-->

Sunday, December 1, 2019

Antiviral Actions of Tetherin Against Ebola Virus Microvesicles and Immunomodulation


          Ebola virus infection is associated with fatal diseases in humans. For example, Ebola hemorrhagic fever is distinguished by the pathological onset of severe inflammation and internal bleeding within infected hosts [2]. Despite a high prevalence of human-human transmission, Ebola is a “zoonotic” virus, meaning it enters the human population via an animal host. For this particular virus, the animal host is a fruit bat, as certain species have been shown to act as reservoirs— carrying an asymptomatic infection able to manifest as pathogenic when transmitted into humans [3]. Until 2014, Ebola was endemic, or regularly found within the population, in West Africa. However, when a mutant strain of the virus emerged, an epidemic was established in the region. This outbreak lasted until 2016 and during this period the virus found its way to eleven individuals within the U.S., which resulted in two fatalities of American citizens [4]. Today, the Democratic Republic of the Congo is currently facing an outbreak of its own, keeping research on the virus and potential ways to control its spread at the forefront of global public health efforts [5].
In terms of structure, the Ebola virus genome is composed of single-stranded, minus-sense RNA that must be converted to positive-sense by its own polymerase before translation can occur. Additionally, its lipid membrane envelope includes just one type of protein, known as a glycoprotein (GP), on its surface. This protein protrudes from Ebola viral particles, playing a key role in the attachment and fusion of the particles to host target cells. In its mature form, GP exists as two subunits: GP1, which includes a receptor binding domain for attaching to target cell surfaces, and GP2, which includes a fusion peptide and transmembrane domain to facilitate this attachment during infection. In the face of such viral infection, host immunity often works in opposition to the invading virus, creating an arms race as each one attempts to overcome the other. For instance, the host restriction factor, tetherin, is an antiviral gene regulated by the innate immune system, or the host’s nonspecific defense that kicks in directly following detection of a virus. Known to function in the suppression of many enveloped viruses, tetherin works to interfere with a virus’s ability to release progeny viral particles from infected cells in order to spread within the host. Specifically, as seen below, tetherin’s unique double-anchored structure inserts itself into both the viral envelope and infected cell membranes in order to anchor budding virions to the cell and thereby prevent free release into the host. 

Tetherin: Traps viral-associated vesicles, as pictured, or progeny virions at the cell surface by embedding one anchor into the releasing membrane and the other into the host cell plasma membrane. [1]

 
           In this study, Nehls et al. were mainly focused on tetherin’s targeting of vesicle release, in addition to trapping budding virions. Previous studies these researchers built upon have reported that, like many viruses, Ebola virus infection releases microvesicles upon infection as a mechanism to emit viral proteins or nucleic acids from infected cells. Specifically, Ebola utilizes these vesicles to carry out the release of its GP protein. To better elucidate the role of these “GP-virosomes” in Ebola’s ability to infect hosts, researchers analyzed these particles and hypothesized that tetherin interacts with the GP-associated vesicles, ultimately interfering with their release. 
            To specifically analyze GP-virosomes, as opposed to exosomes and smaller vesicles, researchers first used a centrifugation technique that separated the seemingly larger virosomes from other smaller vesicles thought to be lacking the GP viral protein. To verify that these virosomes were both vesicles of larger size and carriers of the Ebola virus GP, a western blot, as shown below, was performed showcasing enriched GP in microvesicles (pelleted at 21000xg) as compared to in exosomes and smaller vesicles (pelleted at 100000xg). 

Figure 1C: Verifying Ebola virosomes as GP-containing, non-exosomal vesicles. Supernatants of transfected 293T cells were spun down at different centrifugation speeds and GP levels were detected via western blot. Microvesicles (100-1000nm in size) pelleted at 21000xg and exosomes (<100nm) pelleted at 100000xg.

            After observing via electron microscopy that the release of these GP-virosomes from host cells is similar to the budding of progeny virions, researchers aimed to investigate tetherin’s impact on this release. First, a human embryonic kidney cell line (HEK-293T) commonly used for transfections, or the introduction of DNA to eukaryotic cells, was used to analyze the coexpression of tetherin and GP in cell culture. To track and quantify the amount of GP released by virosomes, researchers performed a western blot on the cell culture media, as this is where the virosomes are expected to be located after successful release from infected cells. However, if tetherin was expressed as well, as shown below, this release of Ebola virus GP-virosomes detected within the media was significantly reduced. This result was replicated in HeLa cells, as this cell line contains endogenous levels of tetherin, and therefore verifies that the suppression of release seen is not an unintended impact of tetherin overexpression in the transfected 293T cells. 
           
Figure 4a: Monitoring the release of GP-virosomes. 293T cells were transfected to express varying amounts of tetherin alone (mock) or in addition to cotransfection with GP. As tetherin expression increased, GP-virosome detection in media decreased in GP-transfected cells. 

            Next, researchers analyzed tetherin mutants to narrow down which components of the protein’s unique anchor shape are vital for its ability to suppress GP-virosome release. Tetherin mutants lacking the C-terminal anchor were discovered via western blot to be incorporated into the cell culture media. This result suggested that the C-terminal anchor facilitates tetherin insertion into the host cell plasma membrane and is therefore vital for the protein’s ability to prohibit virosome release. Without this region, tetherin is not only unable to keep GP-virosomes from entering the cell culture media, but is also incorporated into the virosomes themselves. After narrowing down the vitality of tetherin’s shape to its antiviral function, transfections with tetherin derived from various animals, from hamsters to alligators, were compared to human tetherin functioning. Differential levels of GP detected in the media based on tetherin variation supported a conclusion that the primary sequence of tetherin, as it varies by species, is also important for the efficiency of the protein’s antiviral activity. Turning their sights to regions of the Ebola virus GP important for virosome release, researchers then analyzed the impact of various mutations in either the receptor binding or transmembrane domains of the protein. After transfecting HeLa cells with a GP variant containing a change in its transmembrane domain, tetherin levels were detected via a proximity ligation assay that uses antibodies against tetherin to fluorescently image the protein, as shown in the figure below. Levels of tetherin detected in the GP variant cells (ELE) were significantly reduced as compared to cells with wild-type GP transmembrane domains. This finding implied that this region of the Ebola virus GP is essential to tetherin’s ability to interact and prevent the release of GP-virosomes. 

Figure 5c: The GP transmembrane domain is important for tetherin binding. HeLa cells were transfected with GP or a GP variant with changes to its transmembrane domain (ELE). Cells were imaged via proximity ligation assay (PLA). Tetherin levels are shown in red, with decreased tetherin detection seen in GP-variant cells. 

            Lastly, Nehls et al. aimed to determine how these virosomes contribute to Ebola virus pathogenic abilities, as well as their possible role in viral-mediated immunomodulation within hosts. Interestingly, when a GP mutant strain from the 2014-2016 Ebola outbreak in West Africa was transfected into HeLa cells, the release of GP-virosomes in both the presence and absence of tetherin was significantly amplified as compared to Ebola virus strains with wild-type GP. As demonstrated by the outbreak, this mutant, known as A82V, is highly pathogenic. Therefore, this result supports a correlation between GP-virosome release and viral pathogenicity. As interesting as this result is, the GP-virosome’s role in immunomodulation is perhaps the most fascinating finding of this paper. Using a neutralization assay, the ability of an antibody, KZ52, to neutralize GP was measured via luciferase reporter activity. In supernatants containing GP-virosomes, the neutralizing activity of the antibody was counteracted until the addition of tetherin, in which neutralization levels were restored. These results provide support for a mechanism of virosomes to trap these neutralizing antibodies, ultimately preventing their ability to restrict true viral particles that express GP on their surface. This idea of GP-virosomes as “decoys” for the immune response was further supported by an analysis of cytokine release from macrophages treated with supernatants in the presence and absence of GP. Of the eight cytokines in question, three were seen to have decreased secretion in the presence of GP-virosomes unrestricted by tetherin. This immunomodulatory function of GP-virosomes, and its antagonization by tetherin, is key to understanding the breadth of the tetherin antiviral protein’s contribution to host innate immune responses against Ebola infection. 
            Nehls et al. do not highlight any future directions for their study in particular, however, there are many remaining questions left to be investigated. For instance, GP-virosome release was found by the researchers to be significantly inhibited by tetherin derived from the fruit bat reservoir species. This leads to speculation that tetherin’s role in suppressing GP-virosome release is separate from its role in antagonizing budding viral particles. However, since fruit bat tetherin demonstrates more efficient antiviral activity than the human form, does the idea that these tetherin-suppression processes are separate still hold true in human hosts facing pathogenesis? With human tetherin’s lower antiviral activity, its actions on GP-virosomes could be impacted in the presence of budding virions that tetherin must also work to target. To investigate this, researchers could utilize the same methodology of transfecting with Ebola virus GP, and analyzing its release into cell culture media, but add in the condition of infected cells. If GP-virosome release is still inhibited by human tetherin at the same level as in uninfected cells, this would verify that, in the infected host, these antiviral mechanisms of tetherin are indeed independent of one another. 
            Additionally, in their initial characterization of Ebola virus GP-virosomes, Nehls et al. notice an upregulation of the CD81 marker protein and describe the microvesicles as “CD81-positive,” but do not expand upon this finding. As CD81 is known as an exosomal marker, what role does it have in these non-exosomal GP-virosomes? One possibility is that CD81 contributes to tetherin’s ability to recognize the vesicle, as the protein has been found to also play a role in suppressing exosomal release [6]. To determine if CD81 is important for tetherin to be able to target exosomal and non-exosomal particles alike, the expression of the marker protein could be attenuated using a knockout mechanism such as RNAi. If tetherin’s ability to suppress release in the absence of CD81 is impacted, these findings would suggest that the marker could be a common denominator between tetherin targets. 
            Lastly, with the support these findings provide toward tetherin’s strong role in the innate immune response, it is easy to wonder if tetherin could potentially act as an agent in antiviral therapy. Future investigations could thus consider ways to upregulate tetherin expression on the cell surface of sites such as dendritic and other immune cells that face Ebola in its early stages of replication [7, 8]. Such advancements of therapy against Ebola virus infection could make a difference in facing current outbreaks, as well as future threats to international public health. 

References
[1] Nehls, J., Businger, R., Hoffmann, M., Brinkmann, C., Fehrenbacher, B., Schaller, M., ... & Pöhlmann, S. (2019). Release of Immunomodulatory Ebola Virus Glycoprotein-Containing Microvesicles Is Suppressed by Tetherin in a Species-Specific Manner. Cell reports, 26(7), 1841-1853.

[2] “What is Ebola?” John Hopkins Medicine. Retrieved from https://www.hopkinsmedicine.org/ebola/about-the-ebola-virus.html

[3] Leroy, E. M., Kumulungui, B., Pourrut, X., Rouquet, P., Hassanin, A., Yaba, P., ... & Swanepoel, R. (2005). Fruit bats as reservoirs of Ebola virus. Nature, 438(7068), 575. 

[4] “2014-2016 Ebola Outbreak in West Africa.” (2018, March 8). Centers for Disease Control and Prevention. Retrieved from https://www.cdc.gov/vhf/ebola/history/2014-2016-outbreak/index.html#anchor_1515001427541

[5] Moss, K., Michaud, J., & Kates, J. (2019, September 27). “The Current Ebola Outbreak and the U.S. Role: An Explainer.” Kaiser Family Foundation. Retrieved from

[6] Edgar, J. R., Manna, P. T., Nishimura, S., Banting, G., & Robinson, M. S. (2016). Tetherin is an exosomal tether. Elife, 5, e17180.

[7] Sauter, D., Specht, A., & Kirchhoff, F. (2010). Tetherin: holding on and letting go. Cell, 141(3), 392-398.

[8] Servick, K.  (2014, August 13). “What does Ebola actually do?” Science. Retrieved from https://www.sciencemag.org/news/2014/08/what-does-ebola-actually-do

No comments:

Post a Comment