaorong Meng
- Veterinary Faculty, Institute for Virology, Freie Universität Berlin , Berlin, Germany
| | - Michael Veit
- Veterinary Faculty, Institute for Virology, Freie Universität Berlin , Berlin, Germany
| |
Collapse
|
9
|
Lin L, Wang X, Chen Z, Deng T, Yan Y, Dong W, Huang Y, Zhou J. TRIM21 restricts influenza A virus replication by ubiquitination-dependent degradation of M1. PLoS Pathog 2023; 19:e1011472. [PMID: 37343022 DOI: 10.1371/journal.ppat.1011472] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Tripartite motif-containing protein 21 (TRIM21), an E3 ubiquitin ligase, plays a critical role in the host antiviral response. However, the mechanism and antiviral spectrum of TRIM21 in influenza A virus (IAV) remain unclear. Here, we report that TRIM21 inhibits the replication of various IAV subtypes by targeting matrix protein 1 (M1) from H3/H5/H9, but not H1 and H7 M1. Mechanistically, TRIM21 binds to the residue R95 of M1 and facilitates K48 ubiquitination of M1 K242 for proteasome-dependent degradation, leading to the inhibition of H3, H5, and H9 IAV replication. Interestingly, the recombinant viruses with M1 R95K or K242R mutations were resistance to TRIM21 and exhibited more robust replication and severe pathogenicity. Moreover, the amino acid sequence M1 proteins, mainly from avian influenza such as H5N1, H7N9, H9N2, ranging from 1918 to 2022, reveals a gradual dominant accumulation of the TRIM21-driven R95K mutation when the virus jumps into mammals. Thus, TRIM21 in mammals' functions as a host restriction factor and drives a host adaptive mutation of influenza A virus.
Collapse
Affiliation(s)
- Lulu Lin
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University First Affiliated Hospital, Hangzhou, China
| | - Xingbo Wang
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Zhen Chen
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Tingjuan Deng
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yan Yan
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Weiren Dong
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
| | - Yu Huang
- Institute of Animal Husbandry and Veterinary, Fujian Academy of Agricultural Sciences, Fuzhou, China
| | - Jiyong Zhou
- MOA Key Laboratory of Animal Virology, Zhejiang University Center for Veterinary Sciences, Hangzhou, China
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University First Affiliated Hospital, Hangzhou, China
| |
Collapse
|
10
|
Jiang L, Chen H, Li C. Advances in deciphering the interactions between viral proteins of influenza A virus and host cellular proteins. CELL INSIGHT 2023; 2:100079. [PMID: 37193064 PMCID: PMC10134199 DOI: 10.1016/j.cellin.2023.100079] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/28/2023] [Accepted: 01/28/2023] [Indexed: 05/18/2023]
Abstract
Influenza A virus (IAV) poses a severe threat to the health of animals and humans. The genome of IAV consists of eight single-stranded negative-sense RNA segments, encoding ten essential proteins as well as certain accessory proteins. In the process of virus replication, amino acid substitutions continuously accumulate, and genetic reassortment between virus strains readily occurs. Due to this high genetic variability, new viruses that threaten animal and human health can emerge at any time. Therefore, the study on IAV has always been a focus of veterinary medicine and public health. The replication, pathogenesis, and transmission of IAV involve intricate interplay between the virus and host. On one hand, the entire replication cycle of IAV relies on numerous proviral host proteins that effectively allow the virus to adapt to its host and support its replication. On the other hand, some host proteins play restricting roles at different stages of the viral replication cycle. The mechanisms of interaction between viral proteins and host cellular proteins are currently receiving particular interest in IAV research. In this review, we briefly summarize the current advances in our understanding of the mechanisms by which host proteins affect virus replication, pathogenesis, or transmission by interacting with viral proteins. Such information about the interplay between IAV and host proteins could provide insights into how IAV causes disease and spreads, and might help support the development of antiviral drugs or therapeutic approaches.
Collapse
Affiliation(s)
- Li Jiang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Chengjun Li
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| |
Collapse
|
11
|
Hu D, Zou H, Chen W, Li Y, Luo Z, Wang X, Guo D, Meng Y, Liao F, Wang W, Zhu Y, Wu J, Li G. ZDHHC11 Suppresses Zika Virus Infections by Palmitoylating the Envelope Protein. Viruses 2023; 15:144. [PMID: 36680184 PMCID: PMC9863066 DOI: 10.3390/v15010144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/27/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Zika virus (ZIKV) is an RNA-enveloped virus that belongs to the Flavivirus genus, and ZIKV infections potentially induce severe neurodegenerative diseases and impair male fertility. Palmitoylation is an important post-translational modification of proteins that is mediated by a series of DHHC-palmitoyl transferases, which are implicated in various biological processes and viral infections. However, it remains to be investigated whether palmitoylation regulates ZIKV infections. In this study, we initially observed that the inhibition of palmitoylation by 2-bromopalmitate (2-BP) enhanced ZIKV infections, and determined that the envelope protein of ZIKV is palmitoylated at Cys308. ZDHHC11 was identified as the predominant enzyme that interacts with the ZIKV envelope protein and catalyzes its palmitoylation. Notably, ZDHHC11 suppressed ZIKV infections in an enzymatic activity-dependent manner and ZDHHC11 knockdown promoted ZIKV infection. In conclusion, we proposed that the envelope protein of ZIKV undergoes a novel post-translational modification and identified a distinct mechanism in which ZDHHC11 suppresses ZIKV infections via palmitoylation of the ZIKV envelope protein.
Collapse
Affiliation(s)
- Dingwen Hu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Haimei Zou
- The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Weijie Chen
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Yuting Li
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Ziqing Luo
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Xianyang Wang
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Dekuan Guo
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Yu Meng
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Feng Liao
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Wenbiao Wang
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou 510080, China
| | - Ying Zhu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Geng Li
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
- Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| |
Collapse
|
12
|
Panina IS, Krylov NA, Chugunov AO, Efremov RG, Kordyukova LV. The Mechanism of Selective Recognition of Lipid Substrate by hDHHC20 Enzyme. Int J Mol Sci 2022; 23:ijms232314791. [PMID: 36499114 PMCID: PMC9739150 DOI: 10.3390/ijms232314791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
S-acylation is a post-translational linkage of long chain fatty acids to cysteines, playing a key role in normal physiology and disease. In human cells, the reaction is catalyzed by a family of 23 membrane DHHC-acyltransferases (carrying an Asp-His-His-Cys catalytic motif) in two stages: (1) acyl-CoA-mediated autoacylation of the enzyme; and (2) further transfer of the acyl chain to a protein substrate. Despite the availability of a 3D-structure of human acyltransferase (hDHHC20), the molecular aspects of lipid selectivity of DHHC-acyltransferases remain unclear. In this paper, using molecular dynamics (MD) simulations, we studied membrane-bound hDHHC20 right before the acylation by C12-, C14-, C16-, C18-, and C20-CoA substrates. We found that: (1) regardless of the chain length, its terminal methyl group always reaches the "ceiling" of the enzyme's cavity; (2) only for C16, an optimal "reactivity" (assessed by a simple geometric criterion) permits the autoacylation; (3) in MD, some key interactions between an acyl-CoA and a protein differ from those in the reference crystal structure of the C16-CoA-hDHHS20 mutant complex (probably, because this structure corresponds to a non-native dimer). These features of specific recognition of full-size acyl-CoA substrates support our previous hypothesis of "geometric and physicochemical selectivity" derived for simplified acyl-CoA analogues.
Collapse
Affiliation(s)
- Irina S. Panina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- International Laboratory for Supercomputer Atomistic Modelling and Multi-Scale Analysis, National Research University Higher School of Economics, 101000 Moscow, Russia
| | - Nikolay A. Krylov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- International Laboratory for Supercomputer Atomistic Modelling and Multi-Scale Analysis, National Research University Higher School of Economics, 101000 Moscow, Russia
| | - Anton O. Chugunov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- International Laboratory for Supercomputer Atomistic Modelling and Multi-Scale Analysis, National Research University Higher School of Economics, 101000 Moscow, Russia
- Moscow Institute of Physics and Technology, State University, Dolgoprudny, 141701 Moscow, Russia
| | - Roman G. Efremov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
- International Laboratory for Supercomputer Atomistic Modelling and Multi-Scale Analysis, National Research University Higher School of Economics, 101000 Moscow, Russia
- Moscow Institute of Physics and Technology, State University, Dolgoprudny, 141701 Moscow, Russia
- Correspondence:
| | - Larisa V. Kordyukova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
13
|
Molecular Dynamics of DHHC20 Acyltransferase Suggests Principles of Lipid and Protein Substrate Selectivity. Int J Mol Sci 2022; 23:ijms23095091. [PMID: 35563480 PMCID: PMC9105814 DOI: 10.3390/ijms23095091] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/28/2022] [Accepted: 04/29/2022] [Indexed: 12/17/2022] Open
Abstract
Lipid modification of viral proteins with fatty acids of different lengths (S-acylation) is crucial for virus pathogenesis. The reaction is catalyzed by members of the DHHC family and proceeds in two steps: the autoacylation is followed by the acyl chain transfer onto protein substrates. The crystal structure of human DHHC20 (hDHHC20), an enzyme involved in the acylation of S-protein of SARS-CoV-2, revealed that the acyl chain may be inserted into a hydrophobic cavity formed by four transmembrane (TM) α-helices. To test this model, we used molecular dynamics of membrane-embedded hDHHC20 and its mutants either in the absence or presence of various acyl-CoAs. We found that among a range of acyl chain lengths probed only C16 adopts a conformation suitable for hDHHC20 autoacylation. This specificity is altered if the small or bulky residues at the cavity's ceiling are exchanged, e.g., the V185G mutant obtains strong preferences for binding C18. Surprisingly, an unusual hydrophilic ridge was found in TM helix 4 of hDHHC20, and the responsive hydrophilic patch supposedly involved in association was found in the 3D model of the S-protein TM-domain trimer. Finally, the exchange of critical Thr and Ser residues in the spike led to a significant decrease in its S-acylation. Our data allow further development of peptide/lipid-based inhibitors of hDHHC20 that might impede replication of Corona- and other enveloped viruses.
Collapse
|
14
|
Chua SCJH, Cui J, Engelberg D, Lim LHK. A Review and Meta-Analysis of Influenza Interactome Studies. Front Microbiol 2022; 13:869406. [PMID: 35531276 PMCID: PMC9069142 DOI: 10.3389/fmicb.2022.869406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Annually, the influenza virus causes 500,000 deaths worldwide. Influenza-associated mortality and morbidity is especially high among the elderly, children, and patients with chronic diseases. While there are antivirals available against influenza, such as neuraminidase inhibitors and adamantanes, there is growing resistance against these drugs. Thus, there is a need for novel antivirals for resistant influenza strains. Host-directed therapies are a potential strategy for influenza as host processes are conserved and are less prone mutations as compared to virus-directed therapies. A literature search was performed for papers that performed viral–host interaction screens and the Reactome pathway database was used for the bioinformatics analysis. A total of 15 studies were curated and 1717 common interactors were uncovered among all these studies. KEGG analysis, Enrichr analysis, STRING interaction analysis was performed on these interactors. Therefore, we have identified novel host pathways that can be targeted for host-directed therapy against influenza in our review.
Collapse
Affiliation(s)
- Sonja Courtney Jun Hui Chua
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- CREATE-NUS-HUJ Cellular & Molecular Mechanisms of Inflammation Programme, National University of Singapore, Singapore, Singapore
| | - Jianzhou Cui
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
| | - David Engelberg
- CREATE-NUS-HUJ Cellular & Molecular Mechanisms of Inflammation Programme, National University of Singapore, Singapore, Singapore
- Department of Microbiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Department of Biological Chemistry, The Institute of Life Science, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Hsiu Kim Lim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- *Correspondence: Lina Hsiu Kim Lim,
| |
Collapse
|
15
|
Abdulrahman DA, Meng X, Veit M. S-Acylation of Proteins of Coronavirus and Influenza Virus: Conservation of Acylation Sites in Animal Viruses and DHHC Acyltransferases in Their Animal Reservoirs. Pathogens 2021; 10:669. [PMID: 34072434 PMCID: PMC8227752 DOI: 10.3390/pathogens10060669] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/25/2021] [Indexed: 01/21/2023] Open
Abstract
Recent pandemics of zoonotic origin were caused by members of coronavirus (CoV) and influenza A (Flu A) viruses. Their glycoproteins (S in CoV, HA in Flu A) and ion channels (E in CoV, M2 in Flu A) are S-acylated. We show that viruses of all genera and from all hosts contain clusters of acylated cysteines in HA, S and E, consistent with the essential function of the modification. In contrast, some Flu viruses lost the acylated cysteine in M2 during evolution, suggesting that it does not affect viral fitness. Members of the DHHC family catalyze palmitoylation. Twenty-three DHHCs exist in humans, but the number varies between vertebrates. SARS-CoV-2 and Flu A proteins are acylated by an overlapping set of DHHCs in human cells. We show that these DHHC genes also exist in other virus hosts. Localization of amino acid substitutions in the 3D structure of DHHCs provided no evidence that their activity or substrate specificity is disturbed. We speculate that newly emerged CoVs or Flu viruses also depend on S-acylation for replication and will use the human DHHCs for that purpose. This feature makes these DHHCs attractive targets for pan-antiviral drugs.
Collapse
Affiliation(s)
- Dina A. Abdulrahman
- Department of Virology, Animal Health Research Institute (AHRI), Giza 12618, Egypt;
| | - Xiaorong Meng
- Institute of Virology, Veterinary Faculty, Free University Berlin, 14163 Berlin, Germany;
| | - Michael Veit
- Institute of Virology, Veterinary Faculty, Free University Berlin, 14163 Berlin, Germany;
| |
Collapse
|
16
|
Gadalla MR, Morrison E, Serebryakova MV, Han X, Wolff T, Freund C, Kordyukova L, Veit M. NS1-mediated upregulation of ZDHHC22 acyltransferase in influenza a virus infected cells. Cell Microbiol 2021; 23:e13322. [PMID: 33629465 DOI: 10.1111/cmi.13322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 02/19/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022]
Abstract
Influenza A viruses contain two S-acylated proteins, the ion channel M2 and the glycoprotein hemagglutinin (HA). Acylation of the latter is essential for virus replication. Here we analysed the expression of each of the 23 members of the family of ZDHHC acyltransferases in human airway cells, the site of virus replication. RT-PCR revealed that every ZDHHC acyltransferase (except ZDHHC19) is expressed in A549 and Calu cells. Interestingly, expression of one ZDHHC, ZDHHC22, is upregulated in virus-infected cells; this effect is more pronounced after infection with an avian compared to a human virus strain. The viral protein NS1 triggers ZDHHC22 expression in transfected cells, whereas recombinant viruses lacking a functional NS1 gene did not cause ZDHHC22 upregulation. CRISPR/Cas9 technology was then used to knock-out the ZDHHC22 gene in A549 cells. However, acylation of M2 and HA was not reduced, as analysed for intracellular HA and M2 and the stoichiometry of S-acylation of HA incorporated into virus particles did not change according to MALDI-TOF mass spectrometry analysis. Comparative mass spectrometry of palmitoylated proteins in wt and ΔZDHHC22 cells identified 25 potential substrates of ZDHHC22 which might be involved in virus replication.
Collapse
Affiliation(s)
- Mohamed Rasheed Gadalla
- Institute of Virology, Free University Berlin, Berlin, Germany.,Department of Virology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Eliot Morrison
- Institute of Chemistry and Biochemistry, Free University Berlin, Berlin, Germany
| | - Marina V Serebryakova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Xueijiao Han
- Institute of Virology, Free University Berlin, Berlin, Germany
| | - Thorsten Wolff
- Unit 17: Influenza and Other Respiratory Viruses, Robert Koch Institute, Berlin, Germany
| | - Christian Freund
- Institute of Chemistry and Biochemistry, Free University Berlin, Berlin, Germany
| | - Larisa Kordyukova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Michael Veit
- Institute of Virology, Free University Berlin, Berlin, Germany
| |
Collapse
|
17
|
Influenza A H1 and H3 Transmembrane Domains Interact Differently with Each Other and with Surrounding Membrane Lipids. Viruses 2020; 12:v12121461. [PMID: 33348831 PMCID: PMC7765950 DOI: 10.3390/v12121461] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 02/06/2023] Open
Abstract
Hemagglutinin (HA) is a class I viral membrane fusion protein, which is the most abundant transmembrane protein on the surface of influenza A virus (IAV) particles. HA plays a crucial role in the recognition of the host cell, fusion of the viral envelope and the host cell membrane, and is the major antigen in the immune response during the infection. Mature HA organizes in homotrimers consisting of a sequentially highly variable globular head and a relatively conserved stalk region. Every HA monomer comprises a hydrophilic ectodomain, a pre-transmembrane domain (pre-TMD), a hydrophobic transmembrane domain (TMD), and a cytoplasmic tail (CT). In recent years the effect of the pre-TMD and TMD on the structure and function of HA has drawn some attention. Using bioinformatic tools we analyzed all available full-length amino acid sequences of HA from 16 subtypes across various host species. We calculated several physico-chemical parameters of HA pre-TMDs and TMDs including accessible surface area (ASA), average hydrophobicity (Hav), and the hydrophobic moment (µH). Our data suggests that distinct differences in these parameters between the two major phylogenetic groups, represented by H1 and H3 subtypes, could have profound effects on protein–lipid interactions, trimer formation, and the overall HA ectodomain orientation and antigen exposure.
Collapse
|
18
|
Gomord V, Stordeur V, Fitchette AC, Fixman ED, Tropper G, Garnier L, Desgagnes R, Viel S, Couillard J, Beauverger G, Trepout S, Ward BJ, van Ree R, Faye L, Vézina LP. Design, production and immunomodulatory potency of a novel allergen bioparticle. PLoS One 2020; 15:e0242867. [PMID: 33259521 PMCID: PMC7707610 DOI: 10.1371/journal.pone.0242867] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/10/2020] [Indexed: 02/08/2023] Open
Abstract
Allergen immunotherapy (AIT) is the only disease-modifying treatment with evidence for sustained efficacy. However, it is poorly developed compared to symptomatic drugs. The main reasons come from treatment duration implying monthly injections during 3 to 5 years or daily sublingual use, and the risk of allergic side-effects. To become a more attractive alternative to lifelong symptomatic drug use, improvements to AIT are needed. Among the most promising new immunotherapy strategies is the use of bioparticles for the presentation of target antigen to the immune system as they can elicit strong T cell and B cell immune responses. Virus-like particles (VLPs) are a specific class of bioparticles in which the structural and immunogenic constituents are from viral origin. However, VLPs are ill-suited for use in AIT as their antigenicity is linked to structure. Recently, synthetic biology has been used to produce artificial modular bioparticles, in which supramolecular assemblies are made of elements from heterogeneous biological sources promoting the design and use of in vivo-assembling enveloped bioparticles for viral and non-viral antigens presentation. We have used a coiled-coil hybrid assembly for the design of an enveloped bioparticle (eBP) that present trimers of the Der p 2 allergen at its surface, This bioparticle was produced as recombinant and in vivo assembled eBPs in plant. This allergen biotherapeutic was used to demonstrate i) the capacity of plants to produce synthetic supramolecular allergen bioparticles, and ii) the immunomodulatory potential of naturally-assembled allergen bioparticles. Our results show that allergens exposed on eBPs induced a very strong IgG response consisting predominantly of IgG2a in favor of the TH1 response. Finally, our results demonstrate that rDer p 2 present on the surface of BPs show a very limited potential to stimulate the basophil degranulation of patient allergic to this allergen which is predictive of a high safety potential.
Collapse
Affiliation(s)
- Véronique Gomord
- ANGANY Innovation, Val de Reuil, France
- ANGANY Inc, Québec, Québec, Canada
| | | | | | - Elizabeth D. Fixman
- McGill University Health Centre, Research Institute (RI MUHC), Montreal, Quebec, Canada
| | | | - Lorna Garnier
- Service d’Immunologie Biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | - Sébastien Viel
- Service d’Immunologie Biologique, Hospices Civils de Lyon, Hôpital Lyon Sud, Pierre-Bénite, France
| | | | | | - Sylvain Trepout
- IR2 Inserm, Plateforme de microscopie électronique, INSERM US43/CNRS UMS2016, Institut Curie, Orsay, France
| | - Brian J. Ward
- McGill University Health Centre, Research Institute (RI MUHC), Montreal, Quebec, Canada
| | - Ronald van Ree
- Department of Experimental Immunology, Molecular and Translational Allergy, Amsterdam, Netherlands
| | - Loic Faye
- ANGANY Innovation, Val de Reuil, France
| | | |
Collapse
|
19
|
Selzer L, Su Z, Pintilie GD, Chiu W, Kirkegaard K. Full-length three-dimensional structure of the influenza A virus M1 protein and its organization into a matrix layer. PLoS Biol 2020; 18:e3000827. [PMID: 32997652 PMCID: PMC7549809 DOI: 10.1371/journal.pbio.3000827] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 10/12/2020] [Accepted: 09/08/2020] [Indexed: 02/06/2023] Open
Abstract
Matrix proteins are encoded by many enveloped viruses, including influenza viruses, herpes viruses, and coronaviruses. Underneath the viral envelope of influenza virus, matrix protein 1 (M1) forms an oligomeric layer critical for particle stability and pH-dependent RNA genome release. However, high-resolution structures of full-length monomeric M1 and the matrix layer have not been available, impeding antiviral targeting and understanding of the pH-dependent transitions involved in cell entry. Here, purification and extensive mutagenesis revealed protein–protein interfaces required for the formation of multilayered helical M1 oligomers similar to those observed in virions exposed to the low pH of cell entry. However, single-layered helical oligomers with biochemical and ultrastructural similarity to those found in infectious virions before cell entry were observed upon mutation of a single amino acid. The highly ordered structure of the single-layered oligomers and their likeness to the matrix layer of intact virions prompted structural analysis by cryo-electron microscopy (cryo-EM). The resulting 3.4-Å–resolution structure revealed the molecular details of M1 folding and its organization within the single-shelled matrix. The solution of the full-length M1 structure, the identification of critical assembly interfaces, and the development of M1 assembly assays with purified proteins are crucial advances for antiviral targeting of influenza viruses. Multi-subunit shells of matrix proteins line the interior of infectious influenza virus particles. In this study, biochemical purification of wild-type and mutant influenza M1 proteins allows the structural determination of an oligomer whose shape corresponds to that of infectious virions and suggests mechanisms for its formation and dismantling during infection.
Collapse
Affiliation(s)
- Lisa Selzer
- Departments of Genetics Stanford University School of Medicine, Stanford, California, United States of America
| | - Zhaoming Su
- The State Key Laboratory of Biotherapy and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Sichuan, China
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, California, United States of America
| | - Grigore D. Pintilie
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, California, United States of America
| | - Wah Chiu
- Department of Bioengineering, James H. Clark Center, Stanford University, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail: (KK); (WC)
| | - Karla Kirkegaard
- Departments of Genetics Stanford University School of Medicine, Stanford, California, United States of America
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail: (KK); (WC)
| |
Collapse
|
20
|
Dawson AR, Wilson GM, Coon JJ, Mehle A. Post-Translation Regulation of Influenza Virus Replication. Annu Rev Virol 2020; 7:167-187. [DOI: 10.1146/annurev-virology-010320-070410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Influenza virus exploits cellular factors to complete each step of viral replication. Yet, multiple host proteins actively block replication. Consequently, infection success depends on the relative speed and efficacy at which both the virus and host use their respective effectors. Post-translational modifications (PTMs) afford both the virus and the host means to readily adapt protein function without the need for new protein production. Here we use influenza virus to address concepts common to all viruses, reviewing how PTMs facilitate and thwart each step of the replication cycle. We also discuss advancements in proteomic methods that better characterize PTMs. Although some effectors and PTMs have clear pro- or antiviral functions, PTMs generally play regulatory roles to tune protein functions, levels, and localization. Synthesis of our current understanding reveals complex regulatory schemes where the effects of PTMs are time and context dependent as the virus and host battle to control infection.
Collapse
Affiliation(s)
- Anthony R. Dawson
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin 53706, USA
| | - Gary M. Wilson
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53706, USA
| | - Joshua J. Coon
- Department of Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53706, USA
- Department of Biomolecular Chemistry, University of Wisconsin–Madison, Madison, Wisconsin 53706, USA
| | - Andrew Mehle
- Department of Medical Microbiology and Immunology, University of Wisconsin–Madison, Madison, Wisconsin 53706, USA
| |
Collapse
|
21
|
The native structure of the assembled matrix protein 1 of influenza A virus. Nature 2020; 587:495-498. [PMID: 32908308 DOI: 10.1038/s41586-020-2696-8] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 06/19/2020] [Indexed: 02/03/2023]
Abstract
Influenza A virus causes millions of severe cases of disease during annual epidemics. The most abundant protein in influenza virions is matrix protein 1 (M1), which mediates virus assembly by forming an endoskeleton beneath the virus membrane1. The structure of full-length M1, and how it oligomerizes to mediate the assembly of virions, is unknown. Here we determine the complete structure of assembled M1 within intact virus particles, as well as the structure of M1 oligomers reconstituted in vitro. We find that the C-terminal domain of M1 is disordered in solution but can fold and bind in trans to the N-terminal domain of another M1 monomer, thus polymerizing M1 into linear strands that coat the interior surface of the membrane of the assembling virion. In the M1 polymer, five histidine residues-contributed by three different monomers of M1-form a cluster that can serve as the pH-sensitive disassembly switch after entry into a target cell. These structures therefore reveal mechanisms of influenza virus assembly and disassembly.
Collapse
|
22
|
Hu J, Zhang L, Liu X. Role of Post-translational Modifications in Influenza A Virus Life Cycle and Host Innate Immune Response. Front Microbiol 2020; 11:517461. [PMID: 33013775 PMCID: PMC7498822 DOI: 10.3389/fmicb.2020.517461] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 08/14/2020] [Indexed: 01/01/2023] Open
Abstract
Throughout various stages of its life cycle, influenza A virus relies heavily on host cellular machinery, including the post-translational modifications (PTMs) system. During infection, influenza virus interacts extensively with the cellular PTMs system to aid in its successful infection and dissemination. The complex interplay between viruses and the PTMs system induces global changes in PTMs of the host proteome as well as modifications of specific host or viral proteins. The most common PTMs include phosphorylation, ubiquitination, SUMOylation, acetylation, methylation, NEDDylation, and glycosylation. Many PTMs directly support influenza virus infection, whereas others contribute to modulating antiviral responses. In this review, we describe current knowledge regarding the role of PTMs in different stages of the influenza virus replication cycle. We also discuss the concerted role of PTMs in antagonizing host antiviral responses, with an emphasis on their impact on viral pathogenicity and host range.
Collapse
Affiliation(s)
- Jiao Hu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
| | - Lei Zhang
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, School of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Key Laboratory of Prevention and Control of Biological Hazard Factors (Animal Origin) for Agrifood Safety and Quality, Ministry of Agriculture of China, Yangzhou University, Yangzhou, China
| |
Collapse
|
23
|
Mecate-Zambrano A, Sukumar S, Seebohm G, Ciminski K, Schreiber A, Anhlan D, Greune L, Wixler L, Grothe S, Stein NC, Schmidt MA, Langer K, Schwemmle M, Shi T, Ludwig S, Boergeling Y. Discrete spatio-temporal regulation of tyrosine phosphorylation directs influenza A virus M1 protein towards its function in virion assembly. PLoS Pathog 2020; 16:e1008775. [PMID: 32866218 PMCID: PMC7485975 DOI: 10.1371/journal.ppat.1008775] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 09/11/2020] [Accepted: 07/02/2020] [Indexed: 01/09/2023] Open
Abstract
Small RNA viruses only have a very limited coding capacity, thus most viral proteins have evolved to fulfill multiple functions. The highly conserved matrix protein 1 (M1) of influenza A viruses is a prime example for such a multifunctional protein, as it acts as a master regulator of virus replication whose different functions have to be tightly regulated. The underlying mechanisms, however, are still incompletely understood. Increasing evidence points towards an involvement of posttranslational modifications in the spatio-temporal regulation of M1 functions. Here, we analyzed the role of M1 tyrosine phosphorylation in genuine infection by using recombinant viruses expressing M1 phosphomutants. Presence of M1 Y132A led to significantly decreased viral replication compared to wildtype and M1 Y10F. Characterization of phosphorylation dynamics by mass spectrometry revealed the presence of Y132 phosphorylation in M1 incorporated into virions that is most likely mediated by membrane-associated Janus kinases late upon infection. Molecular dynamics simulations unraveled a potential phosphorylation-induced exposure of the positively charged linker domain between helices 4 and 5, supposably acting as interaction platform during viral assembly. Consistently, M1 Y132A showed a defect in lipid raft localization due to reduced interaction with viral HA protein resulting in a diminished structural stability of viral progeny and the formation of filamentous particles. Importantly, reduced M1-RNA binding affinity resulted in an inefficient viral genome incorporation and the production of non-infectious virions that interferes with virus pathogenicity in mice. This study advances our understanding of the importance of dynamic phosphorylation as a so far underestimated level of regulation of multifunctional viral proteins and emphasizes the potential feasibility of targeting posttranslational modifications of M1 as a novel antiviral intervention.
Collapse
Affiliation(s)
- Angeles Mecate-Zambrano
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
| | - Swathi Sukumar
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
| | - Guiscard Seebohm
- Institute for Genetics of Heart Diseases (IfGH), Department of Cardiovascular Medicine, University Hospital Muenster, Muenster, Germany
| | - Kevin Ciminski
- Institute of Virology, Medical Center–University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - André Schreiber
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
| | - Darisuren Anhlan
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Lilo Greune
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Muenster, Muenster, Germany
| | - Ludmilla Wixler
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
| | - Stephanie Grothe
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Muenster, Germany
| | - Nora Caroline Stein
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Muenster, Germany
| | - M. Alexander Schmidt
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
- Institute of Infectiology, Center for Molecular Biology of Inflammation (ZMBE), University of Muenster, Muenster, Germany
| | - Klaus Langer
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
- Institute of Pharmaceutical Technology and Biopharmacy, University of Muenster, Muenster, Germany
| | - Martin Schwemmle
- Institute of Virology, Medical Center–University of Freiburg, Freiburg, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tianlai Shi
- Immunology, Inflammation and Infectious Diseases (I3) DTA, Roche Pharma Research and Early Development, Roche Innovation Center Basel, Basel, Switzerland
| | - Stephan Ludwig
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
- * E-mail:
| | - Yvonne Boergeling
- Institute of Virology Muenster, University of Muenster, Muenster, Germany
- Cells in Motion Interfaculty Centre (CiM), University of Muenster, Muenster, Germany
| |
Collapse
|
24
|
Yurtsever D, Lorent JH. Structural Modifications Controlling Membrane Raft Partitioning and Curvature in Human and Viral Proteins. J Phys Chem B 2020; 124:7574-7585. [PMID: 32813532 PMCID: PMC7476027 DOI: 10.1021/acs.jpcb.0c03435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Membrane
proteins and lipids have the capacity to associate into
lateral domains in cell membranes through mutual or collective interactions.
Lipid rafts are functional lateral domains that are formed through
collective interactions of certain lipids and which can include or
exclude proteins. These domains have been implicated in cell signaling
and protein trafficking and seem to be of importance for virus–host
interactions. We therefore want to investigate if raft and viral membrane
proteins present similar structural features, and how these features
are distributed throughout viruses. For this purpose, we performed
a bioinformatics analysis of raft and viral membrane proteins from
available online databases and compared them to nonraft proteins.
In general, transmembrane proteins of rafts and viruses had higher
proportions of palmitoyl and phosphoryl residues compared to nonraft
proteins. They differed in terms of transmembrane domain length and
thickness, with viral proteins being generally shorter and having
a smaller accessible surface area per residue. Nontransmembrane raft
proteins had increased amounts of palmitoyl, prenyl, and phosphoryl
moieties while their viral counterparts were largely myristoylated
and phosphorylated. Several of these structural determinants such
as phosphorylation are new to the raft field and are extensively discussed
in terms of raft functionality and phase separation. Surprisingly,
the proportion of palmitoylated viral transmembrane proteins was inversely
correlated to the virus size which indicated the implication of palmitoylation
in virus membrane curvature and possibly budding. The current results
provide new insights into the raft–virus interplay and unveil
possible targets for antiviral compounds.
Collapse
Affiliation(s)
- Deniz Yurtsever
- Theoretical Biology and Bioinformatics, Department of Biology, Utrecht University, Padualaan 8, NL-3584CH Utrecht, The Netherlands
| | - Joseph Helmuth Lorent
- Membrane Biochemistry & Biophysics, Bijvoet Center for Biomolecular Research, Department of Chemistry, Utrecht University, Padualaan 8, NL-3584CH Utrecht, The Netherlands
| |
Collapse
|
25
|
Ting-Hui-Lin, Chia MY, Lin CY, Yeh YQ, Jeng US, Wu WG, Lee MS. Improving immunogenicity of influenza virus H7N9 recombinant hemagglutinin for vaccine development. Vaccine 2020; 37:1897-1903. [PMID: 30857635 DOI: 10.1016/j.vaccine.2018.09.034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 09/08/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Human infections of novel avian influenza A virus (H7N9) emerged in early 2013 and caused about 40% case-fatality through 2017. Therefore, development of influenza H7N9 vaccines is critical for pandemic preparedness. Currently, there are three means of production of commercial influenza vaccines: egg-based, mammalian cell-based, and insect cell-based platforms. The insect cell-based platform has the advantage of high speed in producing recombinant protein. In this study, we evaluate the stability and immunogenicity of two different influenza H7 HA expression constructs generated using the baculovirus system, including membrane-based full-length HA (mH7) and secreted ectodomain-based H7 (sH7). The mH7 construct could form an oligomer-rosette structure and had a high hemagglutinin (HA) titer 8192. In contrast to mH7, the sH7 construct could not form an oligomer-rosette structure and did not have HA titer before cross-linking with anti-His antibody. Thermal stability tests showed that the sH7 and mH7 constructs were unstable at 43 °C and 52 °C, respectively. In a mice immunization study, the mH7 construct but not the sH7 construct could induce robust HI and neutralizing antibody titers. In conclusion, further development of the mH7 vaccine candidate is desirable.
Collapse
Affiliation(s)
- Ting-Hui-Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Yuan Chia
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan; Department of Veterinary Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chun-Yang Lin
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan
| | - Yi-Qi Yeh
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu, Taiwan; Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Guey Wu
- College of Life Science, National Tsing-Hua University, Hsinchu, Taiwan
| | - Min-Shi Lee
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Zhunan, Miaoli County, Taiwan.
| |
Collapse
|
26
|
Stix R, Lee CJ, Faraldo-Gómez JD, Banerjee A. Structure and Mechanism of DHHC Protein Acyltransferases. J Mol Biol 2020; 432:4983-4998. [PMID: 32522557 DOI: 10.1016/j.jmb.2020.05.023] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/26/2020] [Accepted: 05/28/2020] [Indexed: 01/01/2023]
Abstract
S-acylation, whereby a fatty acid chain is covalently linked to a cysteine residue by a thioester linkage, is the most prevalent kind of lipid modification of proteins. Thousands of proteins are targets of this post-translational modification, which is catalyzed by a family of eukaryotic integral membrane enzymes known as DHHC protein acyltransferases (DHHC-PATs). Our knowledge of the repertoire of S-acylated proteins has been rapidly expanding owing to development of the chemoproteomic techniques. There has also been an increasing number of reports in the literature documenting the importance of S-acylation in human physiology and disease. Recently, the first atomic structures of two different DHHC-PATs were determined using X-ray crystallography. This review will focus on the insights gained into the molecular mechanism of DHHC-PATs from these structures and highlight representative data from the biochemical literature that they help explain.
Collapse
Affiliation(s)
- Robyn Stix
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chul-Jin Lee
- Unit on Structural and Chemical Biology of Membrane Proteins, Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - José D Faraldo-Gómez
- Theoretical Molecular Biophysics Laboratory, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anirban Banerjee
- Unit on Structural and Chemical Biology of Membrane Proteins, Neurosciences and Cellular and Structural Biology Division, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
27
|
Pinigin KV, Kondrashov OV, Jiménez-Munguía I, Alexandrova VV, Batishchev OV, Galimzyanov TR, Akimov SA. Elastic deformations mediate interaction of the raft boundary with membrane inclusions leading to their effective lateral sorting. Sci Rep 2020; 10:4087. [PMID: 32139760 PMCID: PMC7058020 DOI: 10.1038/s41598-020-61110-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 02/18/2020] [Indexed: 02/06/2023] Open
Abstract
Liquid-ordered lipid domains represent a lateral inhomogeneity in cellular membranes. These domains have elastic and physicochemical properties different from those of the surrounding membrane. In particular, their thickness exceeds that of the disordered membrane. Thus, elastic deformations arise at the domain boundary in order to compensate for the thickness mismatch. In equilibrium, the deformations lead to an incomplete register of monolayer ordered domains: the elastic energy is minimal if domains in opposing monolayers lie on the top of each other, and their boundaries are laterally shifted by about 3 nm. This configuration introduces a region, composed of one ordered and one disordered monolayers, with an intermediate bilayer thickness. Besides, a jump in a local monolayer curvature takes place in this intermediate region, concentrating here most of the elastic stress. This region can participate in a lateral sorting of membrane inclusions by offering them an optimal bilayer thickness and local curvature conditions. In the present study, we consider the sorting of deformable lipid inclusions, undeformable peripheral and deeply incorporated peptide inclusions, and undeformable transmembrane inclusions of different molecular geometry. With rare exceptions, all types of inclusions have an affinity to the ordered domain boundary as compared to the bulk phases. The optimal lateral distribution of inclusions allows relaxing the elastic stress at the boundary of domains.
Collapse
Affiliation(s)
- Konstantin V Pinigin
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy prospekt, Moscow, 119071, Russia
| | - Oleg V Kondrashov
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy prospekt, Moscow, 119071, Russia
| | - Irene Jiménez-Munguía
- National University of Science and Technology "MISiS", 4 Leninskiy prospect, Moscow, 119049, Russia
| | | | - Oleg V Batishchev
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy prospekt, Moscow, 119071, Russia
| | - Timur R Galimzyanov
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy prospekt, Moscow, 119071, Russia
| | - Sergey A Akimov
- A.N. Frumkin Institute of Physical Chemistry and Electrochemistry, Russian Academy of Sciences, 31/4 Leninskiy prospekt, Moscow, 119071, Russia.
| |
Collapse
|
28
|
Meischel T, Villalon-Letelier F, Saunders PM, Reading PC, Londrigan SL. Influenza A virus interactions with macrophages: Lessons from epithelial cells. Cell Microbiol 2020; 22:e13170. [PMID: 31990121 DOI: 10.1111/cmi.13170] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 11/24/2019] [Accepted: 12/19/2019] [Indexed: 12/25/2022]
Abstract
Influenza viruses are an important cause of respiratory infection worldwide. In humans, infection with seasonal influenza A virus (IAV) is generally restricted to the respiratory tract where productive infection of airway epithelial cells promotes viral amplification, dissemination, and disease. Alveolar macrophages (MΦ) are also among the first cells to detect and respond to IAV, where they play a pivotal role in mounting effective innate immune responses. In contrast to epithelial cells, IAV infection of MΦ is a "dead end" for most seasonal strains, where replication is abortive and newly synthesised virions are not released. Although the key replicative stages leading to productive IAV infection in epithelial cells are defined, there is limited knowledge about the abortive IAV life cycle in MΦ. In this review, we will explore host factors and viral elements that support the early stages (entry) through to the late stages (viral egress) of IAV replication in epithelial cells. Similarities, differences, and unknowns for each key stage of the IAV replicative cycle in MΦ will then be highlighted. Herein, we provide mechanistic insights into MΦ-specific control of seasonal IAV replication through abortive infection, which may in turn, contribute to effective host defence.
Collapse
Affiliation(s)
- Tina Meischel
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Fernando Villalon-Letelier
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Philippa M Saunders
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Patrick C Reading
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.,WHO Collaborating Centre for Reference and Research on Influenza, Victorian Infectious Diseases Reference Laboratory, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sarah L Londrigan
- Department of Microbiology and Immunology, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| |
Collapse
|
29
|
Gadalla MR, Abrami L, van der Goot FG, Veit M. Hemagglutinin of Influenza A, but not of Influenza B and C viruses is acylated by ZDHHC2, 8, 15 and 20. Biochem J 2020; 477:285-303. [PMID: 31872235 DOI: 10.1042/bcj20190752] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Revised: 12/15/2019] [Accepted: 12/23/2019] [Indexed: 02/05/2023]
Abstract
Hemagglutinin (HA), a glycoprotein of Influenza A viruses and its proton channel M2 are site-specifically modified with fatty acids. Whereas two cysteines in the short cytoplasmic tail of HA contain only palmitate, stearate is exclusively attached to one cysteine located at the cytoplasmic border of the transmembrane region (TMR). M2 is palmitoylated at a cysteine positioned in an amphiphilic helix near the TMR. The enzymes catalyzing acylation of HA and M2 have not been identified, but zinc finger DHHC domain-containing (ZDHHC) palmitoyltransferases are candidates. We used a siRNA library to knockdown expression of each of the 23 human ZDHHCs in HA-expressing HeLa cells. siRNAs against ZDHHC2 and 8 had the strongest effect on acylation of HA as demonstrated by Acyl-RAC and confirmed by 3H-palmitate labeling. CRISPR/Cas9 knockout of ZDHHC2 and 8 in HAP1 cells, but also of the phylogenetically related ZDHHCs 15 and 20 strongly reduced acylation of group 1 and group 2 HAs and of M2, but individual ZDHHCs exhibit slightly different substrate preferences. These ZDHHCs co-localize with HA at membranes of the exocytic pathway in a human lung cell line. ZDHHC2, 8, 15 and 20 are not required for acylation of the HA-esterase-fusion protein of Influenza C virus that contains only stearate at one transmembrane cysteine. Knockout of these ZDHHCs also did not compromise acylation of HA of Influenza B virus that contains two palmitoylated cysteines in its cytoplasmic tail. Results are discussed with respect to the acyl preferences and possible substrate recognition features of the identified ZDHHCs.
Collapse
Affiliation(s)
- Mohamed Rasheed Gadalla
- Institute of Virology, Free University Berlin, 14163 Berlin, Germany
- Department of Virology, Faculty of Veterinary Medicine, Cairo University, 12211 Giza, Egypt
| | - Laurence Abrami
- Global Health Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - F Gisou van der Goot
- Global Health Institute, Faculty of Life Sciences, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Michael Veit
- Institute of Virology, Free University Berlin, 14163 Berlin, Germany
| |
Collapse
|
30
|
Gadalla MR, Veit M. Toward the identification of ZDHHC enzymes required for palmitoylation of viral protein as potential drug targets. Expert Opin Drug Discov 2019; 15:159-177. [PMID: 31809605 DOI: 10.1080/17460441.2020.1696306] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Introduction: S-acylation is the attachment of fatty acids not only to cysteines of cellular, but also of viral proteins. The modification is often crucial for the protein´s function and hence for virus replication. Transfer of fatty acids is mediated by one or several of the 23 members of the ZDHHC family of proteins. Since their genes are linked to various human diseases, they represent drug targets.Areas covered: The authors explore whether targeting acylation of viral proteins might be a strategy to combat viral diseases. Many human pathogens contain S-acylated proteins; the ZDHHCs involved in their acylation are currently identified. Based on the 3D structure of two ZDHHCs, the regulation and the biochemistry of the palmitolyation reaction and the lipid and protein substrate specificities are discussed. The authors then speculate how ZDHHCs might recognize S-acylated membrane proteins of Influenza virus.Expert opinion: Although many viral diseases can now be treated, the available drugs bind to viral proteins that rapidly mutate and become resistant. To develop inhibitors for the genetically more stable cellular ZDHHCs, their binding sites for viral substrates need to be identified. If only a few cellular proteins are recognized by the same binding site, development of specific inhibitors may have therapeutic potential.
Collapse
Affiliation(s)
- Mohamed Rasheed Gadalla
- Institute of Virology, Free University Berlin, Berlin, Germany.,Department of Virology, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Michael Veit
- Institute of Virology, Free University Berlin, Berlin, Germany
| |
Collapse
|
31
|
Kordyukova LV, Serebryakova MV, Khrustalev VV, Veit M. Differential S-Acylation of Enveloped Viruses. Protein Pept Lett 2019; 26:588-600. [PMID: 31161979 DOI: 10.2174/0929866526666190603082521] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 04/11/2019] [Accepted: 04/11/2019] [Indexed: 12/18/2022]
Abstract
Post-translational modifications often regulate protein functioning. Covalent attachment of long chain fatty acids to cysteine residues via a thioester linkage (known as protein palmitoylation or S-acylation) affects protein trafficking, protein-protein and protein-membrane interactions. This post-translational modification is coupled to membrane fusion or virus assembly and may affect viral replication in vitro and thus also virus pathogenesis in vivo. In this review we outline modern methods to study S-acylation of viral proteins and to characterize palmitoylproteomes of virus infected cells. The palmitoylation site predictor CSS-palm is critically tested against the Class I enveloped virus proteins. We further focus on identifying the S-acylation sites directly within acyl-peptides and the specific fatty acid (e.g, palmitate, stearate) bound to them using MALDI-TOF MS-based approaches. The fatty acid heterogeneity/ selectivity issue attracts now more attention since the recently published 3D-structures of two DHHC-acyl-transferases gave a hint how this might be achieved.
Collapse
Affiliation(s)
- Larisa V Kordyukova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | - Marina V Serebryakova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russian Federation
| | - Vladislav V Khrustalev
- Department of General Chemistry, Belarusian State Medical University, Minsk 220116, Belarus
| | - Michael Veit
- Institut für Virologie, Vet.-Med. Faculty, Free University Berlin, Berlin 14163, Germany
| |
Collapse
|
32
|
Cholesterol Binding to the Transmembrane Region of a Group 2 Hemagglutinin (HA) of Influenza Virus Is Essential for Virus Replication, Affecting both Virus Assembly and HA Fusion Activity. J Virol 2019; 93:JVI.00555-19. [PMID: 31118253 DOI: 10.1128/jvi.00555-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/14/2019] [Indexed: 12/21/2022] Open
Abstract
Hemagglutinin (HA) of influenza virus is incorporated into cholesterol-enriched nanodomains of the plasma membrane. Phylogenetic group 2 HAs contain the conserved cholesterol consensus motif (CCM) YKLW in the transmembrane region. We previously reported that mutations in the CCM retarded intracellular transport of HA and decreased its nanodomain association. Here, we analyzed whether cholesterol interacts with the CCM. Incorporation of photocholesterol into HA was significantly reduced if the whole CCM is replaced by alanine, both using immunoprecipitated HA and when HA is embedded in the membrane. We next used reverse genetics to investigate the significance of the CCM for virus replication. No virus was rescued if the whole motif is exchanged (YKLW4A); singly (LA) or doubly (YK2A and LW2A) mutated virus showed decreased titers and a comparative fitness disadvantage. In polarized cells, transport of HA mutants to the apical membrane was not disturbed. Reduced amounts of HA and cholesterol were incorporated into the viral membrane. Mutant viruses exhibit a decrease in hemolysis, which is only partially corrected if the membrane is replenished with cholesterol. More specifically, viruses have a defect in hemifusion, as demonstrated by fluorescence dequenching. Cells expressing HA YKLW4A fuse with erythrocytes, but the number of events is reduced. Even after acidification unfused erythrocytes remain cell bound, a phenomenon not observed with wild-type HA. We conclude that cholesterol binding to a group 2 HA is essential for virus replication. It has pleiotropic effects on virus assembly and membrane fusion, mainly on lipid mixing and possibly a preceding step.IMPORTANCE The glycoprotein HA is a major pathogenicity factor of influenza viruses. Whereas the structure and function of HA's ectodomain is known in great detail, similar data for the membrane-anchoring part of the protein are missing. Here, we demonstrate that the transmembrane region of a group 2 HA interacts with cholesterol, the major lipid of the plasma membrane and the defining element of the viral budding site nanodomains of the plasma membrane. The cholesterol binding motif is essential for virus replication. Its partial removal affects various steps of the viral life cycle, such as assembly of new virus particles and their subsequent cell entry via membrane fusion. A cholesterol binding pocket in group 2 HAs might be a promising target for a small lipophilic drug that inactivates the virus.
Collapse
|
33
|
Zhu L, Zhao W, Lu J, Li S, Zhou K, Jiang W, Duan X, Fu L, Yu B, Cai KQ, Gao GF, Liu W, Fang M. Influenza virus matrix protein M1 interacts with SLD5 to block host cell cycle. Cell Microbiol 2019; 21:e13038. [PMID: 31050118 DOI: 10.1111/cmi.13038] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 04/16/2019] [Accepted: 04/29/2019] [Indexed: 01/09/2023]
Abstract
Influenza virus matrix 1 protein (M1) is highly conserved and plays essential roles at many stages of virus life cycle. Here, we used a yeast two-hybrid system to identify the host protein SLD5, a component of the GINS complex, which is essential for the initiation of DNA replication in eukaryotic cells, as a new M1 interacting protein. M1 from several different influenza virus strains all interacted with SLD5. Overexpression of SLD5 suppressed influenza virus replication. Transient, stable, or inducible expression of M1 induced host cell cycle blockade at G0/G1 phase. Moreover, SLD5 partially rescued M1 expression- or influenza virus infection-induced G0/G1 phase accumulation in cell lines and primary mouse embryonic fibroblasts. Importantly, SLD5 transgenic mice exhibited higher resistance and improved lung epithelial regeneration after virus infection compared with wild-type mice. Therefore, influenza virus M1 blocks host cell cycle process by interacting with SLD5. Our finding reveals the multifunctional nature of M1 and provides new insight for understanding influenza virus-host interaction.
Collapse
Affiliation(s)
- Li Zhu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Wenming Zhao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Jiao Lu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shan Li
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Kai Zhou
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wei Jiang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Xuefeng Duan
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Lifeng Fu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Bolan Yu
- Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Kathy Q Cai
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, Pennsylvania, USA
| | - George Fu Gao
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Wenjun Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Min Fang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China.,Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.,International College, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
34
|
Zhang Y, Xu C, Zhang H, Liu GD, Xue C, Cao Y. Targeting Hemagglutinin: Approaches for Broad Protection against the Influenza A Virus. Viruses 2019; 11:v11050405. [PMID: 31052339 PMCID: PMC6563292 DOI: 10.3390/v11050405] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/26/2019] [Accepted: 04/27/2019] [Indexed: 12/13/2022] Open
Abstract
Influenza A viruses are dynamically epidemic and genetically diverse. Due to the antigenic drift and shift of the virus, seasonal vaccines are required to be reformulated annually to match with current circulating strains. However, the mismatch between vaccinal strains and circulating strains occurs frequently, resulting in the low efficacy of seasonal vaccines. Therefore, several “universal” vaccine candidates based on the structure and function of the hemagglutinin (HA) protein have been developed to meet the requirement of a broad protection against homo-/heterosubtypic challenges. Here, we review recent novel constructs and discuss several important findings regarding the broad protective efficacy of HA-based universal vaccines.
Collapse
Affiliation(s)
- Yun Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Cong Xu
- Research Center of Agricultural of Dongguan City, Dongguan 523086, China.
| | - Hao Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - George Dacai Liu
- Firstline Biopharmaceuticals Corporation, 12,050 167th PL NE, Redmond, WA 98052, USA.
| | - Chunyi Xue
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| | - Yongchang Cao
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
35
|
Takizawa N, Ogura Y, Fujita Y, Noda T, Shigematsu H, Hayashi T, Kurokawa K. Local structural changes of the influenza A virus ribonucleoprotein complex by single mutations in the specific residues involved in efficient genome packaging. Virology 2019; 531:126-140. [PMID: 30875489 DOI: 10.1016/j.virol.2019.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 11/15/2022]
Abstract
The influenza A virus genome consists of eight single-stranded negative-sense RNA segments. The noncoding regions located at the 3'- and 5'- ends of each segment are necessary for genome packaging, and the terminal coding regions are required to precisely bundle the eight segments. However, the nucleotide residues important for genome bundling are not defined. Here, we introduced premature termination codons in the hemagglutinin (HA) or matrix protein 2 (M2) gene and constructed virus libraries containing random sequences in the terminal coding regions. Using these virus libraries, we identified nucleotide residues involved in efficient virus propagation. Viral genome packaging was impaired in viruses that contained single mutations at these identified residues. Furthermore, these single mutations altered the local structure of the viral ribonucleoprotein complex. Our results show that specific nucleotide residues in the viral protein coding region are involved in forming the precise structure of the viral ribonucleoprotein complex.
Collapse
Affiliation(s)
- Naoki Takizawa
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), Tokyo, Japan.
| | - Yoshitoshi Ogura
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoko Fujita
- Laboratory of Ultrastructural Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan; Laboratory of Ultrastructural Virology, Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takeshi Noda
- Laboratory of Ultrastructural Virology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan; Laboratory of Ultrastructural Virology, Division of Integrated Life Science, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Hideki Shigematsu
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Hyogo, Japan
| | - Tetsuya Hayashi
- Department of Bacteriology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Kurokawa
- Center for Information Biology, National Institute of Genetics, Shizuoka, Japan
| |
Collapse
|
36
|
Kordyukova LV, Shtykova EV, Baratova LA, Svergun DI, Batishchev OV. Matrix proteins of enveloped viruses: a case study of Influenza A virus M1 protein. J Biomol Struct Dyn 2018; 37:671-690. [PMID: 29388479 DOI: 10.1080/07391102.2018.1436089] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Influenza A virus, a member of the Orthomyxoviridae family of enveloped viruses, is one of the human and animal top killers, and its structure and components are therefore extensively studied during the last decades. The most abundant component, M1 matrix protein, forms a matrix layer (scaffold) under the viral lipid envelope, and the functional roles as well as structural peculiarities of the M1 protein are still under heavy debate. Despite multiple attempts of crystallization, no high resolution structure is available for the full length M1 of Influenza A virus. The likely reason for the difficulties lies in the intrinsic disorder of the M1 C-terminal part preventing diffraction quality crystals to be grown. Alternative structural methods including synchrotron small-angle X-ray scattering (SAXS), atomic force microscopy, cryo-electron microscopy/tomography are therefore widely applied to understand the structure of M1, its self-association and interactions with the lipid membrane and the viral nucleocapsid. These methods reveal striking similarities in the behavior of M1 and matrix proteins of other enveloped RNA viruses, with the differences accompanied by the specific features of the viral lifecycles, thus suggesting common interaction principles and, possibly, common evolutional ancestors. The structural information on the Influenza A virus M1 protein obtained to the date strongly suggests that the intrinsic disorder in the C-terminal domain has important functional implications.
Collapse
Affiliation(s)
- Larisa V Kordyukova
- a Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | - Eleonora V Shtykova
- b Shubnikov Institute of Crystallography of Federal Scientific Research Centre 'Crystallography and Photonics' of Russian Academy of Sciences , Moscow , Russian Federation.,c Semenov Institute of Chemical Physics , Russian Academy of Sciences , Moscow , Russian Federation
| | - Lyudmila A Baratova
- a Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University , Moscow , Russian Federation
| | | | - Oleg V Batishchev
- e Frumkin Institute of Physical Chemistry and Electrochemistry , Russian Academy of Sciences , Moscow , Russian Federation.,f Moscow Institute of Physics and Technology , Dolgoprudniy , Russian Federation
| |
Collapse
|
37
|
Sobocińska J, Roszczenko-Jasińska P, Ciesielska A, Kwiatkowska K. Protein Palmitoylation and Its Role in Bacterial and Viral Infections. Front Immunol 2018; 8:2003. [PMID: 29403483 PMCID: PMC5780409 DOI: 10.3389/fimmu.2017.02003] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 12/26/2017] [Indexed: 12/11/2022] Open
Abstract
S-palmitoylation is a reversible, enzymatic posttranslational modification of proteins in which palmitoyl chain is attached to a cysteine residue via a thioester linkage. S-palmitoylation determines the functioning of proteins by affecting their association with membranes, compartmentalization in membrane domains, trafficking, and stability. In this review, we focus on S-palmitoylation of proteins, which are crucial for the interactions of pathogenic bacteria and viruses with the host. We discuss the role of palmitoylated proteins in the invasion of host cells by bacteria and viruses, and those involved in the host responses to the infection. We highlight recent data on protein S-palmitoylation in pathogens and their hosts obtained owing to the development of methods based on click chemistry and acyl-biotin exchange allowing proteomic analysis of protein lipidation. The role of the palmitoyl moiety present in bacterial lipopolysaccharide and lipoproteins, contributing to infectivity and affecting recognition of bacteria by innate immune receptors, is also discussed.
Collapse
Affiliation(s)
- Justyna Sobocińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Paula Roszczenko-Jasińska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Anna Ciesielska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Kwiatkowska
- Laboratory of Molecular Membrane Biology, Department of Cell Biology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
38
|
Palmitoylation Contributes to Membrane Curvature in Influenza A Virus Assembly and Hemagglutinin-Mediated Membrane Fusion. J Virol 2017; 91:JVI.00947-17. [PMID: 28794042 DOI: 10.1128/jvi.00947-17] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/27/2017] [Indexed: 12/31/2022] Open
Abstract
The highly conserved cytoplasmic tail of influenza virus glycoprotein hemagglutinin (HA) contains three cysteines, posttranslationally modified by covalently bound fatty acids. While viral HA acylation is crucial in virus replication, its physico-chemical role is unknown. We used virus-like particles (VLP) to study the effect of acylation on morphology, protein incorporation, lipid composition, and membrane fusion. Deacylation interrupted HA-M1 interactions since deacylated mutant HA failed to incorporate an M1 layer within spheroidal VLP, and filamentous particles incorporated increased numbers of neuraminidase (NA). While HA acylation did not influence VLP shape, lipid composition, or HA lateral spacing, acylation significantly affected envelope curvature. Compared to wild-type HA, deacylated HA is correlated with released particles with flat envelope curvature in the absence of the matrix (M1) protein layer. The spontaneous curvature of palmitate was calculated by molecular dynamic simulations and was found to be comparable to the curvature values derived from VLP size distributions. Cell-cell fusion assays show a strain-independent failure of fusion pore enlargement among H2 (A/Japan/305/57), H3 (A/Aichi/2/68), and H3 (A/Udorn/72) viruses. In contradistinction, acylation made no difference in the low-pH-dependent fusion of isolated VLPs to liposomes: fusion pores formed and expanded, as demonstrated by the presence of complete fusion products observed using cryo-electron tomography (cryo-ET). We propose that the primary mechanism of action of acylation is to control membrane curvature and to modify HA's interaction with M1 protein, while the stunting of fusion by deacylated HA acting in isolation may be balanced by other viral proteins which help lower the energetic barrier to pore expansion.IMPORTANCE Influenza A virus is an airborne pathogen causing seasonal epidemics and occasional pandemics. Hemagglutinin (HA), a glycoprotein abundant on the virion surface, is important in both influenza A virus assembly and entry. HA is modified by acylation whose removal abrogates viral replication. Here, we used cryo-electron tomography to obtain three-dimensional images to elucidate a role for HA acylation in VLP assembly. Our data indicate that HA acylation contributes to the capability of HA to bend membranes and to HA's interaction with the M1 scaffold protein during virus assembly. Furthermore, our data on VLP and, by hypothesis, virus suggests that HA acylation, while not critical to fusion pore formation, contributes to pore expansion in a target-dependent fashion.
Collapse
|
39
|
Gardner AS, Rahman IA, Lai CT, Hepworth A, Trengove N, Hartmann PE, Geddes DT. Changes in Fatty Acid Composition of Human Milk in Response to Cold-Like Symptoms in the Lactating Mother and Infant. Nutrients 2017; 9:nu9091034. [PMID: 28925980 PMCID: PMC5622794 DOI: 10.3390/nu9091034] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/11/2017] [Accepted: 09/14/2017] [Indexed: 11/16/2022] Open
Abstract
Infants rely on their innate immune systems to protect them from infection. Human milk (HM) contains fatty acids (FAs) and monoacylglycerols that are known to exhibit antiviral and antibacterial properties in vitro. The specific fat content of HM may potentially affect the efficacy of this antimicrobial activity. This preliminary study investigates whether the proportions of FA in HM change in response to infections, leading to cold-like symptoms in the mother or infant. Milk samples were obtained from mothers (n = 26) when they and their infants were healthy, and when mother, infant, or both suffered cold-like symptoms. The milk was hydrolysed and FA proportions were measured using gas chromatography. Fifteen FAs were recorded, of which eight were detected in sufficient quantities for statistical analysis. The proportions of capric (C10:0) and lauric acids (C12:0) in HM were significantly lower, and palmitic acid (C16:0) was higher when mothers and infants were ill compared to healthy samples. Palmitoleic (C16:1, n-7) and stearic acid (C18:0) proportions were higher in HM when the infant was unwell, but were not related to maternal health. Whilst the differences detected were small (less than 0.5%), the effects may be additive and potentially have a protective function. The value of further studies is certainly indicated.
Collapse
Affiliation(s)
- Andrew S Gardner
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| | - Ibrahim A Rahman
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| | - Ching T Lai
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| | - Anna Hepworth
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| | - Naomi Trengove
- School of Health and Sciences, The University of Notre Dame Australia, 32 Mouat Street, Fremantle, WA 6959, Australia.
| | - Peter E Hartmann
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Crawley, Perth, WA 6009, Australia.
| |
Collapse
|
40
|
Kordyukova L. Structural and functional specificity of Influenza virus haemagglutinin and paramyxovirus fusion protein anchoring peptides. Virus Res 2017; 227:183-199. [DOI: 10.1016/j.virusres.2016.09.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/21/2016] [Accepted: 09/23/2016] [Indexed: 02/08/2023]
|
41
|
WANG ZJ, GAO X, YANG JB, SUN WC, WU DL, PENG QS, LIU N. Mass Spectrometric Analysis of S-palmitoylation of Hemagglutinin from Influenza A Virus. CHINESE JOURNAL OF ANALYTICAL CHEMISTRY 2016. [DOI: 10.1016/s1872-2040(16)60962-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
42
|
Takizawa N, Momose F, Morikawa Y, Nomoto A. Influenza A Virus Hemagglutinin is Required for the Assembly of Viral Components Including Bundled vRNPs at the Lipid Raft. Viruses 2016; 8:v8090249. [PMID: 27626438 PMCID: PMC5035963 DOI: 10.3390/v8090249] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/01/2016] [Accepted: 09/02/2016] [Indexed: 11/16/2022] Open
Abstract
The influenza glycoproteins, hemagglutinin (HA) and neuraminidase (NA), which are associated with the lipid raft, have the potential to initiate virion budding. However, the role of these viral proteins in infectious virion assembly is still unclear. In addition, it is not known how the viral ribonucleoprotein complex (vRNP) is tethered to the budding site. Here, we show that HA is necessary for the efficient progeny virion production and vRNP packaging in the virion. We also found that the level of HA does not affect the bundling of the eight vRNP segments, despite reduced virion production. Detergent solubilization and a subsequent membrane flotation analysis indicated that the accumulation of nucleoprotein, viral polymerases, NA, and matrix protein 1 (M1) in the lipid raft fraction was delayed without HA. Based on our results, we inferred that HA plays a role in the accumulation of viral components, including bundled vRNPs, at the lipid raft.
Collapse
Affiliation(s)
- Naoki Takizawa
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan.
| | - Fumitaka Momose
- Kitasato Institute for Life Sciences and Graduate School for Infection Control, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Yuko Morikawa
- Kitasato Institute for Life Sciences and Graduate School for Infection Control, Kitasato University, 5-9-1 Shirokane, Minato-ku, Tokyo 108-8641, Japan.
| | - Akio Nomoto
- Laboratory of Virology, Institute of Microbial Chemistry (BIKAKEN), 3-14-23 Kamiosaki, Shinagawa-ku, Tokyo 141-0021, Japan.
| |
Collapse
|
43
|
Pohl MO, Lanz C, Stertz S. Late stages of the influenza A virus replication cycle-a tight interplay between virus and host. J Gen Virol 2016; 97:2058-2072. [PMID: 27449792 DOI: 10.1099/jgv.0.000562] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
After successful infection and replication of its genome in the nucleus of the host cell, influenza A virus faces several challenges before newly assembled viral particles can bud off from the plasma membrane, giving rise to a new infectious virus. The viral ribonucleoprotein (vRNP) complexes need to exit from the nucleus and be transported to the virus assembly sites at the plasma membrane. Moreover, they need to be bundled to ensure the incorporation of precisely one of each of the eight viral genome segments into newly formed viral particles. Similarly, viral envelope glycoproteins and other viral structural proteins need to be targeted to virus assembly sites for viral particles to form and bud off from the plasma membrane. During all these steps influenza A virus heavily relies on a tight interplay with its host, exploiting host-cell proteins for its own purposes. In this review, we summarize current knowledge on late stages of the influenza virus replication cycle, focusing on the role of host-cell proteins involved in this process.
Collapse
Affiliation(s)
- Marie O Pohl
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Caroline Lanz
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Silke Stertz
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
44
|
Narayanan S, Nieh AH, Kenwood BM, Davis CA, Tosello-Trampont AC, Elich TD, Breazeale SD, Ward E, Anderson RJ, Caldwell SH, Hoehn KL, Hahn YS. Distinct Roles for Intracellular and Extracellular Lipids in Hepatitis C Virus Infection. PLoS One 2016; 11:e0156996. [PMID: 27280294 PMCID: PMC4900644 DOI: 10.1371/journal.pone.0156996] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/23/2016] [Indexed: 12/20/2022] Open
Abstract
Hepatitis C is a chronic liver disease that contributes to progressive metabolic dysfunction. Infection of hepatocytes by hepatitis C virus (HCV) results in reprogramming of hepatic and serum lipids. However, the specific contribution of these distinct pools of lipids to HCV infection remains ill defined. In this study, we investigated the role of hepatic lipogenesis in HCV infection by targeting the rate-limiting step in this pathway, which is catalyzed by the acetyl-CoA carboxylase (ACC) enzymes. Using two structurally unrelated ACC inhibitors, we determined that blockade of lipogenesis resulted in reduced viral replication, assembly, and release. Supplementing exogenous lipids to cells treated with ACC inhibitors rescued HCV assembly with no effect on viral replication and release. Intriguingly, loss of viral RNA was not recapitulated at the protein level and addition of 2-bromopalmitate, a competitive inhibitor of protein palmitoylation, mirrored the effects of ACC inhibitors on reduced viral RNA without a concurrent loss in protein expression. These correlative results suggest that newly synthesized lipids may have a role in protein palmitoylation during HCV infection.
Collapse
Affiliation(s)
- Sowmya Narayanan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, United States of America
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, United States of America
| | - Albert H. Nieh
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, United States of America
| | - Brandon M. Kenwood
- Department of Pharmacology, University of Virginia, Charlottesville, United States of America
| | - Christine A. Davis
- Department of Biology, University of Richmond, Richmond, United States of America
| | | | - Tedd D. Elich
- Cropsolution Inc., Morrisville, United States of America
| | | | - Eric Ward
- Cropsolution Inc., Morrisville, United States of America
| | | | - Stephen H. Caldwell
- Division of Gastroenterology and Hepatology, University of Virginia, Charlottesville, United States of America
| | - Kyle L. Hoehn
- Department of Pharmacology, University of Virginia, Charlottesville, United States of America
| | - Young S. Hahn
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, United States of America
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, United States of America
| |
Collapse
|
45
|
Wang M, Ludwig K, Böttcher C, Veit M. The role of stearate attachment to the hemagglutinin-esterase-fusion glycoprotein HEF of influenza C virus. Cell Microbiol 2016; 18:692-704. [PMID: 26518983 DOI: 10.1111/cmi.12541] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 10/20/2015] [Accepted: 10/27/2015] [Indexed: 12/13/2022]
Abstract
The only spike of influenza C virus, the hemagglutinin-esterase-fusion glycoprotein (HEF) combines receptor binding, receptor hydrolysis and membrane fusion activities. Like other hemagglutinating glycoproteins of influenza viruses HEF is S-acylated, but only with stearic acid at a single cysteine located at the cytosol-facing end of the transmembrane region. Previous studies established the essential role of S-acylation of hemagglutinin for replication of influenza A and B virus by affecting budding and/or membrane fusion, but the function of acylation of HEF was hitherto not investigated. Using reverse genetics we rescued a virus containing non-stearoylated HEF, which was stable during serial passage and showed no competitive fitness defect, but the growth rate of the mutant virus was reduced by one log. Deacylation of HEF does neither affect the kinetics of its plasma membrane transport nor the protein composition of virus particles. Cryo-electron microscopy showed that the shape of viral particles and the hexagonal array of spikes typical for influenza C virus were not influenced by this mutation indicating that virus budding was not disturbed. However, the extent and kinetics of haemolysis were reduced in mutant virus at 37°C, but not at 33°C, the optimal temperature for virus growth, suggesting that non-acylated HEF has a defect in membrane fusion under suboptimal conditions.
Collapse
Affiliation(s)
- Mingyang Wang
- Institute of Virology, Department of Veterinary Medicine, Free University Berlin, Berlin, Germany
| | - Kai Ludwig
- Research Center of Electron Microscopy, Department of Chemistry, Free University Berlin, Berlin, Germany
| | - Christoph Böttcher
- Research Center of Electron Microscopy, Department of Chemistry, Free University Berlin, Berlin, Germany
| | - Michael Veit
- Institute of Virology, Department of Veterinary Medicine, Free University Berlin, Berlin, Germany
| |
Collapse
|
46
|
Chlanda P, Zimmerberg J. Protein-lipid interactions critical to replication of the influenza A virus. FEBS Lett 2016; 590:1940-54. [PMID: 26921878 DOI: 10.1002/1873-3468.12118] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 02/08/2016] [Accepted: 02/21/2016] [Indexed: 12/12/2022]
Abstract
Influenza A virus (IAV) assembles on the plasma membrane where viral proteins localize to form a bud encompassing the viral genome, which ultimately pinches off to give rise to newly formed infectious virions. Upon entry, the virus faces the opposite task-fusion with the endosomal membrane and disassembly to deliver the viral genome to the cytoplasm. There are at least four influenza proteins-hemagglutinin (HA), neuraminidase (NA), matrix 1 protein (M1), and the M2 ion channel-that are known to directly interact with the cellular membrane and modify membrane curvature in order to both assemble and disassemble membrane-enveloped virions. Here, we summarize and discuss current knowledge of the interactions of lipids and membrane proteins involved in the IAV replication cycle.
Collapse
Affiliation(s)
- Petr Chlanda
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
47
|
Wang M, Veit M. Hemagglutinin-esterase-fusion (HEF) protein of influenza C virus. Protein Cell 2016; 7:28-45. [PMID: 26215728 PMCID: PMC4707155 DOI: 10.1007/s13238-015-0193-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 07/06/2015] [Indexed: 01/19/2023] Open
Abstract
Influenza C virus, a member of the Orthomyxoviridae family, causes flu-like disease but typically only with mild symptoms. Humans are the main reservoir of the virus, but it also infects pigs and dogs. Very recently, influenza C-like viruses were isolated from pigs and cattle that differ from classical influenza C virus and might constitute a new influenza virus genus. Influenza C virus is unique since it contains only one spike protein, the hemagglutinin-esterase-fusion glycoprotein HEF that possesses receptor binding, receptor destroying and membrane fusion activities, thus combining the functions of Hemagglutinin (HA) and Neuraminidase (NA) of influenza A and B viruses. Here we briefly review the epidemiology and pathology of the virus and the morphology of virus particles and their genome. The main focus is on the structure of the HEF protein as well as on its co- and post-translational modification, such as N-glycosylation, disulfide bond formation, S-acylation and proteolytic cleavage into HEF1 and HEF2 subunits. Finally, we describe the functions of HEF: receptor binding, esterase activity and membrane fusion.
Collapse
Affiliation(s)
- Mingyang Wang
- Institute of Virology, Veterinary Medicine, Free University Berlin, Berlin, Germany
| | - Michael Veit
- Institute of Virology, Veterinary Medicine, Free University Berlin, Berlin, Germany.
| |
Collapse
|
48
|
Siche S, Brett K, Möller L, Kordyukova LV, Mintaev RR, Alexeevski AV, Veit M. Two Cytoplasmic Acylation Sites and an Adjacent Hydrophobic Residue, but No Other Conserved Amino Acids in the Cytoplasmic Tail of HA from Influenza A Virus Are Crucial for Virus Replication. Viruses 2015; 7:6458-75. [PMID: 26670246 PMCID: PMC4690873 DOI: 10.3390/v7122950] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Revised: 11/24/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023] Open
Abstract
Recruitment of the matrix protein M1 to the assembly site of the influenza virus is thought to be mediated by interactions with the cytoplasmic tail of hemagglutinin (HA). Based on a comprehensive sequence comparison of all sequences present in the database, we analyzed the effect of mutating conserved residues in the cytosol-facing part of the transmembrane region and cytoplasmic tail of HA (A/WSN/33 (H1N1) strain) on virus replication and morphology of virions. Removal of the two cytoplasmic acylation sites and substitution of a neighboring isoleucine by glutamine prevented rescue of infectious virions. In contrast, a conservative exchange of the same isoleucine, non-conservative exchanges of glycine and glutamine, deletion of the acylation site at the end of the transmembrane region and shifting it into the tail did not affect virus morphology and had only subtle effects on virus growth and on the incorporation of M1 and Ribo-Nucleoprotein Particles (RNPs). Thus, assuming that essential amino acids are conserved between HA subtypes we suggest that, besides the two cytoplasmic acylation sites (including adjacent hydrophobic residues), no other amino acids in the cytoplasmic tail of HA are indispensable for virus assembly and budding.
Collapse
Affiliation(s)
- Stefanie Siche
- Institute of Virology, Faculty of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany.
| | - Katharina Brett
- Institute of Virology, Faculty of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany.
| | - Lars Möller
- Robert Koch Institute, Advanced Light and Electron Microscopy (ZBS4), Nordufer 20, 13353 Berlin, Germany.
| | - Larisa V Kordyukova
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Ramil R Mintaev
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- I.I. Mechnikov Research Institute of Vaccines and Sera, Russian Academy of Medical Sciences, 105064 Moscow, Russia.
| | - Andrei V Alexeevski
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia.
- Department of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia.
| | - Michael Veit
- Institute of Virology, Faculty of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany.
| |
Collapse
|
49
|
S-acylation of influenza virus proteins: Are enzymes for fatty acid attachment promising drug targets? Vaccine 2015; 33:7002-7. [DOI: 10.1016/j.vaccine.2015.08.095] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 05/10/2015] [Accepted: 08/28/2015] [Indexed: 11/22/2022]
|
50
|
Scolari S, Imkeller K, Jolmes F, Veit M, Herrmann A, Schwarzer R. Modulation of cell surface transport and lipid raft localization by the cytoplasmic tail of the influenza virus hemagglutinin. Cell Microbiol 2015; 18:125-36. [PMID: 26243691 PMCID: PMC7162421 DOI: 10.1111/cmi.12491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Revised: 06/28/2015] [Accepted: 07/13/2015] [Indexed: 11/30/2022]
Abstract
Viral glycoproteins are highly variable in their primary structure, but on the other hand feature a high functional conservation to fulfil their versatile tasks during the pathogenic life cycle. Typically, all protein domains are optimized in that indispensable functions can be assigned to small conserved motifs or even individual amino acids. The cytoplasmic tail of many viral spike proteins, although of particular relevance for the virus biology, is often only insufficiently characterized. Hemagglutinin (HA), the receptor-binding protein of the influenza virus comprises a short cytoplasmic tail of 13 amino acids that exhibits three highly conserved palmitoylation sites. However, the particular importance of these modifications and the tail in general for intracellular trafficking and lateral membrane organization remains elusive. In this study, we generated HA core proteins consisting of transmembrane domain, cytoplasmic tail and a minor part of the ectodomain, tagged with a yellow fluorescent protein. Different mutation and truncation variants of these chimeric proteins were investigated using confocal microscopy, to characterize the role of cytoplasmic tail and palmitoylation for the intracellular trafficking to plasma membrane and Golgi apparatus. In addition, we assessed raft partitioning of the variants by Foerster resonance energy transfer with an established raft marker. We revealed a substantial influence of the cytoplasmic tail length on the intracellular distribution and surface exposure of the proteins. A complete removal of the tail hampers a physiological trafficking of the protein, whereas a partial truncation can be compensated by cytoplasmic palmitoylations. Plasma membrane raft partitioning on the other hand was found to imperatively require palmitoylations, and the cysteine at position 551 turned out to be of most relevance. Our data shed further light on the tight interconnection between cytoplasmic elements and intracellular trafficking and suggest a function of HA palmitoylations in both lateral sorting and anterograde trafficking of the glycoprotein.
Collapse
Affiliation(s)
- Silvia Scolari
- Department of Biology, Molecular Biophysics, Humboldt University Berlin, 10115, Berlin, Germany
| | - Katharina Imkeller
- Department of Biology, Molecular Biophysics, Humboldt University Berlin, 10115, Berlin, Germany
| | - Fabian Jolmes
- Department of Biology, Molecular Biophysics, Humboldt University Berlin, 10115, Berlin, Germany
| | - Michael Veit
- Department of Immunology and Molecular Biology, Free University, 14163, Berlin, Germany
| | - Andreas Herrmann
- Department of Biology, Molecular Biophysics, Humboldt University Berlin, 10115, Berlin, Germany
| | - Roland Schwarzer
- Department of Biology, Molecular Biophysics, Humboldt University Berlin, 10115, Berlin, Germany.,Department of Biological Chemistry, Weizmann Institute of Science, 76100, Rehovot, Israel
| |
Collapse
|