Source: VIRGINIA POLYTECHNIC INSTITUTE submitted to NRP
ELUCIDATING THE MECHANISMS OF PRRSV IMMUNE EVASION MEDIATED BY NSP1A AND NSP2
Sponsoring Institution
National Institute of Food and Agriculture
Project Status
COMPLETE
Funding Source
Reporting Frequency
Annual
Accession No.
1011110
Grant No.
2017-67011-26045
Cumulative Award Amt.
$95,000.00
Proposal No.
2016-04845
Multistate No.
(N/A)
Project Start Date
Jan 1, 2017
Project End Date
Jun 30, 2019
Grant Year
2017
Program Code
[A7101]- AFRI Predoctoral Fellowships
Recipient Organization
VIRGINIA POLYTECHNIC INSTITUTE
(N/A)
BLACKSBURG,VA 24061
Performing Department
Biomed Sci and Pathobiology
Non Technical Summary
Porcine reproductive and respiratory syndrome (PRRS) is one of the most economically devastating diseases affecting the global swine industry. In the United States alone, PRRS is estimated to cause more than $600 million in production losses each year. The etiological agent of the disease, PRRS virus (PRRSV), suppresses several facets of the host immune response and therefore thwarts efforts of current vaccine programs in controlling the disease. The central hypothesis to be explored in this proposal is the PRRSV NSP1a and NSP2 proteins suppress the antiviral immune response via mechanisms operating at both the translational and transcriptional levels, respectively. To address this hypothesis, I will (1) delineate the molecular basis for NSP2-mediated immune suppression at the transcriptional level using bioinformatics and rational mutagenesis and (2) identify NSP1a interacting proteins and the underlying mechanisms involved in suppression of cap-dependent translation using immunoprecipitation coupled with mass spectrometry (IP-MS). The knowledge generated from this proposal will contribute to the development of novel and safe vaccines against this devastating pathogen. Efficiently treating and eradicating PRRSV will significantly improve the economic stability and viability of the agricultural economy in the United States, a critical priority of the USDA Agricultural and Food Research Initiative. Completing this pre-doctoral fellowship project will also benefit my development into a responsible and independent scientist in the field of molecular virology by affording me the opportunity to manage all aspects of a research project.
Animal Health Component
0%
Research Effort Categories
Basic
100%
Applied
0%
Developmental
0%
Classification

Knowledge Area (KA)Subject of Investigation (SOI)Field of Science (FOS)Percent
31140301101100%
Knowledge Area
311 - Animal Diseases;

Subject Of Investigation
4030 - Viruses;

Field Of Science
1101 - Virology;
Goals / Objectives
Several facets of the host immune response are suppressed during PRRSV infection. The goal of this fellowship project is to therefore further delineate the molecular basis of immune modulation mediated by the PRRSV NSP1α and NSP2 proteins. The central hypothesis in this fellowship proposal is that the PRRSV NSP1α and NSP2 proteins suppress the antiviral immune response via mechanisms operating at both the translational and transcriptional levels, respectively. By looking at how viral proteins can manipulate multiple levels of gene expression, we can develop more holistic and effective approaches in vaccine development. To test this hypothesis, I will attempt to accomplish the following objectives:Objective 1: Define the molecular basis for NSP2-mediated immune suppression at the transcriptional level. The PRRSV NSP2 protein has been previously shown to modulate the antiviral response by inhibiting NF-κB translocation into the nucleus and subsequent transcription of proinflammatory genes. Several viruses have been shown to suppress NF-κB signaling pathways by interfering with β-TrCP. Therefore, I hypothesize that the PRRSV NSP2 protein interacts with β-TrCP via its PDL motif and this interaction leads to alterations in NF-κB signaling pathways to suppress the antiviral response at the level of transcription. To investigate this hypothesis, I will employ reverse genetics approaches to explore the role of the putative NSP2 PDL motif in NF-κB signaling.Objective 2: Identify NSP1α interacting proteins and the underlying mechanisms involved in suppression of cap-dependent translation. Recent studies have indicated that the PRRSV NSP1α protein is involved in suppressing the host's immune response during viral infection. Viruses commonly modulate the host translation machinery to suppress the antiviral response. My own preliminary data suggests that PRRSV NSP1α suppresses cap-dependent translation of luciferase reporter genes. Therefore, I hypothesize that PRRSV NSP1α protein globally suppresses cap-dependent translation of host proteins through modulating key components of the host translation machinery. To test this hypothesis, I will use co-immunoprecipitation coupled with mass spectrometry (IP-MS) to identify candidate NSP1α interacting proteins. Once candidate interacting proteins have been identified, I will determine how NSP1α modulates their function to affect host translation in infected cells.
Project Methods
Objective 1: Define the molecular basis of NSP2-mediated immune suppression at the transcriptional level. I will first examine the function of the PDL motif that is present in the PRRSV NSP2 protein using co-immunoprecipitation experiments. To accomplish this, NSP2 derived from wild type VR2385 virus strain will be overexpressed in MARC-145 cells, a monkey kidney cell line permissive for PRRSV replication. Cell lysates will then be precipitated with an anti-PRRSV NSP2 specific antibody, followed by immunoblot analysis with a β-TrCP specific antibody. Subsequently, I will explore the function of the specific amino acid residues within the PDL motif of PRRSV NSP2 to determine which amino acid residues are critical for interaction with β-TrCP. To accomplish this, I will use a reverse genetics approach and co-immunoprecipitation experiments. Briefly, point mutations at functionally relevant amino acid positions in the PDL motif will be generated using site-directed mutagenesis. Co-immunoprecipitation experiments will then be performed as described above, in which the capacity of PRRSV NSP2 PDL mutants to interact with β-TrCP will be assessed. Next, I will determine if the PDL motif is necessary to modulate NF-κB signaling. To accomplish this, I will perform luciferase reporter assays to measure the NF-κB activity in MARC-145 cells. Cells will be co-transfected with NSP2 wild-type or PDL mutants, NF-κB response element (NF-κB-RE) firefly luciferase and HSK-tk nano luciferase. Cells will then be stimulated with TNF-α, an inducer of NF-κB activation, and relative luciferase activity will be calculated by normalizing firefly to nano luciferase activity and compared to controls. Additionally, I will monitor NF-κB subcellular localization as readout of activation using immunoblot and indirect immunofluorescence assays (IFA). Finally, I will use reverse genetics approaches to recover viable virus with introducing point mutations in the PRRSV NSP2 PDL motif and characterize NF-κB activation as described above. The expression of several pro-inflammatory cytokines such as IL-1, IL-6 and TNF- α will also be examined during viral infection with the NSP2 PDL mutants at both the mRNA and protein levels using RT-qPCR and ELISA, respectively.Objective 2: Identify PRRSV NSP1a interacting proteins involved in the suppression of cap-dependent translation. In parallel with the work proposed above in Objective 1, I will also attempt to identify candidate proteins interacting with NSP1α using immunoprecipitation coupled with mass spectrometry (IP-MS). I will overexpress FLAG-tagged NSP1α derived from wild-type VR2385 virus strain as well as the mutant NSP1 α G90A in porcine kidney PK-15 cells. Cell lysates will be collected and NSP1α interacting proteins will be precipitated using a FLAG-specific antibody. Precipitated proteins will then be prepared and analyzed by IP-MS using commercial services. The proteins involved in the suppression of cap-dependent translation will be identified by comparison of IP-MS analysis of host proteins differentially precipitated by NSP1α wild-type and G90A mutant. I will then examine how PRRSV NSP1α alters the candidate host proteins' function. IFA and western blot analyses will be used to assess candidate protein stability, post-translational modification and subcellular localization during viral infection. A reverse genetics approach will then be used to rescue a viable virus containing the NSP1 α G90A mutation. The G90A mutation will be introduced into both wild-type and NSP2 PDL point mutants generated in Objective 1. The expression of several pro-inflammatory cytokines such as IL-1, IL-6 and TNF- α will then be examined during viral infection with these mutant viruses at both the mRNA and protein levels using RT-qPCR and ELISA, respectively.

Progress 01/01/17 to 06/30/19

Outputs
Target Audience:Veterinarians and research scientists are informed of the results from this project through scientific meeting presentations and through the publication of data and results in peer-reviewed journal articles. Changes/Problems:Since we could not identify a role for the PRRSV NSP2 putative PDL motif in modulating transcriptional/translational processes within host cells, or identify NSP1a interacting proteins, we decided to study the mechanistic roles of stress granules (SGs) during PRRSV infection. SGs have been shown to play significant roles in regulating immune responses, at both the transcriptional and translational levels to viral infection. As such, we were among the first to establish PRRSV induces SG formation at late times post infection, and that these complexes are associated with viral replication complexes (VRCs). Furthermore, we also determined that SGs are dispensible for viral replication, as the small interferring RNA (siRNA)-mediated knockdown of critical SG components (G3BP1 and G3BP2) did not affect the ability of the virus to replicate. Additional research also revealed that the PRRSV-induced SGs form in a PERK-dependent manner, suggesting that the virus was also able to induce additional cellular stress responses. This finding led us to investigate the role of the unfolded protein response (UPR), a key response to adverse conditions, such as overloaded ER resulting from viral infection. This line of research led us to establish an antiviral role for the UPR during PRRSV infection, as chemical induction of the UPR significantly suppressed PRRSV replication. Collectively, these results suggest that SGs may be involved in viral replication, but are not crucial to the virus life cycle. However, our studies clearly show that the UPR negatively regulates PRRSV infection. Ultimately, we were able to achieve our goal of further elucidating the molecular mechanisms of PRRSV replication. The studies supported by this fellowship significantly enhance our understanding of PRRSV replication and pathogenesis. What opportunities for training and professional development has the project provided?Nicholas Catanzaro was afforded the opportunity to complete Virginia Tech's Graduate School, "Preparing the Future Professoriate" teaching certificate in which he extensively learned about topics pertaining to higher education and contemporary pedagogy. This opportunity significantly enhanced his professional development such that one day he may achieve his goal of becoming a tenure track faculty member at a RO1 research institute. The fellowship has also led him to present his research findings at a major PRRSV conference, as well as publish his results in the journal, Virus Research. How have the results been disseminated to communities of interest?We have presented the results of this project at scientific meetings. A major manuscript reporting the results from this project have now been published in the journal, Virus Research. Another manuscript has also been submitted to the same journal and is currently under peer review. What do you plan to do during the next reporting period to accomplish the goals? Nothing Reported

Impacts
What was accomplished under these goals? The orginial specific objectives were (1) Define the molecular basis for NSP2-mediated immune suppression at the transcriptional level. (2) Identify NSP1a interacting proteins and the underlying mechanisms involved in suppression of cap-dependent translation. The data generated from preliminary experiments does not support our initial hypothesis forumulated in Objective 1 that NSP2 contains a PDL-motif to modulate NFkB signaling during PRRSV infection. Therefore, we pursued another line of investigation examining how PRRSV modulates the host immune response via manipulation of the host's RNA stress granule pathway. Stress granules are dynamic regulators of host transcription and translation and have been shown to be important for the regulation of host antiviral defense mechanisms. Through our work, we have clearly established induction of stress granules during PRRSV infection and their underlying molecular pathways. Although we have modified the original proposed approaches to acheive the ultimate goal, the ultimate goal of the project remained the same. Thus, we have successfully accomplished the ultimate goal of the project which was to determine additional molecular mechanisms of PRRSV replication and pathogenesis. Specifically, for the original objectives I & II (100% completed) we clearly established the induction of stress granules during PRRSV infection. As such, we delineated the molecular mechanisms regulating the SG response to PRRSV infection. Using confocal microscopy, we first demonstrated that infection with PRRSV strain VR2385 induces an accumulation of the SG markers G3BP1, G3BP2, TIAR, eIF3b, and USP10 as well as mRNAs into punctate structures in the cytoplasm of infected host cells. Subsequently, we demonstrated that the PRRSV-induced SGs were in close proximity to viral replication complexes (VRCs) and processing bodies (P-bodies), and that SG formation was coordinated with inhibition of host cellular translation. Treatment of infected cells with cycloheximide disrupted the PRRSV-induced SGs. Furthermore, impairment of SG assembly by the shRNA-mediated knockdown of G3BP1, G3BP2 and USP10 did not affect viral replication. Overall impact:Collectively, these results demonstrate that PRRSV infection induces formation of SGs associated with VRCs, which is coordinated with the suppression of host cell protein synthesis. This is the first study to extensively characterize the formation and underlying mechanism ofbona fideSGs during PRRSV infection. Our findings have important implications in understanding the mechanism of PRRSV-host interactions.

Publications

  • Type: Journal Articles Status: Published Year Published: 2019 Citation: Catanzaro N, Meng XJ. Porcine reproductive and respiratory syndrome virus (PRRSV)-induced stress granules are associated with viral replication complexes and suppression of host translation. Virus Research. 2019 Feb 28; 265:47-56.


Progress 01/01/17 to 12/31/17

Outputs
Target Audience:The information generated by the work described in this proposal is expected to advance the knowledge of other researchers in the fields of molecular virology and antiviral vaccine development, especially swine veterinarians and researchers. Changes/Problems:The data generated thus far does not support our initial hypothesis formulated in Objective 1 that NSP2 contains a PDL-motif to modulate NFkB signaling during PRRSV infection. Thus, we are pursuing another line of investigation in looking at how PRRSV modulates the host immune response via manipulation of the host's RNA stress granule pathway. Stress granules are dynamic regulators of host transcription and translation and have been shown to be important for the regulation of host antiviral defense mechanisms. Thus far, we have demonstrated that PRRSV induces a unique class of stress granules that are compositionally and functionally distinct from canonical stress granules. We will continue this line of investigation. What opportunities for training and professional development has the project provided?I have successfully completed the Future Professoriate Teaching Certificate offered through the Graduate School at Virginia Tech. Completion of the certificate involved taking 3, 3 credit courses including, Preparing the Future Professoriate, Contemporary Pedagogy and Citizen Scholar. Completion of the certificate and these courses guided my professional development and has helped me prepare for my goal of obtaining a job as a tenure track faculty member at a reasearch institute. How have the results been disseminated to communities of interest? Nothing Reported What do you plan to do during the next reporting period to accomplish the goals?The data generated thus far does not support our initial hypothesis that NSP2 contains a putative PDL-motif involved in modulating the transcription of antiviral cytokines through the NFkB signaling pathway. In light of these results, we have started to pursue other lines of investigation looking into other mechanisms regulating the transciption and translation of antiviral host factors during PRRSV infection. Specifically, we are examining the antiviral properties of RNA stress granules during PRRSV infection. Stress granules are dynamic regulators of host transcription and translation and have been shown to be important for the regulation of host antiviral defense mechanisms. Using various molecular biology techniques, such as immunofluorescence assays, western blotting and RNA interference assays, we will continue this investigation during the next reporting period to accomplish this goal of further understanding modulation of the host immune response during PRRSV infection.

Impacts
What was accomplished under these goals? Objective 1: Define the molecular basis for NSP2- mediated immune suppression at the transcriptional level. Reverse genetics approaches have been utilized to investigate the potential role of the NSP2-mediated immune suppression observed during PRRSV infection. Point mutations were generated in the putative PDL-motif speculated to be involved in the suppression of the host innate immune response, and the ability of the protein to suppress NF-KB signaling pathways was investigated using transient transfection and luciferase reporter assays in cell culture. No significant changes were observed between the wild-type and mutant proteins. Thus, we conclude the PRRSV NSP2 PDL-motif is not involved in mediating immune suppression. Objective 2: Identify NSP1a interacting proteins and the underlying mechanisms involved in suppression of cap-dependent translation. We are still investigating the role of NSP1a in suppression of cap-dependent translation and have nothing to report on this objective.

Publications