152 results on '"Margaret Kielian"'
Search Results
2. Rubella virus assembly requirements and evolutionary relationships with novel rubiviruses
- Author
-
Pratyush Kumar Das and Margaret Kielian
- Subjects
rubella virus ,rubivirus ,virus assembly ,virus budding ,Microbiology ,QR1-502 - Abstract
ABSTRACT Rubella virus (RuV) is an enveloped virus that usually causes mild disease in children, but can produce miscarriage or severe congenital birth defects. While in nature RuV only infects humans, the discovery of the related Ruhugu (RuhV) and Rustrela (RusV) viruses highlights the spillover potential of mammalian rubiviruses to humans. RuV buds into the Golgi, but its assembly and exit are not well understood. We identified a potential late domain motif 278PPAY281 at the C-terminus of the RuV E2 envelope protein. Such late domain motifs can promote virus budding by recruiting the cellular ESCRT machinery. An E2 Y281A mutation reduced infectious virus production by >3 logs and inhibited virus particle production. However, RuV was insensitive to inhibition by dominant-negative VPS4, and thus appeared ESCRT-independent. The E2 Y281A mutation did not significantly inhibit the production of the viral structural proteins capsid (Cp), E2, and E1, or dimerization, glycosylation, Golgi transport, and colocalization of E2 and E1. However, E2 Y281A significantly reduced glycoprotein-Cp colocalization and interaction, and inhibited Cp localization to the Golgi. Revertants of the E2 Y281A mutant contained an E2 281V substitution or the second site mutations [E2 N277I + Cp D215A]. These mutations promoted virus growth, particle production, E2/Cp colocalization and Cp-Golgi localization. Both the E2 substitutions 281V and 277I were found at the corresponding positions in the RuhV E2 protein. Taken together, our data identify a key interaction of the RuV E2 endodomain with the Cp during RuV biogenesis, and support the close evolutionary relationship between human and animal rubiviruses.IMPORTANCERubella virus (RuV) is an enveloped virus that only infects humans, where transplacental infection can cause miscarriage or congenital birth defects. We identified a potential late domain, 278PPAY281, at the C terminus of the E2 envelope protein. However, rather than this domain recruiting the cellular ESCRT machinery as predicted, our data indicate that E2 Y281 promotes a critical interaction of the E2 endodomain with the capsid protein, leading to capsid's localization to the Golgi where virus budding occurs. Revertant analysis demonstrated that two substitutions on the E2 protein could partially rescue virus growth and Cp-Golgi localization. Both residues were found at the corresponding positions in Ruhugu virus E2, supporting the close evolutionary relationship between RuV and Ruhugu virus, a recently discovered rubivirus from bats.
- Published
- 2024
- Full Text
- View/download PDF
3. 4′-Fluorouridine inhibits alphavirus replication and infection in vitro and in vivo
- Author
-
Peiqi Yin, Nicholas A. May, Laura Sandra Lello, Atef Fayed, M. Guston Parks, Adam M. Drobish, Sainan Wang, Meghan Andrews, Zachary Sticher, Alexander A. Kolykhalov, Michael G. Natchus, George R. Painter, Andres Merits, Margaret Kielian, and Thomas E. Morrison
- Subjects
alphavirus ,chikungunya virus ,mayaro virus ,antiviral ,RNA replication ,Microbiology ,QR1-502 - Abstract
ABSTRACT Chikungunya virus (CHIKV) is an enveloped, positive-sense RNA virus that has re-emerged to cause millions of human infections worldwide. In humans, acute CHIKV infection causes fever and severe muscle and joint pain. Chronic and debilitating arthritis and joint pain can persist for months to years. To date, there are no approved antivirals against CHIKV. Recently, the ribonucleoside analog 4′-fluorouridine (4′-FlU) was reported as a highly potent orally available inhibitor of SARS-CoV-2, respiratory syncytial virus, and influenza virus replication. In this study, we assessed 4′-FlU’s potency and breadth of inhibition against a panel of alphaviruses including CHIKV, and found that it broadly suppressed alphavirus production in cell culture. 4′-FlU acted on the viral RNA replication step, and the first 4 hours post-infection were the critical time for its antiviral effect. In vitro replication assays identified nsP4 as the target of inhibition. In vivo, treatment with 4′-FlU reduced disease signs, inflammatory responses, and viral tissue burden in mouse models of CHIKV and Mayaro virus infection. Treatment initiated at 2 hours post-infection was most effective; however, treatment initiated as late as 24–48 hours post-infection produced measurable antiviral effects in the CHIKV mouse model. 4′-FlU showed effective oral delivery in our mouse model and resulted in the accumulation of both 4′-FlU and its bioactive triphosphate form in tissues relevant to arthritogenic alphavirus pathogenesis. Together, our data indicate that 4′-FlU inhibits CHIKV infection in vitro and in vivo and is a promising oral therapeutic candidate against CHIKV infection.IMPORTANCEAlphaviruses including chikungunya virus (CHIKV) are mosquito-borne positive-strand RNA viruses that can cause various diseases in humans. Although compounds that inhibit CHIKV and other alphaviruses have been identified in vitro, there are no licensed antivirals against CHIKV. Here, we investigated a ribonucleoside analog, 4′-fluorouridine (4′-FlU), and demonstrated that it inhibited infectious virus production by several alphaviruses in vitro and reduced virus burden in mouse models of CHIKV and Mayaro virus infection. Our studies also indicated that 4′-FlU treatment reduced CHIKV-induced footpad swelling and reduced the production of pro-inflammatory cytokines. Inhibition in the mouse model correlated with effective oral delivery of 4′-FlU and accumulation of both 4′-FlU and its bioactive form in relevant tissues. In summary, 4′-FlU exhibits potential as a novel anti-alphavirus agent targeting the replication of viral RNA.
- Published
- 2024
- Full Text
- View/download PDF
4. A single-point mutation in the rubella virus E1 glycoprotein promotes rescue of recombinant vesicular stomatitis virus
- Author
-
Pratyush Kumar Das, Paulina Alatriste Gonzalez, Rohit K. Jangra, Peiqi Yin, and Margaret Kielian
- Subjects
rubella ,rubivirus ,vesicular stomatitis virus ,virus budding ,virus fusion ,Microbiology ,QR1-502 - Abstract
ABSTRACT Rubella virus (RuV) is an enveloped plus-sense RNA virus and a member of the Rubivirus genus. RuV infection in pregnant women can lead to miscarriage or an array of severe birth defects known as congenital rubella syndrome. Novel rubiviruses were recently discovered in various mammals, highlighting the spillover potential of other rubiviruses to humans. Many features of the rubivirus infection cycle remain unexplored. To promote the study of rubivirus biology, here, we generated replication-competent recombinant VSV-RuV (rVSV-RuV) encoding the RuV transmembrane glycoproteins E2 and E1. Sequencing of rVSV-RuV showed that the RuV glycoproteins acquired a single-point mutation W448R in the E1 transmembrane domain. The E1 W448R mutation did not detectably alter the intracellular expression, processing, glycosylation, colocalization, or dimerization of the E2 and E1 glycoproteins. Nonetheless, the mutation enhanced the incorporation of RuV E2/E1 into VSV particles, which bud from the plasma membrane rather than the RuV budding site in the Golgi. Neutralization by E1 antibodies, calcium dependence, and cell tropism were comparable between WT-RuV and either rVSV-RuV or RuV containing the E1 W448R mutation. However, the E1 W448R mutation strongly shifted the threshold for the acid pH-triggered virus fusion reaction, from pH 6.2 for the WT RuV to pH 5.5 for the mutant. These results suggest that the increased resistance of the mutant RuV E1 to acidic pH promotes the ability of viral envelope proteins to generate infectious rVSV and provide insights into the regulation of RuV fusion during virus entry and exit.IMPORTANCERubella virus (RuV) infection in pregnant women can cause miscarriage or severe fetal birth defects. While a highly effective vaccine has been developed, RuV cases are still a significant problem in areas with inadequate vaccine coverage. In addition, related viruses have recently been discovered in mammals, such as bats and mice, leading to concerns about potential virus spillover to humans. To facilitate studies of RuV biology, here, we generated and characterized a replication-competent vesicular stomatitis virus encoding the RuV glycoproteins (rVSV-RuV). Sequence analysis of rVSV-RuV identified a single-point mutation in the transmembrane region of the E1 glycoprotein. While the overall properties of rVSV-RuV are similar to those of WT-RuV, the mutation caused a marked shift in the pH dependence of virus membrane fusion. Together, our studies of rVSV-RuV and the identified W448R mutation expand our understanding of rubivirus biology and provide new tools for its study.
- Published
- 2024
- Full Text
- View/download PDF
5. Multiple capsid protein binding sites mediate selective packaging of the alphavirus genomic RNA
- Author
-
Rebecca S. Brown, Dimitrios G. Anastasakis, Markus Hafner, and Margaret Kielian
- Subjects
Science - Abstract
Alphaviruses need to selectively package genomic viral RNA for transmission, but the packaging mechanism remains unclear. Here, Brown et al. combine PAR-CLIP with biotinylated capsid protein (Cp) retrieval and identify multiple Cp binding sites on genomic viral RNA that promote virion formation.
- Published
- 2020
- Full Text
- View/download PDF
6. Transcriptional and Translational Dynamics of Zika and Dengue Virus Infection
- Author
-
Kamini Singh, Maria Guadalupe Martinez, Jianan Lin, James Gregory, Trang Uyen Nguyen, Rawan Abdelaal, Kristy Kang, Kristen Brennand, Arnold Grünweller, Zhengqing Ouyang, Hemali Phatnani, Margaret Kielian, and Hans-Guido Wendel
- Subjects
ZIKV ,polyamine pathways ,ATF3/CHOP ,BACH1/2 ,dengue ,eIF5A hypusination ,Microbiology ,QR1-502 - Abstract
Zika virus (ZIKV) and dengue virus (DENV) are members of the Flaviviridae family of RNA viruses and cause severe disease in humans. ZIKV and DENV share over 90% of their genome sequences, however, the clinical features of Zika and dengue infections are very different reflecting tropism and cellular effects. Here, we used simultaneous RNA sequencing and ribosome footprinting to define the transcriptional and translational dynamics of ZIKV and DENV infection in human neuronal progenitor cells (hNPCs). The gene expression data showed induction of aminoacyl tRNA synthetases (ARS) and the translation activating PIM1 kinase, indicating an increase in RNA translation capacity. The data also reveal activation of different cell stress responses, with ZIKV triggering a BACH1/2 redox program, and DENV activating the ATF/CHOP endoplasmic reticulum (ER) stress program. The RNA translation data highlight activation of polyamine metabolism through changes in key enzymes and their regulators. This pathway is needed for eIF5A hypusination and has been implicated in viral translation and replication. Concerning the viral RNA genomes, ribosome occupancy readily identified highly translated open reading frames and a novel upstream ORF (uORF) in the DENV genome. Together, our data highlight both the cellular stress response and the activation of RNA translation and polyamine metabolism during DENV and ZIKV infection.
- Published
- 2022
- Full Text
- View/download PDF
7. Berberine Chloride is an Alphavirus Inhibitor That Targets Nucleocapsid Assembly
- Author
-
Judy J. Wan, Rebecca S. Brown, and Margaret Kielian
- Subjects
berberine ,RNA packaging ,alphavirus ,antiviral inhibitor ,nucleocapsid assembly ,Microbiology ,QR1-502 - Abstract
ABSTRACT Alphaviruses are enveloped positive-sense RNA viruses that can cause serious human illnesses such as polyarthritis and encephalitis. Despite their widespread distribution and medical importance, there are no licensed vaccines or antivirals to combat alphavirus infections. Berberine chloride (BBC) is a pan-alphavirus inhibitor that was previously identified in a replicon-based small-molecule screen. This work showed that BBC inhibits alphavirus replication but also suggested that BBC might have additional effects later in the viral life cycle. Here, we show that BBC has late effects that target the virus nucleocapsid (NC) core. Infected cells treated with BBC late in infection were unable to form stable cytoplasmic NCs or assembly intermediates, as assayed by gradient sedimentation. In vitro studies with recombinant capsid protein (Cp) and purified genomic RNA (gRNA) showed that BBC perturbs core-like particle formation and potentially traps the assembly process in intermediate states. Particles produced from BBC-treated cells were less infectious, despite efficient particle production and only minor decreases in genome packaging. In addition, BBC treatment of free virus particles strongly decreased alphavirus infectivity. In contrast, the infectivity of the negative-sense RNA virus vesicular stomatitis virus was resistant to BBC treatment of infected cells or free virus. Together, our data indicate that BBC alters alphavirus Cp-gRNA interactions and oligomerization and suggest that this may cause defects in NC assembly and in disassembly during subsequent virus entry. Thus, BBC may be considered a novel alphavirus NC assembly inhibitor. IMPORTANCE The alphavirus chikungunya virus (CHIKV) is an example of an emerging human pathogen with increased and rapid global spread. Although an acute CHIKV infection is rarely fatal, many patients suffer from debilitating chronic arthralgia for years. Antivirals against chikungunya and other alphaviruses have been identified in vitro, but to date none have been shown to be efficacious and have been licensed for human use. Here, we investigated a small molecule, berberine chloride (BBC), and showed that it inhibited infectious virus production by several alphaviruses including CHIKV. BBC acted on a late step in the alphavirus exit pathway, namely the formation of the nucleocapsid containing the infectious viral RNA. Better understanding of nucleocapsid formation and its inhibition by BBC will provide important information on the mechanisms of infectious alphavirus production and may enable their future targeting in antiviral strategies.
- Published
- 2020
- Full Text
- View/download PDF
8. Atovaquone and Berberine Chloride Reduce SARS-CoV-2 Replication In Vitro
- Author
-
Bruno A. Rodriguez-Rodriguez, Maria G. Noval, Maria E. Kaczmarek, Kyung Ku Jang, Sara A. Thannickal, Angelica Cifuentes Kottkamp, Rebecca S. Brown, Margaret Kielian, Ken Cadwell, and Kenneth A. Stapleford
- Subjects
antiviral ,coronavirus ,atovaquone ,berberine chloride ,Microbiology ,QR1-502 - Abstract
Epidemic RNA viruses seem to arise year after year leading to countless infections and devastating disease. SARS-CoV-2 is the most recent of these viruses, but there will undoubtedly be more to come. While effective SARS-CoV-2 vaccines are being deployed, one approach that is still missing is effective antivirals that can be used at the onset of infections and therefore prevent pandemics. Here, we screened FDA-approved compounds against SARS-CoV-2. We found that atovaquone, a pyrimidine biosynthesis inhibitor, is able to reduce SARS-CoV-2 infection in human lung cells. In addition, we found that berberine chloride, a plant-based compound used in holistic medicine, was able to inhibit SARS-CoV-2 infection in cells through direct interaction with the virion. Taken together, these studies highlight potential avenues of antiviral development to block emerging viruses. Such proactive approaches, conducted well before the next pandemic, will be essential to have drugs ready for when the next emerging virus hits.
- Published
- 2021
- Full Text
- View/download PDF
9. Specific Recognition of a Stem-Loop RNA Structure by the Alphavirus Capsid Protein
- Author
-
Rebecca S. Brown, Lisa Kim, and Margaret Kielian
- Subjects
alphavirus ,nucleocapsid ,capsid protein ,RNA packaging ,RNA binding ,Microbiology ,QR1-502 - Abstract
Alphaviruses are small enveloped viruses with positive-sense RNA genomes. During infection, the alphavirus capsid protein (Cp) selectively packages and assembles with the viral genomic RNA to form the nucleocapsid core, a process critical to the production of infectious virus. Prior studies of the alphavirus Semliki Forest virus (SFV) showed that packaging and assembly are promoted by Cp binding to multiple high affinity sites on the genomic RNA. Here, we developed an in vitro Cp binding assay based on fluorescently labeled RNA oligos. We used this assay to explore the RNA sequence and structure requirements for Cp binding to site #1, the top binding site identified on the genomic RNA during all stages of virus assembly. Our results identify a stem-loop structure that promotes specific binding of the SFV Cp to site #1 RNA. This structure is also recognized by the Cps of the related alphaviruses chikungunya virus and Ross River virus.
- Published
- 2021
- Full Text
- View/download PDF
10. Human monoclonal antibodies against chikungunya virus target multiple distinct epitopes in the E1 and E2 glycoproteins.
- Author
-
Jose A Quiroz, Ryan J Malonis, Larissa B Thackray, Courtney A Cohen, Jesper Pallesen, Rohit K Jangra, Rebecca S Brown, Daniel Hofmann, Frederick W Holtsberg, Sergey Shulenin, Elisabeth K Nyakatura, Lorellin A Durnell, Vinayak Rayannavar, Johanna P Daily, Andrew B Ward, M Javad Aman, John M Dye, Kartik Chandran, Michael S Diamond, Margaret Kielian, and Jonathan R Lai
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes persistent arthritis in a subset of human patients. We report the isolation and functional characterization of monoclonal antibodies (mAbs) from two patients infected with CHIKV in the Dominican Republic. Single B cell sorting yielded a panel of 46 human mAbs of diverse germline lineages that targeted epitopes within the E1 or E2 glycoproteins. MAbs that recognized either E1 or E2 proteins exhibited neutralizing activity. Viral escape mutations localized the binding epitopes for two E1 mAbs to sites within domain I or the linker between domains I and III; and for two E2 mAbs between the β-connector region and the B-domain. Two of the E2-specific mAbs conferred protection in vivo in a stringent lethal challenge mouse model of CHIKV infection, whereas the E1 mAbs did not. These results provide insight into human antibody response to CHIKV and identify candidate mAbs for therapeutic intervention.
- Published
- 2019
- Full Text
- View/download PDF
11. Surface (S) Layer Proteins of Lactobacillus acidophilus Block Virus Infection via DC-SIGN Interaction
- Author
-
Mariano Prado Acosta, Eileen M. Geoghegan, Bernd Lepenies, Sandra Ruzal, Margaret Kielian, and Maria Guadalupe Martinez
- Subjects
S-layer ,DC-SIGN ,alphavirus ,flavivirus ,Lactobacillus ,Microbiology ,QR1-502 - Abstract
Alphaviruses and flaviviruses are important human pathogens that include Chikungunya virus (CHIKV), Dengue virus (DENV), and Zika virus (ZIKV), which can cause diseases in humans ranging from arthralgia to hemorrhagic fevers and microcephaly. It was previously shown that treatment with surface layer (S-layer) protein, present on the bacterial cell-envelope of Lactobacillus acidophilus, is able to inhibit viral and bacterial infections by blocking the pathogen’s interaction with DC-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN), a trans-membrane protein that is a C-type calcium-dependent lectin. DC-SIGN is known to act as an attachment factor for several viruses including alphaviruses and flaviviruses. In the present study, we used alphaviruses as a model system to dissect the mechanism of S-layer inhibition. We first evaluated the protective effect of S-layer using 3T3 cells, either wild type or stably expressing DC-SIGN, and infecting with the alphaviruses Semliki Forest virus (SFV) and CHIKV and the flaviviruses ZIKV and DENV. DC-SIGN expression significantly enhanced infection by all four viruses. Treatment of the cells with S-layer prior to infection decreased infectivity of all viruses only in cells expressing DC-SIGN. In vitro ELISA experiments showed a direct interaction between S-layer and DC-SIGN; however, confocal microscopy and flow cytometry demonstrated that S-layer binding to the cells was independent of DC-SIGN expression. S-layer protein prevented SFV binding and internalization in DC-SIGN-expressing cells but had no effect on virus binding to DC-SIGN-negative cells. Inhibition of virus binding occurred in a time-dependent manner, with a significant reduction of infection requiring at least a 30-min pre-incubation of S-layer with DC-SIGN-expressing cells. These results suggest that S-layer has a different mechanism of action compared to mannan, a common DC-SIGN-binding compound that has an immediate effect in blocking viral infection. This difference could reflect slower kinetics of S-layer binding to the DC-SIGN present at the plasma membrane (PM). Alternatively, the S-layer/DC-SIGN interaction may trigger the activation of signaling pathways that are required for the inhibition of viral infection. Together our results add important information relevant to the potential use of L. acidophilus S-layer protein as an antiviral therapy.
- Published
- 2019
- Full Text
- View/download PDF
12. BHK-21 Cell Clones Differ in Chikungunya Virus Infection and MXRA8 Receptor Expression
- Author
-
Peiqi Yin and Margaret Kielian
- Subjects
alphavirus ,chikungunya virus ,receptor ,MXRA8 ,BHK-21 cells ,Microbiology ,QR1-502 - Abstract
Baby hamster kidney-21 (BHK-21) cells are widely used to propagate and study many animal viruses using infection and transfection techniques. Among various BHK-21 cell clones, the fibroblast-like BHK-21/C-13 line and the epithelial-like BHK-21/WI-2 line are commonly used cell clones for alphavirus research. Here we report that BHK-21/WI-2 cells were significantly less susceptible to primary infection by the alphavirus chikungunya virus (CHIKV) than were BHK-21/C-13 cells. The electroporation efficiency of alphavirus RNA into BHK-21/WI-2 was also lower than that of BHK-21/C-13. The growth of CHIKV was decreased in BHK-21/WI-2 compared to BHK-21/C-13, while primary infection and growth of the alphavirus Sindbis virus (SINV) were equivalent in the two cell lines. Our results suggested that CHIKV entry could be compromised in BHK-21/WI-2. Indeed, we found that the mRNA level of the CHIKV receptor MXRA8 in BHK-21/WI-2 cells was much lower than that in BHK-21/C-13 cells, and exogenous expression of either human MXRA8 or hamster MXRA8 rescued CHIKV infection. Our results affirm the importance of the MXRA8 receptor for CHIKV infection, and document differences in its expression in two clonal cell lines derived from the original BHK-21 cell cultures. Our results also indicate that CHIKV propagation and entry studies in BHK-21 cells will be significantly more efficient in BHK-21/C-13 than in BHK-21/WI-2 cells.
- Published
- 2021
- Full Text
- View/download PDF
13. An Alphavirus E2 Membrane-Proximal Domain Promotes Envelope Protein Lateral Interactions and Virus Budding
- Author
-
Emily A. Byrd and Margaret Kielian
- Subjects
virus assembly ,virus budding ,alphavirus ,virus entry ,virus fusion ,virus structure ,Microbiology ,QR1-502 - Abstract
ABSTRACT Alphaviruses are members of a group of small enveloped RNA viruses that includes important human pathogens such as Chikungunya virus and the equine encephalitis viruses. The virus membrane is covered by a lattice composed of 80 spikes, each a trimer of heterodimers of the E2 and E1 transmembrane proteins. During virus endocytic entry, the E1 glycoprotein mediates the low-pH-dependent fusion of the virus membrane with the endosome membrane, thus initiating virus infection. While much is known about E1 structural rearrangements during membrane fusion, it is unclear how the E1/E2 dimer dissociates, a step required for the fusion reaction. A recent Alphavirus cryo-electron microscopy reconstruction revealed a previously unidentified D subdomain in the E2 ectodomain, close to the virus membrane. A loop within this region, here referred to as the D-loop, contains two highly conserved histidines, H348 and H352, which were hypothesized to play a role in dimer dissociation. We generated Semliki Forest virus mutants containing the single and double alanine substitutions H348A, H352A, and H348/352A. The three D-loop mutations caused a reduction in virus growth ranging from 1.6 to 2 log but did not significantly affect structural protein biosynthesis or transport, dimer stability, virus fusion, or specific infectivity. Instead, growth reduction was due to inhibition of a late stage of virus assembly at the plasma membrane. The virus particles that are produced show reduced thermostability compared to the wild type. We propose the E2 D-loop as a key region in establishing the E1-E2 contacts that drive glycoprotein lattice formation and promote Alphavirus budding from the plasma membrane. IMPORTANCE Alphavirus infection causes severe and debilitating human diseases for which there are no effective antiviral therapies or vaccines. In order to develop targeted therapeutics, detailed molecular understanding of the viral entry and exit mechanisms is required. In this report, we define the role of the E2 protein juxtamembrane D-loop, which contains highly conserved histidine residues at positions 348 and 352. These histidines do not play an important role in virus fusion and infection. However, mutation of the D-loop histidines causes significant decreases in the assembly and thermostability of Alphavirus particles. Our results suggest that the E2 D-loop interacts with the E1 protein to promote Alphavirus budding.
- Published
- 2017
- Full Text
- View/download PDF
14. BST2/Tetherin Inhibition of Alphavirus Exit
- Author
-
Yaw Shin Ooi, Mathieu Dubé, and Margaret Kielian
- Subjects
alphavirus ,tetherin/BST2 ,virus budding ,rubella virus ,dengue virus ,Microbiology ,QR1-502 - Abstract
Alphaviruses such as chikungunya virus (CHIKV) and Semliki Forest virus (SFV) are small enveloped RNA viruses that bud from the plasma membrane. Tetherin/BST2 is an interferon-induced host membrane protein that inhibits the release of many enveloped viruses via direct tethering of budded particles to the cell surface. Alphaviruses have highly organized structures and exclude host membrane proteins from the site of budding, suggesting that their release might be insensitive to tetherin inhibition. Here, we demonstrated that exogenously-expressed tetherin efficiently inhibited the release of SFV and CHIKV particles from host cells without affecting virus entry and infection. Alphavirus release was also inhibited by the endogenous levels of tetherin in HeLa cells. While rubella virus (RuV) and dengue virus (DENV) have structural similarities to alphaviruses, tetherin inhibited the release of RuV but not DENV. We found that two recently identified tetherin isoforms differing in length at the N-terminus exhibited distinct capabilities in restricting alphavirus release. SFV exit was efficiently inhibited by the long isoform but not the short isoform of tetherin, while both isoforms inhibited vesicular stomatitis virus exit. Thus, in spite of the organized structure of the virus particle, tetherin specifically blocks alphavirus release and shows an interesting isoform requirement.
- Published
- 2015
- Full Text
- View/download PDF
15. Intercellular Extensions Are Induced by the Alphavirus Structural Proteins and Mediate Virus Transmission.
- Author
-
Maria Guadalupe Martinez and Margaret Kielian
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Alphaviruses are highly organized enveloped RNA viruses with an internal nucleocapsid surrounded by a membrane containing the E2 and E1 transmembrane proteins. Alphavirus budding takes place at the plasma membrane and requires the interaction of the cytoplasmic domain of E2 with the capsid protein. Here we used WT alphaviruses and Sindbis virus in which E2 was fused to a fluorescent protein to characterize virus exit from host cells. Our results show that alphavirus infection induced striking modifications of the host cell cytoskeleton and resulted in the formation of stable intercellular extensions that emanated exclusively from the infected cell. The intercellular extensions were long (> 10 μM), contained actin and tubulin, and formed flattened contacts with neighboring cells, but did not mediate membrane or cytoplasmic continuity between cells. Receptor down-regulation studies indicated that formation of stable extensions did not require the virus receptor, and that extensions promoted cell-to-cell virus transmission to receptor-depleted cells. Virus mutant experiments demonstrated that formation of extensions required the E2-capsid interaction but not active particle budding, while intercellular transmission of infection required the production of fusion-active virus particles. Protein expression studies showed that even in the absence of virus infection, the viral structural proteins alone induced intercellular extensions, and that these extensions were preferentially targeted to non-expressing cells. Together, our results identify a mechanism for alphavirus cell-to-cell transmission and define the key viral protein interactions that it requires.
- Published
- 2016
- Full Text
- View/download PDF
16. Alphavirus Entry and Membrane Fusion
- Author
-
Margaret Kielian, Chantal Chanel-Vos, and Maofu Liao
- Subjects
virus entry ,membrane fusion ,alphavirus ,class II fusion protein ,endocytosis ,Microbiology ,QR1-502 - Abstract
The study of enveloped animal viruses has greatly advanced our understanding of the general properties of membrane fusion and of the specific pathways that viruses use to infect the host cell. The membrane fusion proteins of the alphaviruses and flaviviruses have many similarities in structure and function. As reviewed here, alphaviruses use receptor-mediated endocytic uptake and low pH-triggered membrane fusion to deliver their RNA genomes into the cytoplasm. Recent advances in understanding the biochemistry and structure of the alphavirus membrane fusion protein provide a clearer picture of this fusion reaction, including the protein’s conformational changes during fusion and the identification of key domains. These insights into the alphavirus fusion mechanism suggest new areas for experimental investigation and potential inhibitor strategies for anti-viral therapy.
- Published
- 2010
- Full Text
- View/download PDF
17. Haploid Genetic Screen Reveals a Profound and Direct Dependence on Cholesterol for Hantavirus Membrane Fusion
- Author
-
Lara M. Kleinfelter, Rohit K. Jangra, Lucas T. Jae, Andrew S. Herbert, Eva Mittler, Katie M. Stiles, Ariel S. Wirchnianski, Margaret Kielian, Thijn R. Brummelkamp, John M. Dye, and Kartik Chandran
- Subjects
Microbiology ,QR1-502 - Abstract
ABSTRACT Hantaviruses cause hemorrhagic fever with renal syndrome (HFRS) in the Old World and a highly fatal hantavirus cardiopulmonary syndrome (HCPS) in the New World. No vaccines or antiviral therapies are currently available to prevent or treat hantavirus disease, and gaps in our understanding of how hantaviruses enter cells challenge the search for therapeutics. We performed a haploid genetic screen in human cells to identify host factors required for entry by Andes virus, a highly virulent New World hantavirus. We found that multiple genes involved in cholesterol sensing, regulation, and biosynthesis, including key components of the sterol response element-binding protein (SREBP) pathway, are critical for Andes virus entry. Genetic or pharmacological disruption of the membrane-bound transcription factor peptidase/site-1 protease (MBTPS1/S1P), an SREBP control element, dramatically reduced infection by virulent hantaviruses of both the Old World and New World clades but not by rhabdoviruses or alphaviruses, indicating that this pathway is broadly, but selectively, required by hantaviruses. These results could be fully explained as arising from the modest depletion of cellular membrane cholesterol that accompanied S1P disruption. Mechanistic studies of cells and with protein-free liposomes suggested that high levels of cholesterol are specifically needed for hantavirus membrane fusion. Taken together, our results indicate that the profound dependence on target membrane cholesterol is a fundamental, and unusual, biophysical property of hantavirus glycoprotein-membrane interactions during entry. IMPORTANCE Although hantaviruses cause important human diseases worldwide, no specific antiviral treatments are available. One of the major obstacles to the development of new therapies is a lack of understanding of how hantaviruses hijack our own host factors to enter cells. Here, we identified multiple cellular genes that control the levels of cholesterol in cellular membranes to be important for hantavirus entry. Our findings suggest that high concentrations of cholesterol in cellular membranes are required at a specific step in the entry process—fusion between viral and cellular membranes—that allows escape of the hantavirus genome into the host cell cytoplasm to initiate infection. Our findings uncover a fundamental feature of the hantavirus infection mechanism and point to cholesterol-lowering drugs as a potential new treatment of hantaviral infections.
- Published
- 2015
- Full Text
- View/download PDF
18. Rubella virus: first calcium-requiring viral fusion protein.
- Author
-
Mathieu Dubé, Felix A Rey, and Margaret Kielian
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Rubella virus (RuV) infection of pregnant women can cause fetal death, miscarriage, or severe fetal malformations, and remains a significant health problem in much of the underdeveloped world. RuV is a small enveloped RNA virus that infects target cells by receptor-mediated endocytosis and low pH-dependent membrane fusion. The structure of the RuV E1 fusion protein was recently solved in its postfusion conformation. RuV E1 is a member of the class II fusion proteins and is structurally related to the alphavirus and flavivirus fusion proteins. Unlike the other known class II fusion proteins, however, RuV E1 contains two fusion loops, with a metal ion complexed between them by the polar residues N88 and D136. Here we demonstrated that RuV infection specifically requires Ca(2+) during virus entry. Other tested cations did not substitute. Ca(2+) was not required for virus binding to cell surface receptors, endocytic uptake, or formation of the low pH-dependent E1 homotrimer. However, Ca(2+) was required for low pH-triggered E1 liposome insertion, virus fusion and infection. Alanine substitution of N88 or D136 was lethal. While the mutant viruses were efficiently assembled and endocytosed by host cells, E1-membrane insertion and fusion were specifically blocked. Together our data indicate that RuV E1 is the first example of a Ca(2+)-dependent viral fusion protein and has a unique membrane interaction mechanism.
- Published
- 2014
- Full Text
- View/download PDF
19. A mechanistic paradigm for broad-spectrum antivirals that target virus-cell fusion.
- Author
-
Frederic Vigant, Jihye Lee, Axel Hollmann, Lukas B Tanner, Zeynep Akyol Ataman, Tatyana Yun, Guanghou Shui, Hector C Aguilar, Dong Zhang, David Meriwether, Gleyder Roman-Sosa, Lindsey R Robinson, Terry L Juelich, Hubert Buczkowski, Sunwen Chou, Miguel A R B Castanho, Mike C Wolf, Jennifer K Smith, Ashley Banyard, Margaret Kielian, Srinivasa Reddy, Markus R Wenk, Matthias Selke, Nuno C Santos, Alexander N Freiberg, Michael E Jung, and Benhur Lee
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
LJ001 is a lipophilic thiazolidine derivative that inhibits the entry of numerous enveloped viruses at non-cytotoxic concentrations (IC50 ≤ 0.5 µM), and was posited to exploit the physiological difference between static viral membranes and biogenic cellular membranes. We now report on the molecular mechanism that results in LJ001's specific inhibition of virus-cell fusion. The antiviral activity of LJ001 was light-dependent, required the presence of molecular oxygen, and was reversed by singlet oxygen ((1)O2) quenchers, qualifying LJ001 as a type II photosensitizer. Unsaturated phospholipids were the main target modified by LJ001-generated (1)O2. Hydroxylated fatty acid species were detected in model and viral membranes treated with LJ001, but not its inactive molecular analog, LJ025. (1)O2-mediated allylic hydroxylation of unsaturated phospholipids leads to a trans-isomerization of the double bond and concurrent formation of a hydroxyl group in the middle of the hydrophobic lipid bilayer. LJ001-induced (1)O2-mediated lipid oxidation negatively impacts on the biophysical properties of viral membranes (membrane curvature and fluidity) critical for productive virus-cell membrane fusion. LJ001 did not mediate any apparent damage on biogenic cellular membranes, likely due to multiple endogenous cytoprotection mechanisms against phospholipid hydroperoxides. Based on our understanding of LJ001's mechanism of action, we designed a new class of membrane-intercalating photosensitizers to overcome LJ001's limitations for use as an in vivo antiviral agent. Structure activity relationship (SAR) studies led to a novel class of compounds (oxazolidine-2,4-dithiones) with (1) 100-fold improved in vitro potency (IC50
- Published
- 2013
- Full Text
- View/download PDF
20. Genome-wide RNAi screen identifies novel host proteins required for alphavirus entry.
- Author
-
Yaw Shin Ooi, Katie M Stiles, Catherine Y Liu, Gwen M Taylor, and Margaret Kielian
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
The enveloped alphaviruses include important and emerging human pathogens such as Chikungunya virus and Eastern equine encephalitis virus. Alphaviruses enter cells by clathrin-mediated endocytosis, and exit by budding from the plasma membrane. While there has been considerable progress in defining the structure and function of the viral proteins, relatively little is known about the host factors involved in alphavirus infection. We used a genome-wide siRNA screen to identify host factors that promote or inhibit alphavirus infection in human cells. Fuzzy homologue (FUZ), a protein with reported roles in planar cell polarity and cilia biogenesis, was required for the clathrin-dependent internalization of both alphaviruses and the classical endocytic ligand transferrin. The tetraspanin membrane protein TSPAN9 was critical for the efficient fusion of low pH-triggered virus with the endosome membrane. FUZ and TSPAN9 were broadly required for infection by the alphaviruses Sindbis virus, Semliki Forest virus, and Chikungunya virus, but were not required by the structurally-related flavivirus Dengue virus. Our results highlight the unanticipated functions of FUZ and TSPAN9 in distinct steps of alphavirus entry and suggest novel host proteins that may serve as targets for antiviral therapy.
- Published
- 2013
- Full Text
- View/download PDF
21. In vitro and in vivo studies identify important features of dengue virus pr-E protein interactions.
- Author
-
Aihua Zheng, Mahadevaiah Umashankar, and Margaret Kielian
- Subjects
Immunologic diseases. Allergy ,RC581-607 ,Biology (General) ,QH301-705.5 - Abstract
Flaviviruses bud into the endoplasmic reticulum and are transported through the secretory pathway, where the mildly acidic environment triggers particle rearrangement and allows furin processing of the prM protein to pr and M. The peripheral pr peptide remains bound to virus at low pH and inhibits virus-membrane interaction. Upon exocytosis, the release of pr at neutral pH completes virus maturation to an infectious particle. Together this evidence suggests that pr may shield the flavivirus fusion protein E from the low pH environment of the exocytic pathway. Here we developed an in vitro system to reconstitute the interaction of dengue virus (DENV) pr with soluble truncated E proteins. At low pH recombinant pr bound to both monomeric and dimeric forms of E and blocked their membrane insertion. Exogenous pr interacted with mature infectious DENV and specifically inhibited virus fusion and infection. Alanine substitution of E H244, a highly conserved histidine residue in the pr-E interface, blocked pr-E interaction and reduced release of DENV virus-like particles. Folding, membrane insertion and trimerization of the H244A mutant E protein were preserved, and particle release could be partially rescued by neutralization of the low pH of the secretory pathway. Thus, pr acts to silence flavivirus fusion activity during virus secretion, and this function can be separated from the chaperone activity of prM. The sequence conservation of key residues involved in the flavivirus pr-E interaction suggests that this protein-protein interface may be a useful target for broad-spectrum inhibitors.
- Published
- 2010
- Full Text
- View/download PDF
22. prM-reactive antibodies reveal a role for partially mature virions in dengue virus pathogenesis
- Author
-
Kimberly A. Dowd, Devika Sirohi, Scott D. Speer, Laura A. VanBlargan, Rita E. Chen, Swati Mukherjee, Bradley M. Whitener, Jennifer Govero, Maya Aleshnick, Bridget Larman, Soila Sukupolvi-Petty, Madhumati Sevvana, Andrew S. Miller, Thomas Klose, Aihua Zheng, Scott Koenig, Margaret Kielian, Richard J. Kuhn, Michael S. Diamond, and Theodore C. Pierson
- Subjects
Multidisciplinary - Abstract
Cleavage of the flavivirus premembrane (prM) structural protein during maturation can be inefficient. The contribution of partially mature flavivirus virions that retain uncleaved prM to pathogenesis during primary infection is unknown. To investigate this question, we characterized the functional properties of newly-generated dengue virus (DENV) prM-reactive monoclonal antibodies (mAbs) in vitro and using a mouse model of DENV disease. Anti-prM mAbs neutralized DENV infection in a virion maturation state–dependent manner. Alanine scanning mutagenesis and cryoelectron microscopy of anti-prM mAbs in complex with immature DENV defined two modes of attachment to a single antigenic site. In vivo, passive transfer of intact anti-prM mAbs resulted in an antibody-dependent enhancement of disease. However, protection against DENV-induced lethality was observed when the transferred mAbs were genetically modified to inhibit their ability to interact with Fcγ receptors. These data establish that in addition to mature forms of the virus, partially mature infectious prM + virions can also contribute to pathogenesis during primary DENV infections.
- Published
- 2023
23. Identification of human and mosquito receptors for alphaviruses
- Author
-
Caroline K. Martin and Margaret Kielian
- Subjects
Multidisciplinary - Published
- 2022
24. Near-germline human monoclonal antibodies neutralize and protect against multiple arthritogenic alphaviruses
- Author
-
Michael S. Diamond, M. Javad Aman, James T. Earnest, Rohit K. Jangra, Jonathan R. Lai, Frederick W. Holtsberg, Ryan J. Malonis, Kartik Chandran, Margaret Kielian, Arthur S. Kim, Matthew Angeliadis, and Johanna P. Daily
- Subjects
Multidisciplinary ,biology ,medicine.drug_class ,viruses ,virus diseases ,Alphavirus ,biology.organism_classification ,medicine.disease ,Monoclonal antibody ,medicine.disease_cause ,Virology ,Epitope ,Virus ,Immunity ,biology.protein ,medicine ,Chikungunya ,Antibody ,Alphavirus infection - Abstract
Arthritogenic alphaviruses are globally distributed, mosquito-transmitted viruses that cause rheumatological disease in humans and include Chikungunya virus (CHIKV), Mayaro virus (MAYV), and others. Although serological evidence suggests that some antibody-mediated heterologous immunity may be afforded by alphavirus infection, the extent to which broadly neutralizing antibodies that protect against multiple arthritogenic alphaviruses are elicited during natural infection remains unknown. Here, we describe the isolation and characterization of MAYV-reactive alphavirus monoclonal antibodies (mAbs) from a CHIKV-convalescent donor. We characterized 33 human mAbs that cross-reacted with CHIKV and MAYV and engaged multiple epitopes on the E1 and E2 glycoproteins. We identified five mAbs that target distinct regions of the B domain of E2 and potently neutralize multiple alphaviruses with differential breadth of inhibition. These broadly neutralizing mAbs (bNAbs) contain few somatic mutations and inferred germline-revertants retained neutralizing capacity. Two bNAbs, DC2.M16 and DC2.M357, protected against both CHIKV- and MAYV-induced musculoskeletal disease in mice. These findings enhance our understanding of the cross-reactive and cross-protective antibody response to human alphavirus infections.
- Published
- 2021
25. Third Tofo Advanced Study Week on Emerging and Re-emerging Viruses, 2018
- Author
-
Rachel Fearns, Rolf Hilgenfeld, Yi Shi, Julien Lescar, Eduardo Samo Gudo, Susan Daniel, Margaret Kielian, Felicity J. Burt, Albrecht von Brunn, Athanase Badolo, and Susan R. Weiss
- Subjects
0301 basic medicine ,Rift Valley fever virus ,medicine.drug_class ,viruses ,030106 microbiology ,medicine.disease_cause ,Communicable Diseases, Emerging ,Article ,Disease Outbreaks ,Dengue fever ,03 medical and health sciences ,Virology ,parasitic diseases ,medicine ,Animals ,Humans ,RNA Viruses ,Chikungunya ,Socioeconomics ,Mozambique ,Pharmacology ,biology ,Flavivirus ,virus diseases ,Outbreak ,Zika Virus ,Congresses as Topic ,Hemorrhagic Fever, Ebola ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Lassa virus ,Geography ,Hemorrhagic Fever Virus, Crimean-Congo ,Chikungunya Fever ,Hemorrhagic Fever, Crimean ,Antiviral drug ,Crimean Congo hemorrhagic fever virus - Abstract
The Third Tofo Advanced Study Week on Emerging and Re-Emerging Viruses (3rd TASW) was held in Praia do Tofo, Mozambique, from September 02 to 06, 2018. It brought together 55 participants from 10 African countries as well as from Belgium, China, Germany, Singapore, and the USA. Meeting sessions covered aspects of the epidemiology, diagnosis, molecular and structural biology, vaccine development, and antiviral drug discovery for emerging RNA viruses that are current threats in Africa and included flaviviruses (dengue and Zika), alphaviruses (chikungunya), coronaviruses, filoviruses (Ebola), influenza viruses, Crimean Congo hemorrhagic fever virus, Rift Valley fever Virus, Lassa virus, and others. Data were presented on recent flavivirus and/or chikungunyavirus outbreaks in Angola, Burkina Faso, and Mozambique. In addition, these viruses are endemic in many sub-Saharan countries. The TASW series on emerging viruses is unique in Africa and successful in promoting collaborations between researchers in Africa and other parts of the world, as well as among African scientists. This report summarizes the lectures held at the meeting and highlights advances in the field., Highlights • The 3rd Tofo Advanced Study Week on Emerging and Re-emerging Viruses took place from September 2–6, 2018. • African attendees came from Angola, Botswana, Burkina Faso, the CAR, Mozambique, Nigeria, S Africa, Tanzania and Zimbabwe. • Other participants were from Europe, China, Singapore, and the USA. • This unique meeting enabled scientists from Africa and elsewhere to discuss problems and initiate new collaborations. • Presentations covered dengue virus, Zika, chikungunya, coronaviruses, Ebola, influenza, Rift Valley fever, CCHF, and RSV.
- Published
- 2019
26. Molecular and Structural Insights into the Life Cycle of Rubella Virus
- Author
-
Pratyush Kumar Das and Margaret Kielian
- Subjects
Comparative genomics ,Congenital rubella syndrome ,Immunology ,Human pathogen ,Rubella virus ,Biology ,medicine.disease_cause ,Pathogenicity ,medicine.disease ,Microbiology ,Virology ,Viral entry ,Insect Science ,Vaccination coverage ,medicine ,Protein homology ,Minireview - Abstract
Rubella virus (RUBV), a rubivirus, is an airborne human pathogen that generally causes mild measles-like symptoms in children or adults. However, RUBV infection of pregnant women can result in miscarriage or congenital rubella syndrome (CRS), a collection of long-term birth defects, including incomplete organ development and mental retardation. Worldwide vaccination campaigns have significantly reduced the number of RUBV infections, but RUBV continues to be a problem in countries with low vaccination coverage. Furthermore, the recent discovery of pathogenic rubiviruses in other mammals emphasizes the spillover potential of rubella-related viruses to humans. In the last decade, our understanding of RUBV has been significantly increased by virological, biochemical, and structural studies, providing a platform to begin understanding the life cycle of RUBV at the molecular level. This review concentrates on recent work on RUBV, focusing on the virion; its structural components; and its entry, fusion, and assembly mechanisms. Important features of RUBV are compared with those of viruses from other families. We also use comparative genomics, manual curation, and protein homology modeling to highlight distinct features of RUBV that are evolutionarily conserved in the nonhuman rubiviruses. Since rubella-like viruses may potentially have higher pathogenicity and transmissibility to humans, we also propose a framework for utilizing RUBV as a model to study its more pathogenic cousins.
- Published
- 2021
27. The Enigmatic Capsid Protein of an Encephalitic Rubivirus
- Author
-
Pratyush Kumar Das and Margaret Kielian
- Subjects
0303 health sciences ,030306 microbiology ,Transmission (medicine) ,Immunology ,Rubella virus ,Biology ,medicine.disease_cause ,medicine.disease ,Microbiology ,Virology ,Rubella ,03 medical and health sciences ,Capsid ,Genus ,Insect Science ,Viral evolution ,medicine ,Letter to the Editor ,030304 developmental biology - Abstract
The genus Rubivirus was previously comprised of a single member, Rubella virus (RuV), which is spread by airborne or maternal-fetal transmission and only infects humans (1).….
- Published
- 2021
28. Advances in Virus Research
- Author
-
Margaret Kielian, Marilyn J. Roossinck, Margaret Kielian, and Marilyn J. Roossinck
- Subjects
- Virology--Research
- Abstract
Advances in Virus Research, Volume 115, the latest release in this comprehensive serial that highlights new advances in the field, includes updates on a variety of timely topics, including Plant viral nanotools, Mycoviruses, Rift Valley Fever virus entry and infection, and more. - Provides the authority and expertise of leading contributors from an international board of authors - Presents the latest release in the Advances in Virus Research series
- Published
- 2023
29. Insertion of Dengue E into lipid bilayers studied by neutron reflectivity and molecular dynamics simulations
- Author
-
Sushil K. Satija, Sadie La Bauve, Aihua Zheng, Juan M. Vanegas, Bryan Carson, Susan B. Rempe, David M. Rogers, Briana C. Vernon, Bulent Akgun, Michael S. Kent, Frank Heinrich, and Margaret Kielian
- Subjects
Models, Molecular ,0301 basic medicine ,Lipid Bilayers ,Biophysics ,Virus Attachment ,Trimer ,Molecular Dynamics Simulation ,Spodoptera ,Membrane Fusion ,Biochemistry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Molecular dynamics ,Viral Envelope Proteins ,Animals ,Amino Acid Sequence ,Lipid bilayer ,POPC ,Cells, Cultured ,Neutrons ,030102 biochemistry & molecular biology ,Hydrogen bond ,technology, industry, and agriculture ,Lipid bilayer fusion ,Hydrogen Bonding ,Cell Biology ,Virus Internalization ,030104 developmental biology ,Membrane ,chemistry ,lipids (amino acids, peptides, and proteins) ,Neutron reflectometry ,Protein Binding - Abstract
The envelope (E) protein of Dengue virus rearranges to a trimeric hairpin to mediate fusion of the viral and target membranes, which is essential for infectivity. Insertion of E into the target membrane serves to anchor E and possibly also to disrupt local order within the membrane. Both aspects are likely to be affected by the depth of insertion, orientation of the trimer with respect to the membrane normal, and the interactions that form between trimer and membrane. In the present work, we resolved the depth of insertion, the tilt angle, and the fundamental interactions for the soluble portion of Dengue E trimers (sE) associated with planar lipid bilayer membranes of various combinations of l-palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-palmitoyl-2-oleoyl-sn-glycero-3-phospho-rac-glycerol (POPG), l-palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), and cholesterol (CHOL) by neutron reflectivity (NR) and by molecular dynamics (MD) simulations. The results show that the tip of E containing the fusion loop (FL) is located at the interface of the headgroups and acyl chains of the outer leaflet of the lipid bilayers, in good agreement with prior predictions. The results also indicate that E tilts with respect to the membrane normal upon insertion, promoted by either the anionic lipid POPG or CHOL. The simulations show that tilting of the protein correlates with hydrogen bond formation between lysines and arginines located on the sides of the trimer close to the tip (K246, K247, and R73) and nearby lipid headgroups. These hydrogen bonds provide a major contribution to the membrane anchoring and may help to destabilize the target membrane.
- Published
- 2018
30. Advances in Virus Research
- Author
-
Thomas Mettenleiter, Margaret Kielian, Marilyn J. Roossinck, Thomas Mettenleiter, Margaret Kielian, and Marilyn J. Roossinck
- Abstract
Advances in Virus Research, Volume 111, the latest release in a serial that highlights new advances in the field, presents interesting and timely chapters authored by an international board of subject matter experts. - Provides the authority and expertise of leading contributors from an international board of authors - Presents the latest release in the Advances in Virus Research series
- Published
- 2021
31. Enhancing host cell infection by SARS-CoV-2
- Author
-
Margaret Kielian
- Subjects
2019-20 coronavirus outbreak ,animal structures ,Coronavirus disease 2019 (COVID-19) ,viruses ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Pneumonia, Viral ,Severe Acute Respiratory Syndrome ,Betacoronavirus ,Pandemic ,Neuropilin 1 ,medicine ,Humans ,skin and connective tissue diseases ,Pandemics ,Multidisciplinary ,biology ,SARS-CoV-2 ,COVID-19 ,respiratory system ,biology.organism_classification ,medicine.disease ,Virology ,Neuropilin-1 ,Pneumonia ,embryonic structures ,Coronavirus Infections - Abstract
Neuropilin-1 binds the furin-processed spike protein of SARS-CoV-2 to promote virus entry
- Published
- 2020
32. Multiple capsid protein binding sites mediate selective packaging of the alphavirus genomic RNA
- Author
-
Markus Hafner, Dimitrios G. Anastasakis, Rebecca S. H. Brown, and Margaret Kielian
- Subjects
0301 basic medicine ,Models, Molecular ,Science ,viruses ,Alphaviruses ,General Physics and Astronomy ,Viral transmission ,Alphavirus ,Semliki Forest virus ,Virus Replication ,Virus ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Capsid ,Chlorocebus aethiops ,Animals ,Guide RNA ,Binding site ,lcsh:Science ,Nucleocapsid ,Vero Cells ,Multidisciplinary ,Binding Sites ,030102 biochemistry & molecular biology ,biology ,Chemistry ,Virus Assembly ,Viral nucleocapsid ,RNA ,virus diseases ,General Chemistry ,Genomics ,biochemical phenomena, metabolism, and nutrition ,biology.organism_classification ,Semliki forest virus ,Cell biology ,030104 developmental biology ,Viral replication ,Biotinylation ,RNA, Viral ,lcsh:Q ,Capsid Proteins ,Chikungunya virus ,Protein Binding - Abstract
The alphavirus capsid protein (Cp) selectively packages genomic RNA (gRNA) into the viral nucleocapsid to produce infectious virus. Using photoactivatable ribonucleoside crosslinking and an innovative biotinylated Cp retrieval method, here we comprehensively define binding sites for Semliki Forest virus (SFV) Cp on the gRNA. While data in infected cells demonstrate Cp binding to the proposed genome packaging signal (PS), mutagenesis experiments show that PS is not required for production of infectious SFV or Chikungunya virus. Instead, we identify multiple Cp binding sites that are enriched on gRNA-specific regions and promote infectious SFV production and gRNA packaging. Comparisons of binding sites in cytoplasmic vs. viral nucleocapsids demonstrate that budding causes discrete changes in Cp-gRNA interactions. Notably, Cp’s top binding site is maintained throughout virus assembly, and specifically binds and assembles with Cp into core-like particles in vitro. Together our data suggest a model for selective alphavirus genome recognition and assembly., Alphaviruses need to selectively package genomic viral RNA for transmission, but the packaging mechanism remains unclear. Here, Brown et al. combine PAR-CLIP with biotinylated capsid protein (Cp) retrieval and identify multiple Cp binding sites on genomic viral RNA that promote virion formation.
- Published
- 2019
33. Human monoclonal antibodies against chikungunya virus target multiple distinct epitopes in the E1 and E2 glycoproteins
- Author
-
Ryan J. Malonis, Jesper Pallesen, Daniel Hofmann, Elisabeth K. Nyakatura, Andrew B. Ward, Johanna P. Daily, Larissa B. Thackray, Jonathan R. Lai, M. Javad Aman, Courtney A. Cohen, John M. Dye, Rohit K. Jangra, Margaret Kielian, Kartik Chandran, Rebecca S. H. Brown, Vinayak Rayannavar, Michael S. Diamond, Jose A. Quiroz, Frederick W. Holtsberg, Lorellin A. Durnell, and Sergey Shulenin
- Subjects
RNA viruses ,Viral Diseases ,Physiology ,medicine.disease_cause ,Antibodies, Viral ,Pathology and Laboratory Medicine ,Biochemistry ,Epitope ,Epitopes ,Mice ,Viral Envelope Proteins ,Immune Physiology ,Medicine and Health Sciences ,Chikungunya ,Biology (General) ,Enzyme-Linked Immunoassays ,chemistry.chemical_classification ,0303 health sciences ,Mice, Inbred ICR ,Chikungunya Virus ,Immune System Proteins ,biology ,030302 biochemistry & molecular biology ,Microbial Mutation ,virus diseases ,Antibodies, Monoclonal ,3. Good health ,Precipitation Techniques ,Infectious Diseases ,Medical Microbiology ,Viral Pathogens ,Viruses ,Antibody ,Pathogens ,Research Article ,Neglected Tropical Diseases ,Adult ,Viral Entry ,QH301-705.5 ,medicine.drug_class ,Alphaviruses ,Immunology ,Alphavirus ,Viral Structure ,Monoclonal antibody ,Research and Analysis Methods ,Microbiology ,Virus ,Antibodies ,Togaviruses ,03 medical and health sciences ,Viral entry ,Virology ,Genetics ,medicine ,Animals ,Humans ,Immunoprecipitation ,Immunoassays ,Molecular Biology ,Microbial Pathogens ,030304 developmental biology ,Glycoproteins ,Biology and life sciences ,Organisms ,Chikungunya Infection ,Proteins ,RC581-607 ,biology.organism_classification ,Tropical Diseases ,Antibodies, Neutralizing ,Mice, Inbred C57BL ,chemistry ,biology.protein ,Immunologic Techniques ,Chikungunya Fever ,Parasitology ,Immunologic diseases. Allergy ,Glycoprotein ,Viral Transmission and Infection - Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes persistent arthritis in a subset of human patients. We report the isolation and functional characterization of monoclonal antibodies (mAbs) from two patients infected with CHIKV in the Dominican Republic. Single B cell sorting yielded a panel of 46 human mAbs of diverse germline lineages that targeted epitopes within the E1 or E2 glycoproteins. MAbs that recognized either E1 or E2 proteins exhibited neutralizing activity. Viral escape mutations localized the binding epitopes for two E1 mAbs to sites within domain I or the linker between domains I and III; and for two E2 mAbs between the β-connector region and the B-domain. Two of the E2-specific mAbs conferred protection in vivo in a stringent lethal challenge mouse model of CHIKV infection, whereas the E1 mAbs did not. These results provide insight into human antibody response to CHIKV and identify candidate mAbs for therapeutic intervention., Author summary Chikungunya virus (CHIKV) is a globally emerging virus that can cause significant disease, including a prolonged and painful arthritis. The virus is spread by mosquitoes that circulate in many regions of the world including the United States. Currently, there are no available vaccines or therapies to treat CHIKV infection. In this report, we identified and characterized a large panel of antibodies against CHIKV from two donors that contracted the viral infection in the Dominican Republic. These antibodies target a number of different regions of the membrane proteins that coat the surface of the virus, and many can inhibit the ability of CHIKV to infect cells. Two of the antibodies were shown to protect mice from a lethal dose of CHIKV. These antibodies have therapeutic potential, and provide insight into the human immune response that may facilitate vaccine development.
- Published
- 2019
34. Surface (S) Layer Proteins of Lactobacillus acidophilus Block Virus Infection via DC-SIGN Interaction
- Author
-
Maria Guadalupe Martinez, Bernd Lepenies, Sandra M. Ruzal, Margaret Kielian, Eileen M. Geoghegan, and Mariano Prado Acosta
- Subjects
Microbiology (medical) ,S-LAYER ,viruses ,lcsh:QR1-502 ,Alphavirus ,Dengue virus ,Semliki Forest virus ,medicine.disease_cause ,Microbiology ,DC-SIGN ,Virus ,lcsh:Microbiology ,S-layer ,purl.org/becyt/ford/1 [https] ,03 medical and health sciences ,LACTOBACILLUS ,flavivirus ,medicine ,alphavirus ,Chikungunya ,purl.org/becyt/ford/1.6 [https] ,030304 developmental biology ,Original Research ,Infectivity ,0303 health sciences ,biology ,030306 microbiology ,virus diseases ,biology.organism_classification ,FLAVIVIRUS ,Virology ,3. Good health ,Flavivirus ,Lactobacillus ,ALPHAVIRUS ,biology.protein - Abstract
Alphaviruses and flaviviruses are important human pathogens that include Chikungunya virus (CHIKV), Dengue virus (DENV), and Zika virus (ZIKV), which can cause diseases in humans ranging from arthralgia to hemorrhagic fevers and microcephaly. It was previously shown that treatment with surface layer (S-layer) protein, present on the bacterial cell-envelope of Lactobacillus acidophilus, is able to inhibit viral and bacterial infections by blocking the pathogen’s interaction with DC-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN), a trans-membrane protein that is a C-type calcium-dependent lectin. DC-SIGN is known to act as an attachment factor for several viruses including alphaviruses and flaviviruses. In the present study, we used alphaviruses as a model system to dissect the mechanism of S-layer inhibition. We first evaluated the protective effect of S-layer using 3T3 cells, either wild type or stably expressing DC-SIGN, and infecting with the alphaviruses Semliki Forest virus (SFV) and CHIKV and the flaviviruses ZIKV and DENV. DC-SIGN expression significantly enhanced infection by all four viruses. Treatment of the cells with S-layer prior to infection decreased infectivity of all viruses only in cells expressing DC-SIGN. In vitro ELISA experiments showed a direct interaction between S-layer and DC-SIGN; however, confocal microscopy and flow cytometry demonstrated that S-layer binding to the cells was independent of DC-SIGN expression. S-layer protein prevented SFV binding and internalization in DC-SIGN-expressing cells but had no effect on virus binding to DC-SIGN-negative cells. Inhibition of virus binding occurred in a time-dependent manner, with a significant reduction of infection requiring at least a 30-min pre-incubation of S-layer with DC-SIGN-expressing cells. These results suggest that S-layer has a different mechanism of action compared to mannan, a common DC-SIGN-binding compound that has an immediate effect in blocking viral infection. This difference could reflect slower kinetics of S-layer binding to the DC-SIGN present at the plasma membrane (PM). Alternatively, the S-layer/DC-SIGN interaction may trigger the activation of signaling pathways that are required for the inhibition of viral infection. Together our results add important information relevant to the potential use of L. acidophilus S-layer protein as an antiviral therapy. Fil: Prado Acosta, Mariano. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina. Albert Einstein College of Medicine; Estados Unidos Fil: Geoghegan, Eileen M.. Albert Einstein College of Medicine; Estados Unidos Fil: Lepenies, Bernd. University of Veterinary Medicine Hannover; Alemania Fil: Ruzal, Sandra Mónica. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Ciudad Universitaria. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales. Universidad de Buenos Aires. Facultad de Ciencias Exactas y Naturales. Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales; Argentina Fil: Kielian, Margaret. Albert Einstein College of Medicine; Estados Unidos Fil: Martinez, Maria Guadalupe. Albert Einstein College of Medicine; Estados Unidos
- Published
- 2019
35. Mechanism of Tetherin Inhibition of Alphavirus Release
- Author
-
Yaw Shin Ooi, Judy J. Wan, and Margaret Kielian
- Subjects
viruses ,media_common.quotation_subject ,Immunology ,Endocytic cycle ,Alphavirus ,Biology ,Endocytosis ,Microbiology ,Cell Line ,Viral envelope ,Cricetinae ,Virology ,Animals ,Humans ,Protein Isoforms ,Internalization ,Virus Release ,media_common ,Host factor ,Alphavirus Infections ,Bone Marrow Stromal Antigen 2 ,HEK 293 cells ,NF-kappa B ,Virion ,biology.organism_classification ,Virus-Cell Interactions ,Cell biology ,HEK293 Cells ,Insect Science ,Tetherin - Abstract
Tetherin is an interferon-inducible, antiviral host factor that broadly restricts enveloped virus release by tethering budded viral particles to the plasma membrane. In response, many viruses have evolved tetherin antagonists. The human tetherin gene can express two isoforms, long and short, due to alternative translation initiation sites in the N-terminal cytoplasmic tail. The long isoform (L-tetherin) contains 12 extra amino acids in its N terminus, including a dual tyrosine motif (YDYCRV) that is an internalization signal for clathrin-mediated endocytosis and a determinant of NF-κB activation. Tetherin restricts alphaviruses, which are highly organized enveloped RNA viruses that bud from the plasma membrane. L-tetherin is more efficient than S-tetherin in inhibiting alphavirus release in 293 cells. Here, we demonstrated that alphaviruses do not encode an antagonist for either of the tetherin isoforms. Instead, the isoform specificity reflected a requirement for tetherin endocytosis. The YXY motif in L-tetherin was necessary for alphavirus restriction in 293 cells but was not required for rhabdovirus restriction. L-tetherin’s inhibition of alphavirus release correlated with its internalization but did not involve NF-κB activation. In contrast, in U-2 OS cells, the YXY motif and the L-tetherin N-terminal domain were not required for either robust tetherin internalization or alphavirus inhibition. Tetherin forms that were negative for restriction accumulated at the surface of infected cells, while the levels of tetherin forms that restrict were decreased. Together, our results suggest that tetherin-mediated virus internalization plays an important role in the restriction of alphavirus release and that cell-type-specific cofactors may promote tetherin endocytosis. IMPORTANCE The mechanisms of tetherin’s antiviral activities and viral tetherin antagonism have been studied in detail for a number of different viruses. Although viral countermeasures against tetherin can differ significantly, overall, tetherin’s antiviral activity correlates with physical tethering of virus particles to prevent their release. While tetherin can mediate virus endocytic uptake and clearance, this has not been observed to be required for restriction. Here we show that efficient tetherin inhibition of alphavirus release requires efficient tetherin endocytosis. Our data suggest that this endocytic uptake can be mediated by tetherin itself or by a tetherin cofactor that promotes uptake of an endocytosis-deficient variant of tetherin.
- Published
- 2019
36. BHK-21 Cell Clones Differ in Chikungunya Virus Infection and MXRA8 Receptor Expression
- Author
-
Margaret Kielian and Peiqi Yin
- Subjects
0301 basic medicine ,Sindbis virus ,receptor ,viruses ,Receptor expression ,030106 microbiology ,Cell ,BHK-21 cells ,Gene Expression ,Alphavirus ,medicine.disease_cause ,Microbiology ,Article ,Virus ,Cell Line ,03 medical and health sciences ,Cricetinae ,Virology ,medicine ,Animals ,Humans ,alphavirus ,Chikungunya ,chikungunya virus ,MXRA8 ,biology ,urogenital system ,Membrane Proteins ,virus diseases ,Transfection ,biology.organism_classification ,QR1-502 ,030104 developmental biology ,Infectious Diseases ,medicine.anatomical_structure ,Cell culture ,Host-Pathogen Interactions ,Chikungunya Fever - Abstract
Baby hamster kidney-21 (BHK-21) cells are widely used to propagate and study many animal viruses using infection and transfection techniques. Among various BHK-21 cell clones, the fibroblast-like BHK-21/C-13 line and the epithelial-like BHK-21/WI-2 line are commonly used cell clones for alphavirus research. Here we report that BHK-21/WI-2 cells were significantly less susceptible to primary infection by the alphavirus chikungunya virus (CHIKV) than were BHK-21/C-13 cells. The electroporation efficiency of alphavirus RNA into BHK-21/WI-2 was also lower than that of BHK-21/C-13. The growth of CHIKV was decreased in BHK-21/WI-2 compared to BHK-21/C-13, while primary infection and growth of the alphavirus Sindbis virus (SINV) were equivalent in the two cell lines. Our results suggested that CHIKV entry could be compromised in BHK-21/WI-2. Indeed, we found that the mRNA level of the CHIKV receptor MXRA8 in BHK-21/WI-2 cells was much lower than that in BHK-21/C-13 cells, and exogenous expression of either human MXRA8 or hamster MXRA8 rescued CHIKV infection. Our results affirm the importance of the MXRA8 receptor for CHIKV infection, and document differences in its expression in two clonal cell lines derived from the original BHK-21 cell cultures. Our results also indicate that CHIKV propagation and entry studies in BHK-21 cells will be significantly more efficient in BHK-21/C-13 than in BHK-21/WI-2 cells.
- Published
- 2021
37. Virus Assembly and Exit Pathways
- Author
-
Margaret Kielian, Thomas Mettenleiter, Marilyn J. Roossinck, Margaret Kielian, Thomas Mettenleiter, and Marilyn J. Roossinck
- Subjects
- Viruses, Virology
- Abstract
Advances in Virus Research, Volume 108, in this ongoing series, highlights new advances in the field, with this new volume presenting interesting chapters on topics including Virus infections of the developing brain, Geminivirus assembly, Flavivirus assembly, Cell-cell transmission, Archael virus assembly, Potyvirus assembly, Poxvirus assembly and exit, Mycovirus assembly, Reo/orbivirus assembly and exit, Giant virus assembly, Quasi-enveloped virus assembly/exit, and Betaherpesvirus assembly and exit. - Provides the authority and expertise of leading contributors from an international board of authors - Presents the latest release in the Advances in Virus Research series - Includes the latest information on virus assembly and exit pathways
- Published
- 2020
38. Advances in Virus Research
- Author
-
Thomas Mettenleiter, Margaret Kielian, Marilyn J. Roossinck, Thomas Mettenleiter, Margaret Kielian, and Marilyn J. Roossinck
- Subjects
- Virology, Viruses
- Abstract
Advances in Virus Research, Volume 103, the latest in the Advances in Virus Research series, contains new, informative updates on the topic. First published in 1953, this series covers a diverse range of in-depth reviews, providing a valuable overview of the current field of virology. Updates to this release include a Review on phage genes that affect bacterial phenotypes, Phage lysis: new perspectives on the bacterial cell envelope, CRISPRs as the adaptive immune system of archaea and bacteria, CRESS viruses, Tupanviruses, and Giant virus evolution, amongst other topics. Contains contributions from leading authorities in the field of virology Informs and updates on all the latest developments in the field
- Published
- 2019
39. Virus Entry
- Author
-
Thomas Mettenleiter, Margaret Kielian, Marilyn J. Roossinck, Thomas Mettenleiter, Margaret Kielian, and Marilyn J. Roossinck
- Abstract
Virus Entry, Volume 104, the latest release in the Advances in Virus Research series, highlights new advances in the field, with this new volume presenting interesting chapters on plant virus cell-to-cell entry, plant virus entry via insect transmission, VSV/Rabies virus entry, Papovavirus entry, New approaches to study fusion proteins, Hantavirus receptors, Gamma Herpesvirus entry, and many other interesting topics. - Provides the authority and expertise of leading contributors from an international board of authors - Presents the latest release in the Advances in Virus Research series - Includes the latest information on virus structure and function
- Published
- 2019
40. Calcium-Dependent Rubella Virus Fusion Occurs in Early Endosomes
- Author
-
Margaret Kielian, Maximilian Fels, Loïc Etienne, and Mathieu Dubé
- Subjects
0301 basic medicine ,Protein Conformation ,Endosome ,Immunology ,chemistry.chemical_element ,Endosomes ,Calcium ,Biology ,Membrane Fusion ,Microbiology ,Calcium in biology ,03 medical and health sciences ,Viral entry ,Virology ,Chlorocebus aethiops ,Animals ,Vero Cells ,Rubella ,030102 biochemistry & molecular biology ,Membrane fusion protein ,Virus Assembly ,Cell Membrane ,Lipid bilayer fusion ,Hydrogen-Ion Concentration ,Virus Internalization ,Fusion protein ,Virus-Cell Interactions ,Cell biology ,030104 developmental biology ,Membrane protein ,chemistry ,Insect Science ,Liposomes ,Mutation ,Rubella virus ,Viral Fusion Proteins - Abstract
The E1 membrane protein of rubella virus (RuV) is a class II membrane fusion protein structurally related to the fusion proteins of the alphaviruses, flaviviruses, and phleboviruses. Virus entry is mediated by a low pH-dependent fusion reaction through E1's insertion into the cell membrane and refolding to a stable homotrimer. Unlike the other described class II proteins, RuV E1 contains 2 fusion loops, which complex a metal ion between them by interactions with residues N88 and D136. Insertion of the E1 protein into the target membrane, fusion, and infection require calcium and are blocked by alanine substitution of N88 or D136. Here we addressed the requirements of E1 for calcium binding and the intracellular location of the calcium requirement during virus entry. Our results demonstrated that N88 and D136 are optimally configured to support RuV fusion and are strongly selected for during the virus life cycle. While E1 has some similarities with cellular proteins that bind calcium and anionic lipids, RuV binding to the membrane was independent of anionic lipids. Virus fusion occurred within early endosomes, and chelation of intracellular calcium showed that calcium within the early endosome was required for virus fusion and infection. Calcium triggered the reversible insertion of E1 into the target membrane at neutral pH, but E1 homotrimer formation and fusion required a low pH. Thus, RuV E1, unlike other known class II fusion proteins, has distinct triggers for membrane insertion and fusion protein refolding mediated, respectively, by endosomal calcium and low pH. IMPORTANCE Rubella virus causes a mild disease of childhood, but infection of pregnant women frequently results in miscarriage or severe birth defects. In spite of an effective vaccine, RuV disease remains a serious problem in many developing countries. RuV infection of host cells involves endocytic uptake and low pH-triggered membrane fusion and is unusual in its requirement for calcium binding by the membrane fusion protein. Here we addressed the mechanism of the calcium requirement and the required location of calcium during virus entry. Both calcium and low pH were essential during the virus fusion reaction, which was shown to occur in the early endosome compartment.
- Published
- 2016
41. The Alphavirus E2 Membrane-Proximal Domain Impacts Capsid Interaction and Glycoprotein Lattice Formation
- Author
-
Emily A. Byrd and Margaret Kielian
- Subjects
viruses ,Immunology ,Alphavirus ,Microbiology ,Virus ,Cell Line ,Capsid ,Viral envelope ,Viral Envelope Proteins ,Virology ,Animals ,Humans ,Amino Acid Sequence ,Nucleocapsid ,Virus Release ,Glycoproteins ,chemistry.chemical_classification ,Membranes ,biology ,Alphavirus Infections ,Virus Assembly ,Structure and Assembly ,Cell Membrane ,RNA ,biology.organism_classification ,Cell biology ,Ectodomain ,chemistry ,Insect Science ,Host cell plasma membrane ,Capsid Proteins ,Sindbis Virus ,Glycoprotein - Abstract
Alphaviruses are small enveloped RNA viruses that bud from the host cell plasma membrane. Alphavirus particles have a highly organized structure, with a nucleocapsid core containing the RNA genome surrounded by the capsid protein, and a viral envelope containing 80 spikes, each a trimer of heterodimers of the E1 and E2 glycoproteins. The capsid protein and envelope proteins are both arranged in organized lattices that are linked via the interaction of the E2 cytoplasmic tail/endodomain with the capsid protein. We previously characterized the role of two highly conserved histidine residues, H348 and H352, located in an external, juxtamembrane region of the E2 protein termed the D-loop. Alanine substitutions of H348 and H352 inhibit virus growth by impairing late steps in the assembly/budding of virus particles at the plasma membrane. To investigate this budding defect, we selected for revertants of the E2-H348/352A double mutant. We identified eleven second-site revertants with improved virus growth and mutations in the capsid, E2 and E1 proteins. Multiple isolates contained the mutation E2-T402K in the E2 endodomain or E1-T317I in the E1 ectodomain. Both of these mutations were shown to partially restore H348/352A growth and virus assembly/budding, while neither rescued the decreased thermostability of H348/352A. Within the alphavirus particle, these mutations are positioned to affect the E2-capsid interaction or the E1-mediated intertrimer interactions at the 5-fold axis of symmetry. Together, our results support a model in which the E2 D-loop promotes the formation of the glycoprotein lattice and its interactions with the internal capsid protein lattice. IMPORTANCE Alphaviruses include important human pathogens such as Chikungunya and the encephalitic alphaviruses. There are currently no licensed alphavirus vaccines or effective antiviral therapies, and more molecular information on virus particle structure and function is needed. Here, we highlight the important role of the E2 juxtamembrane D-loop in mediating virus budding and particle production. Our results demonstrated that this E2 region affects both the formation of the external glycoprotein lattice and its interactions with the internal capsid protein shell.
- Published
- 2018
42. Preface to Volume 100: History and Looking Forward
- Author
-
Margaret Kielian, Thomas C. Mettenleiter, and Marilyn J. Roossinck
- Subjects
0301 basic medicine ,03 medical and health sciences ,Virology ,MEDLINE ,Historical Article ,Biology ,History, 20th Century ,Periodicals as Topic ,030112 virology ,History, 21st Century ,Classics ,Volume (compression) - Published
- 2018
43. Mechanisms of Virus Membrane Fusion Proteins
- Author
-
Margaret Kielian
- Subjects
biology ,Cell ,Lipid bilayer fusion ,Alphavirus ,biology.organism_classification ,Virology ,Fusion protein ,Virus ,Cell biology ,medicine.anatomical_structure ,Viral envelope ,Viral entry ,medicine ,Intracellular - Abstract
Enveloped viruses infect host cells by a membrane fusion reaction that takes place at the cell surface or in intracellular compartments following virus uptake. Fusion is mediated by the membrane interactions and conformational changes of specialized virus envelope proteins termed membrane fusion proteins. This article discusses the structures and refolding reactions of specific fusion proteins and the methods for their study and highlights outstanding questions in the field.
- Published
- 2014
44. Imaging the Alphavirus Exit Pathway
- Author
-
Geoffrey S. Perumal, Maria Guadalupe Martinez, Erik L. Snapp, Frank P. Macaluso, and Margaret Kielian
- Subjects
Sindbis virus ,viruses ,Immunology ,Alphavirus ,Microbiology ,Virus ,Cell Line ,VP40 ,Viral envelope ,Virology ,Animals ,Humans ,Viral shedding ,Instrumentation ,Virus Release ,Budding ,biology ,Alphavirus Infections ,Chemistry ,Virus Assembly ,Cell Membrane ,Viral membrane ,biology.organism_classification ,Transmembrane protein ,Virus-Cell Interactions ,Cell biology ,Capsid ,Insect Science ,Sindbis Virus ,Fluorescence Recovery After Photobleaching - Abstract
Alphaviruses are small enveloped RNA viruses with highly organized structures that exclude host cell proteins. They contain an internal nucleocapsid and an external lattice of the viral E2 and E1 transmembrane proteins. Alphaviruses bud from the plasma membrane (PM), but the process and dynamics of alphavirus assembly and budding are poorly understood. Here we generated Sindbis viruses (SINVs) with fluorescent protein labels on the E2 envelope protein and exploited them to characterize virus assembly and budding in living cells. During virus infection, E2 became enriched in localized patches on the PM and in filopodium-like extensions. These E2-labeled patches and extensions contained all of the viral structural proteins. Correlative light and electron microscopy studies established that the patches and extensions colocalized with virus budding structures, while light microscopy showed that they excluded a freely diffusing PM marker protein. Exclusion required the interaction of the E2 protein with the capsid protein, a critical step in virus budding, and was associated with the immobilization of the envelope proteins on the cell surface. Virus infection induced two distinct types of extensions: tubulin-negative extensions that were ∼2 to 4 μm in length and excluded the PM marker, and tubulin-positive extensions that were >10 μm long, contained the PM marker, and could transfer virus particles to noninfected cells. Tubulin-positive extensions were selectively reduced in cells infected with a nonbudding SINV mutant. Together, our data support a model in which alphavirus infection induces reorganization of the PM and cytoskeleton, leading to virus budding from specialized sites. IMPORTANCE Alphaviruses are important and widely distributed human pathogens for which vaccines and antiviral therapies are urgently needed. These small highly organized viruses bud from the host cell PM. Virus assembly and budding are critical but little understood steps in the alphavirus life cycle. We developed alphaviruses with fluorescent protein tags on one of the viral membrane (envelope) proteins and used a variety of microscopy techniques to follow the envelope protein and a host cell PM protein during budding. We showed that alphavirus infection induced the formation of patches and extensions on the PM where the envelope proteins accumulate. These sites excluded other PM proteins and correlated with virus budding structures. Exclusion of PM proteins required specific interactions of the viral envelope proteins with the internal capsid protein. Together, our data indicate that alphaviruses extensively reorganize the cell surface and cytoskeleton to promote their assembly and budding.
- Published
- 2014
45. A Key Interaction between the Alphavirus Envelope Proteins Responsible for Initial Dimer Dissociation during Fusion
- Author
-
Whitney Fields and Margaret Kielian
- Subjects
Dimer ,Immunology ,Fluorescent Antibody Technique ,CHO Cells ,Alphavirus ,Semliki Forest virus ,Microbiology ,Dissociation (chemistry) ,Endosome membrane ,chemistry.chemical_compound ,Cricetulus ,Viral Envelope Proteins ,Viral entry ,Cricetinae ,Virology ,Animals ,Furin ,Membrane Glycoproteins ,biology ,Hydrogen Bonding ,Hydrogen-Ion Concentration ,Virus Internalization ,biology.organism_classification ,Semliki forest virus ,Fusion protein ,Virus-Cell Interactions ,Biochemistry ,chemistry ,Mutagenesis ,Insect Science ,Biophysics ,biology.protein ,Dimerization - Abstract
Alphaviruses such as Semliki Forest virus (SFV) are enveloped viruses whose surface is covered by an organized lattice composed of trimers of E2-E1 heterodimers. The E1 envelope protein, a class II fusion protein, contains the hydrophobic fusion loop and refolds to drive virus fusion with the endosome membrane. The E2 protein is synthesized as a precursor p62, whose processing by furin primes the heterodimer for dissociation during virus entry. Dissociation of the E2-E1 heterodimer is an essential step during low-pH-triggered fusion, while the dissociation of the immature p62-E1 dimer is relatively pH resistant. Previous structural studies described an “acid-sensitive region” in E2 that becomes disordered at low pH. Within this region, the conserved E2 H170 is in position to form a hydrogen bond with the underlying E1 S57. Here we experimentally tested the role of this interaction in regulating dimer dissociation in mature and immature virus. Alanine substitutions of E1 S57 and E2 H170 destabilized the heterodimer and produced a higher pH threshold for exposure of the E1 fusion loop and for fusion of the immature virus. E1 S57K or S57D mutations were lethal and caused transport and assembly defects that were partially abrogated by neutralization of the exocytic pathway. The lethal phenotype of E1 S57K was rescued by second-site mutations at E2 H170/M171. Together, our results define a key role for the E1 S57-E2 H170 interaction in dimer stability and the pH dependence of fusion and provide evidence for stepwise dissociation of the E2-E1 dimer at low pH.
- Published
- 2013
46. Flaviviruses: braking the entering
- Author
-
Margaret Kielian and Theodore C. Pierson
- Subjects
Models, Molecular ,biology ,Mechanism (biology) ,Extramural ,Flavivirus ,viruses ,Lipid bilayer fusion ,Virus Internalization ,biology.organism_classification ,Antiviral Agents ,Models, Biological ,Virology ,Article ,Virus ,Cell biology ,Viral Envelope Proteins ,Viral entry ,Animals ,Humans - Abstract
Flaviviruses are small spherical virus particles covered by a dense icosahedral array of envelope (E) proteins that mediate virus attachment to cells and the fusion of viral and cellular membranes. Our understanding of the mechanism by which flavivirus E proteins orchestrate entry into cells has been advanced by studies of E structure and arrangement on the virion at different steps of the virus entry/membrane fusion process. When combined with an increasingly clear (albeit still incomplete) view of the cell biology of virus entry, these advances suggest new antiviral strategies. Indeed, inhibitors that target cellular and viral processes involved in entry show promise as powerful tools to study this critical step of the viral lifecycle, and with luck, may ultimately lead to therapeutic advances.
- Published
- 2013
47. Method for measuring the unbinding energy of strongly-bound membrane-associated proteins
- Author
-
Bryan Carson, Michael S. Kent, Dongmei Ye, Briana C. Vernon, Susan B. Rempe, Cathryn M. Siegrist, David M. Rogers, Andrew P. Shreve, Margaret Kielian, Aihua Zheng, and Elisa La Bauve
- Subjects
0301 basic medicine ,Binding energy ,Enthalpy ,Lipid Bilayers ,Biophysics ,Activation energy ,Biochemistry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,Viral Envelope Proteins ,Animals ,Lipid bilayer ,POPC ,Cells, Cultured ,Unilamellar Liposomes ,030102 biochemistry & molecular biology ,Peripheral membrane protein ,Phosphatidylglycerols ,Cell Biology ,Dengue Virus ,Hydrogen-Ion Concentration ,Crystallography ,Kinetics ,030104 developmental biology ,Monomer ,Membrane ,Drosophila melanogaster ,chemistry ,Phosphatidylcholines ,Thermodynamics ,Protein Binding - Abstract
We describe a new method to measure the activation energy for unbinding (enthalpy ΔH*u and free energy ΔG*u) of a strongly-bound membrane-associated protein from a lipid membrane. It is based on measuring the rate of release of a liposome-bound protein during centrifugation on a sucrose gradient as a function of time and temperature. The method is used to determine ΔH*u and ΔG*u for the soluble dengue virus envelope protein (sE) strongly bound to 80:20 POPC:POPG liposomes at pH 5.5. ΔH*u is determined from the Arrhenius equation whereas ΔG*u is determined by fitting the data to a model based on mean first passage time for escape from a potential well. The binding free energy ΔGb of sE was also measured at the same pH for the initial, predominantly reversible, phase of binding to a 70:30 PC:PG lipid bilayer. The unbinding free energy (20 +/− 3 kcal/mol, 20% PG) was found to be roughly three times the binding energy per monomer, (7.8 +/− 0.3 kcal/mol for 30% PG, or est. 7.0 kcal/mol for 20% PG). This is consistent with data showing that free sE is a monomer at pH 5.5, but assembles into trimers after associating with membranes. This new method to determine unbinding energies should be useful to understand better the complex interactions of integral monotopic proteins and strongly-bound peripheral membrane proteins with lipid membranes.
- Published
- 2016
48. Role of TSPAN9 in Alphavirus Entry and Early Endosomes
- Author
-
Katie M. Stiles and Margaret Kielian
- Subjects
0301 basic medicine ,Sindbis virus ,Endosome ,Tetraspanins ,viruses ,Immunology ,Endocytic cycle ,Intracellular Space ,Vesicular Transport Proteins ,Gene Expression ,Alphavirus ,Endosomes ,Semliki Forest virus ,Microbiology ,Cell Line ,03 medical and health sciences ,0302 clinical medicine ,Virology ,Humans ,Late endosome ,biology ,Alphavirus Infections ,virus diseases ,biochemical phenomena, metabolism, and nutrition ,Hydrogen-Ion Concentration ,Virus Internalization ,biology.organism_classification ,Semliki forest virus ,Endocytosis ,Virus-Cell Interactions ,Protein Transport ,030104 developmental biology ,Cholesterol ,Vesicular stomatitis virus ,030220 oncology & carcinogenesis ,Insect Science ,Early endosome membrane ,Host-Pathogen Interactions ,Peptide Hydrolases - Abstract
Alphaviruses are small enveloped RNA viruses that infect cells via clathrin-mediated endocytosis and low-pH-triggered fusion in the early endosome. Using a small interfering RNA (siRNA) screen in human cells, we previously identified TSPAN9 as a host factor that promotes infection by the alphaviruses Sindbis virus (SINV), Semliki Forest virus (SFV), and chikungunya virus (CHIKV). Depletion of TSPAN9 specifically decreases SFV membrane fusion in endosomes. TSPAN9 is a member of the tetraspanin family of multipass membrane proteins, but its cellular function is currently unknown. Here we used U-2 OS cells stably overexpressing TSPAN9 to show that TSPAN9 is localized at the plasma membrane and in early and late endosomes. Internalized SFV particles colocalized with TSPAN9 in vesicles early during infection. Depletion of TSPAN9 led to reductions in the amounts of the late endosomal proteins LAMP1 and CD63 and an increase in the amount of LAMP2. However, TSPAN9 depletion did not alter the delivery of SFV to early endosomes or change their pH or protease activity. Comparative studies showed that TSPAN9 depletion strongly inhibited infection by several viruses that fuse in early endosomes (SFV, SINV, CHIKV, and vesicular stomatitis virus [VSV]), while viruses that fuse in the late endosome (recombinant VSV-Lassa and VSV-Junin), including an SFV point mutant with a lower pH threshold for fusion (SFV E2 T12I), were relatively resistant. Our data suggest that TSPAN9 modulates the early endosome compartment to make it more permissive for membrane fusion of early-penetrating viruses. IMPORTANCE Alphaviruses are spread by mosquitoes and can cause serious human diseases such as arthritis and encephalitis. Recent outbreaks of CHIKV infection are responsible for millions of cases of acute illness and long-term complications. There are no vaccines or antiviral treatments for these important human pathogens. Alphaviruses infect host cells by utilizing the endocytic machinery of the cell and fusing their membrane with that of the endosome. Although the mechanism of virus-membrane fusion is well studied, we still know relatively little about the host cell proteins that are involved in alphavirus entry. Here we characterized the role of the host membrane protein TSPAN9 in alphavirus infection. TSPAN9 was localized to early endosomes containing internalized alphavirus, and depletion of TSPAN9 inhibited virus fusion with the early endosome membrane. In contrast, infection of viruses that enter through the late endosome was relatively resistant to TSPAN9 depletion, suggesting an important role for TSPAN9 in the early endosome.
- Published
- 2016
49. The Interaction of Alphavirus E1 Protein with Exogenous Domain III Defines Stages in Virus-Membrane Fusion
- Author
-
Margaret Kielian and Gleyder Roman-Sosa
- Subjects
Immunology ,Immunoglobulins ,Trimer ,Alphavirus ,Kidney ,Virus Replication ,Semliki Forest virus ,Microbiology ,Cell membrane ,Viral Envelope Proteins ,Viral envelope ,Cricetinae ,Virology ,medicine ,Animals ,Immunoprecipitation ,Cells, Cultured ,biology ,Alphavirus Infections ,Cell Membrane ,Lipid bilayer fusion ,Virus Internalization ,biology.organism_classification ,Fusion protein ,Molecular biology ,Recombinant Proteins ,Transmembrane protein ,Virus-Cell Interactions ,Protein Structure, Tertiary ,medicine.anatomical_structure ,Insect Science ,Liposomes ,Biophysics ,Protein Multimerization - Abstract
Alphaviruses such as Semliki Forest virus (SFV) are enveloped viruses that infect cells through a low-pH-triggered membrane fusion reaction mediated by the transmembrane fusion protein E1. E1 drives fusion by insertion of its hydrophobic fusion loop into the cell membrane and refolding to a stable trimeric hairpin. In this postfusion conformation, the immunoglobulin-like domain III (DIII) and the stem region pack against the central core of the trimer. Membrane fusion and infection can be specifically inhibited by exogenous DIII, which binds to an intermediate in the E1 refolding pathway. Here we characterized the properties of the E1 target for interaction with exogenous DIII. The earliest target for DIII binding was an extended membrane-inserted E1 trimer, which was not detectable by assays for the stable postfusion hairpin. DIII binding provided a tool to detect this extended trimer and to define a series of SFV fusion-block mutants. DIII binding studies showed that the mutants were blocked in distinct steps in fusion protein refolding. Our results suggested that formation of the initial extended trimer was reversible and that it was stabilized by the progressive fold-back of the DIII and stem regions.
- Published
- 2011
50. In Vitro Reconstitution Reveals Key Intermediate States of Trimer Formation by the Dengue Virus Membrane Fusion Protein
- Author
-
Margaret Kielian, Claudia Sánchez-San Martín, Aihua Zheng, and Maofu Liao
- Subjects
Protein Folding ,Immunology ,Trimer ,Biology ,Microbiology ,Cell membrane ,Viral Envelope Proteins ,Viral envelope ,Virology ,medicine ,Membrane fusion protein ,Cell Membrane ,Lipid bilayer fusion ,Dengue Virus ,Hydrogen-Ion Concentration ,Virus Internalization ,Fusion protein ,Molecular biology ,Transmembrane protein ,Virus-Cell Interactions ,medicine.anatomical_structure ,Insect Science ,Biophysics ,Protein folding ,Protein Multimerization ,Viral Fusion Proteins - Abstract
The flavivirus dengue virus (DV) infects cells through a low-pH-triggered membrane fusion reaction mediated by the viral envelope protein E. E is an elongated transmembrane protein with three domains and is organized as a homodimer on the mature virus particle. During fusion, the E protein homodimer dissociates, inserts the hydrophobic fusion loop into target membranes, and refolds into a trimeric hairpin in which domain III (DIII) packs against the central trimer. It is clear that E refolding drives membrane fusion, but the steps in hairpin formation and their pH requirements are unclear. Here, we have used truncated forms of the DV E protein to reconstitute trimerization in vitro . Protein constructs containing domains I and II (DI/II) were monomeric and interacted with membranes to form core trimers. DI/II-membrane interaction and trimerization occurred efficiently at both neutral and low pH. The DI/II core trimer was relatively unstable and could be stabilized by binding exogenous DIII or by the formation of mixed trimers containing DI/II plus E protein with all three domains. The mixed trimer had unoccupied DIII interaction sites that could specifically bind exogenous DIII at either low or neutral pH. Truncated DV E proteins thus reconstitute hairpin formation and define properties of key domain interactions during DV fusion.
- Published
- 2010
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.