1
|
Mahajan S, Kumar R, Singh A, Dhaka P, Pareek A, Kumar P, Tomar S. Targeting the host protein G3BP1 for the discovery of novel antiviral inhibitors against Chikungunya virus. Virology 2025; 608:110551. [PMID: 40306108 DOI: 10.1016/j.virol.2025.110551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 04/15/2025] [Accepted: 04/17/2025] [Indexed: 05/02/2025]
Abstract
The molecular interactions between Chikungunya virus (CHIKV) non-structural protein 3 (nsP3) and the host GTPase Activating SH3 Domain Binding Protein 1 (G3BP1) are critical for CHIKV replication. The C-terminus hypervariable domain (HVD) of nsP3 protein binds to the nuclear transport factor 2 (NTF2)-like domain of G3BP1 through two tandem FGDF motifs, aiding in the disruption of stress granule (SG) formation. Given G3BP1's role in the antiviral response, it presents an attractive target for antiviral drug development. In this study, seven potential small molecules targeting the FGDF motif binding pocket of G3BP1 were identified using a structure-based virtual screening approach. The binding modes of these molecules were further investigated through molecular docking and simulations. Surface Plasmon Resonance (SPR) and Isothermal Titration Calorimetry (ITC) experiments confirmed their binding to purified G3BP1 with micromolar (μM) affinity. The antiviral efficacy of these molecules was assessed using in vitro cell culture-based assays, revealing that L-7, WIN, SB2, NAL, DHD, GSK, and FLU effectively inhibited CHIKV replication with EC50 values of 1.99, 0.40, 5.38, 1.52, 7.39, 3.66, and 0.61 μM, respectively. Additionally, CHIKV-infected cells treated with these compounds exhibited fewer virus-induced SGs compared to untreated controls without affecting SG formation under oxidative stress conditions. These findings indicate that identified inhibitors successfully block G3BP1-nsP3 interactions and suppress CHIKV replication. This is one of the first reports of small antiviral molecules targeting G3BP1, a host protein essential for stress granule formation in the antiviral cellular response and CHIKV replication.
Collapse
Affiliation(s)
- Supreeti Mahajan
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Ravi Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Akshay Pareek
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India, 247667.
| |
Collapse
|
2
|
de Souza WM, Lecuit M, Weaver SC. Chikungunya virus and other emerging arthritogenic alphaviruses. Nat Rev Microbiol 2025:10.1038/s41579-025-01177-8. [PMID: 40335675 DOI: 10.1038/s41579-025-01177-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 05/09/2025]
Abstract
Arthritogenic alphaviruses are arboviruses (arthropod-borne viruses) that are genetically and serologically related positive-strand RNA viruses and cause epidemics on a global scale. They are transmitted by mosquitoes and cause diseases in humans that are mainly characterized by fever and often debilitating, sometimes chronic polyarthralgia. At present, approved treatments or vaccines are not available for most arthritogenic alphaviruses, and recently licensed vaccines against chikungunya virus are awaiting implementation in endemic areas. Most arthritogenic alphaviruses are currently limited to specific geographic areas due to vector distributions and availability of amplifying hosts, but they pose a substantial risk of emergence in other regions. The exception is chikungunya virus, which has emerged repeatedly from Africa, established sustained and efficient transmission in urban areas (including in temperate climates) and has caused major epidemics across the world. In this Review, we highlight recent advances in our understanding of the transmission cycles of arthritogenic alphaviruses, their vectors, epidemiology, transmission dynamics, evolution, pathophysiology and immune responses. We also outline strategies and countermeasures to anticipate and mitigate the impact of arthritogenic alphaviruses on human health.
Collapse
Affiliation(s)
- William M de Souza
- Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky, College of Medicine, Lexington, KY, USA
| | - Marc Lecuit
- Institut Pasteur, Université Paris Cité, Inserm U1117, Biology of Infection Unit, Paris, France
- Department of Infectious Diseases and Tropical Medicine, Assistance Publique-Hôpitaux de Paris, Institut Imagine, Necker-Enfants Malades University Hospital, Paris, France
| | - Scott C Weaver
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infections and Immunity, and Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
3
|
Castellón JO, Yuen C, Han B, Andrews KH, Ofori S, Julio AR, Boatner LM, Palafox MF, Perumal N, Damoiseaux R, Backus KM. An activation-based high throughput screen identifies caspase-10 inhibitors. RSC Chem Biol 2025; 6:604-617. [PMID: 40013156 PMCID: PMC11854450 DOI: 10.1039/d5cb00017c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
Caspases are a family of highly homologous cysteine proteases that play critical roles in inflammation and apoptosis. Small molecule inhibitors are useful tools for studying caspase biology, complementary to genetic approaches. However, achieving inhibitor selectivity for individual members of this highly homologous enzyme family remains a major challenge in developing such tool compounds. Prior studies have revealed that one strategy to tackle this selectivity gap is to target the precursor or zymogen forms of individual caspases, which share reduced structural homology when compared to active proteases. To establish a screening assay that favors the discovery of zymogen-directed caspase-10 selective inhibitors, we engineered a low-background and high-activity tobacco etch virus (TEV)-activated caspase-10 protein. We then subjected this turn-on protease to a high-throughput screen of approximately 100 000 compounds, with an average Z' value of 0.58 across all plates analyzed. Counter screening, including against TEV protease, delineated bona fide procaspase-10 inhibitors. Confirmatory studies identified a class of thiadiazine-containing compounds that undergo isomerization and oxidation to generate cysteine-reactive compounds with caspase-10 inhibitory activity. In parallel, mode-of-action studies revealed that pifithrin-μ (PFTμ), a reported TP53 inhibitor, also functions as a promiscuous caspase inhibitor. Both inhibitor classes showed preferential zymogen inhibition. Given the generalized utility of activation assays, we expect our screening platform to have widespread applications in identifying state-specific protease inhibitors.
Collapse
Affiliation(s)
- José O Castellón
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
| | - Constance Yuen
- California NanoSystems Institute (CNSI), UCLA Los Angeles CA 90095 USA
- Department of Molecular and Medical Pharmacology, UCLA Los Angeles CA 90095 USA
| | - Brandon Han
- California NanoSystems Institute (CNSI), UCLA Los Angeles CA 90095 USA
| | - Katrina H Andrews
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
| | - Samuel Ofori
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
| | - Ashley R Julio
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
- Department of Chemistry and Biochemistry UCLA CA 90095 USA
| | - Lisa M Boatner
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
- Department of Chemistry and Biochemistry UCLA CA 90095 USA
| | - Maria F Palafox
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
- Department of Chemistry and Biochemistry UCLA CA 90095 USA
- Department of Human Genetics, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
| | - Nithesh Perumal
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
- Department of Chemistry and Biochemistry UCLA CA 90095 USA
| | - Robert Damoiseaux
- California NanoSystems Institute (CNSI), UCLA Los Angeles CA 90095 USA
- Department of Molecular and Medical Pharmacology, UCLA Los Angeles CA 90095 USA
- Department of Bioengineering, Samueli School of Engineering, UCLA Los Angeles CA 90095 USA
- Jonsson Comprehensive Cancer Center, UCLA Los Angeles CA 90095 USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA Los Angeles CA 90095 USA
| | - Keriann M Backus
- Biological Chemistry Department, David Geffen School of Medicine, UCLA Los Angeles CA 90095 USA
- Department of Chemistry and Biochemistry UCLA CA 90095 USA
- California NanoSystems Institute (CNSI), UCLA Los Angeles CA 90095 USA
- Jonsson Comprehensive Cancer Center, UCLA Los Angeles CA 90095 USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, UCLA Los Angeles CA 90095 USA
- UCLA DOE Institute for Genomics and Proteomics, UCLA Los Angeles CA 90095 USA
| |
Collapse
|
4
|
Thiruvaiyaru A, Mattila S, Sadeghi M, Naumenko K, Merits A, Varjosalo M, Ahola T. Proximity interactome of alphavirus replicase component nsP3 includes proviral host factors eIF4G and AHNAK. PLoS Pathog 2025; 21:e1013050. [PMID: 40193402 PMCID: PMC12005498 DOI: 10.1371/journal.ppat.1013050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/17/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025] Open
Abstract
All positive-strand RNA viruses replicate their genomes in association with modified intracellular membranes, inducing either membrane invaginations termed spherules, or double-membrane vesicles. Alphaviruses encode four non-structural proteins nsP1-nsP4, all of which are essential for RNA replication and spherule formation. To understand the host factors associated with the replication complex, we fused the efficient biotin ligase miniTurbo with Semliki Forest virus (SFV) nsP3, which is located on the cytoplasmic surface of the spherules. We characterized the proximal proteome of nsP3 in three cell lines, including cells unable to form stress granules, and identified >300 host proteins constituting the microenvironment of nsP3. These included all the nsPs, as well as several previously characterized nsP3 binding proteins. However, the majority of the identified interactors had no previously identified roles in alphavirus replication, including 39 of the top 50 interacting proteins. The most prominent biological processes involving the proximal proteins were nucleic acid metabolism, translational regulation, cytoskeletal rearrangement and membrane remodeling. siRNA silencing confirmed six novel proviral factors, USP10, AHNAK, eIF4G1, SH3GL1, XAB2 and ANKRD17, which are associated with distinct cellular functions. All of these except SH3GL1 were also important for the replication of chikungunya virus. We discovered that the small molecule 4E1RCat, which inhibits the interaction between the canonical translation initiation factors eIF4G and eIF4E, exhibits antiviral activity against SFV. Since the same molecule was previously found to inhibit coronaviruses, this suggest the possibility that translation initiation factors could be considered as targets for broadly acting antivirals.
Collapse
Affiliation(s)
- Aditya Thiruvaiyaru
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Sari Mattila
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Mohammadreza Sadeghi
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | | | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Markku Varjosalo
- Institute of Biotechnology, HiLIFE Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tero Ahola
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| |
Collapse
|
5
|
Firdaus MER, Dukhno E, Kapoor R, Gerlach P. Two Birds With One Stone: RNA Virus Strategies to Manipulate G3BP1 and Other Stress Granule Components. WILEY INTERDISCIPLINARY REVIEWS. RNA 2025; 16:e70005. [PMID: 40170442 PMCID: PMC11962251 DOI: 10.1002/wrna.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 04/03/2025]
Abstract
Stress granules (SGs) are membrane-less organelles forming in the cytoplasm in response to various types of stress, including viral infection. SGs and SG-associated proteins can play either a proviral role, by facilitating viral replication, or an antiviral role, by limiting the translation capacity, sequestering viral RNA, or contributing to the innate immune response of the cell. Consequently, viruses frequently target stress granules while counteracting cellular translation shut-off and the antiviral response. One strategy is to sequester SG components, not only to impair their assembly but also to repurpose and incorporate them into viral replication sites. G3BP1 is a key SG protein, driving its nucleation through protein-protein and protein-RNA interactions. Many cellular proteins, including other SG components, interact with G3BP1 via their ΦxFG motifs. Notably, SARS-CoV N proteins and alphaviral nsP3 proteins contain similar motifs, allowing them to compete for G3BP1. Several SG proteins have been shown to interact with the flaviviral capsid protein, which is primarily responsible for anchoring the viral genome inside the virion. There are also numerous examples of structured elements within coronaviral and flaviviral RNAs recruiting or sponging SG proteins. Despite these insights, the structural and biochemical details of SG-virus interactions remain largely unexplored and are known only for a handful of cases. Exploring their molecular relevance for infection and discovering new examples of direct SG-virus contacts is highly important, as advances in this area will open new possibilities for the design of targeted therapies and potentially broad-spectrum antivirals.
Collapse
Affiliation(s)
- Moh Egy Rahman Firdaus
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | - Eliana Dukhno
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| | | | - Piotr Gerlach
- IMol Polish Academy of SciencesWarsawPoland
- ReMedy International Research Agenda UnitIMol Polish Academy of SciencesWarsawPoland
| |
Collapse
|
6
|
Li ZQ, Zhao LX, Wang SY, Hu CY, Wang YY, Yang Y. YBX1 is required for assembly of viral replication complexes of chikungunya virus and replication of multiple alphaviruses. J Virol 2025; 99:e0201524. [PMID: 39745458 PMCID: PMC11852927 DOI: 10.1128/jvi.02015-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/06/2024] [Indexed: 02/26/2025] Open
Abstract
Chikungunya virus (CHIKV), an enveloped positive-sense RNA virus, is a member of the alphaviruses and cause fever and arthralgia in humans. We performed genome-wide CRISPR/Cas9-based screens and identified Y-box binding protein 1 (YBX1) as an essential cellular factor for CHIKV. Deficiency of YBX1 inhibited CHIKV RNA replication and impaired virus production. Upon CHIKV infection, YBX1 showed a striking re-localization to viral replication complexes (vRCs), where it co-localized with CHIKV nsP3 and dsRNA intermediates. YBX1 directly interacted with CHIKV nsP3, and mutation of the YBX1-binding motif in CHIKV nsP3 suppressed viral replication in host cells. Furthermore, YBX1 bound to viral RNA and increased the viral RNA-binding activity of CHIKV nsP3. Consistently, the RNA-binding activity of YBX1, as well as the ability of nsP3 to bind to YBX1, was required for efficient CHIKV replication. In addition to CHIKV, YBX1 was also essential for replication of all examined alphaviruses including the prototypic alphavirus. Our findings suggest that YBX1 acts as a scaffold for assembly of chikungunya vRCs and an important factor for replication of multiple alphaviruses, which may serve as a potential target for the development of anti-alphavirus therapies.IMPORTANCEAlphaviruses are a group of mosquito-transmitted, enveloped, positive-strand RNA viruses in the Togaviridae family. Most alphaviruses are important pathogens that continue to cause human disease ranging from severe and potentially fatal neurological disease to chronic arthritic disease on a global scale. Here, we found that YBX1 promotes binding of CHIKV genomic RNA to nsP3, which is a key component of the replication complex, and is therefore pivotal for CHIKV replication. Deficiency of YBX1 results in reduced replication of multiple alphaviruses, including arthritogenic and encephalitic alphaviruses. These findings suggest that YBX1 is an important cellular factor for multiple alphaviruses and a potential target for preventing alphavirus infections.
Collapse
Affiliation(s)
- Zhen-Qi Li
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Li-Xin Zhao
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Su-Yun Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Chu-Yu Hu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yan-Yi Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Yan Yang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei, China
- University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
7
|
Zhang X, Li K, Zhou S, Zhang L, Wang L, Liu Y, Wang S, Xu G, Liang P, Xu Z, Song C. G3BP1 Regulates the Cell Cycle by Promoting IFNβ Production to Promote PCV2 Replication and Promotes Nuclear Transfer of Viral Proteins by Direct Binding. Int J Mol Sci 2025; 26:1083. [PMID: 39940851 PMCID: PMC11817264 DOI: 10.3390/ijms26031083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/27/2024] [Accepted: 01/09/2025] [Indexed: 02/16/2025] Open
Abstract
Porcine circovirus type 2 (PCV2) is a significant pathogen responsible for porcine circovirus-associated diseases (PCVAD), and it is widely prevalent in pig farms, leading to huge economic losses for the pig industry. Currently, the ability of PCV2 to enhance its own replication by using the antiviral inflammatory factors IFNα, IFNβ, and IL-2 and its complex immune escape mechanism remain unclear, which has attracted wide attention. Research has indicated that GTPase-activating protein (SH3 domain)-binding protein 1 (G3BP1) is involved in the innate immune response to a variety of viruses, primarily by regulating and composing stress granules (SGs) to inhibit viral replication. Our initial studies identified elevated G3BP1 expression during PCV2 infection, paradoxically promoting PCV2 replication. In light of this phenomenon, this study aims to elucidate how PCV2 regulates G3BP1 to enhance its replication. Our findings demonstrate that G3BP1 overexpression further activates PCV2-induced expression of RIG-I, MDA5, cGAS and STING, thereby promoting IFNβ production and affecting cell cycle arrest in the S phase, facilitating PCV2 replication. Moreover, interactions were observed between PCV2 Cap protein and G3BP1's RGG domain, and between PCV2 Rep protein and G3BP1's NTF2 and RRM domains, potentially promoting viral protein nuclear transfer. In summary, PCV2 enhances its replication by modulating G3BP1 to induce IFNβ production and directly binds viral proteins to promote viral protein nuclear transfer. This research provides a foundation for further investigation into the immune evasion mechanisms of PCV2.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zheng Xu
- College of Animal Science & National Engineering Center for Swine Breeding Industry, South China Agriculture University, Guangzhou 510642, China; (X.Z.); (K.L.); (S.Z.); (L.Z.); (L.W.); (Y.L.); (S.W.); (G.X.); (P.L.)
| | - Changxu Song
- College of Animal Science & National Engineering Center for Swine Breeding Industry, South China Agriculture University, Guangzhou 510642, China; (X.Z.); (K.L.); (S.Z.); (L.Z.); (L.W.); (Y.L.); (S.W.); (G.X.); (P.L.)
| |
Collapse
|
8
|
Larkin CI, Dunn MD, Shoemaker JE, Klimstra WB, Faeder JR. A detailed kinetic model of Eastern equine encephalitis virus replication in a susceptible host cell. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.13.628424. [PMID: 39764060 PMCID: PMC11703215 DOI: 10.1101/2024.12.13.628424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Eastern equine encephalitis virus (EEEV) is an arthropod-borne, positive-sense RNA alphavirus posing a substantial threat to public health. Unlike similar viruses such as SARS-CoV-2, EEEV replicates efficiently in neurons, producing progeny viral particles as soon as 3-4 hours post-infection. EEEV infection, which can cause severe encephalitis with a human mortality rate surpassing 30%, has no licensed, targeted therapies, leaving patients to rely on supportive care. Although the general characteristics of EEEV infection within the host cell are well-studied, it remains unclear how these interactions lead to rapid production of progeny viral particles, limiting development of antiviral therapies. Here, we present a novel rule-based model that describes attachment, entry, uncoating, replication, assembly, and export of both infectious virions and virus-like particles within mammalian cells. Additionally, it quantitatively characterizes host ribosome activity in EEEV replication via a model parameter defining ribosome density on viral RNA. To calibrate the model, we performed experiments to quantify viral RNA, protein, and infectious particle production during acute infection. We used Bayesian inference to calibrate the model, discovering in the process that an additional constraint was required to ensure consistency with previous experimental observations of a high ratio between the amounts of full-length positive-sense viral genome and negative-sense template strand. Overall, the model recapitulates the experimental data and predicts that EEEV rapidly concentrates host ribosomes densely on viral RNA. Dense packing of host ribosomes was determined to be critical to establishing the characteristic positive to negative RNA strand ratio because of its role in governing the kinetics of transcription. Sensitivity analysis identified viral transcription as the critical step for infectious particle production, making it a potential target for future therapeutic development.
Collapse
Affiliation(s)
- Caroline I. Larkin
- Joint Carnegie Mellon University - University of Pittsburgh PhD Program in Computational Biology, Pittsburgh, Pennsylvania, United States of America
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Matthew D. Dunn
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Jason E. Shoemaker
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Chemical and Petroleum Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - William B. Klimstra
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - James R. Faeder
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
9
|
Long S, Guzyk M, Perez Vidakovics L, Han X, Sun R, Wang M, Panas MD, Urgard E, Coquet JM, Merits A, Achour A, McInerney GM. SARS-CoV-2 N protein recruits G3BP to double membrane vesicles to promote translation of viral mRNAs. Nat Commun 2024; 15:10607. [PMID: 39638802 PMCID: PMC11621422 DOI: 10.1038/s41467-024-54996-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Ras-GTPase-activating protein SH3-domain-binding proteins (G3BP) are critical for the formation of stress granules (SGs) through their RNA- and ribosome-binding properties. SARS-CoV-2 nucleocapsid (N) protein exhibits strong binding affinity for G3BP and inhibits infection-induced SG formation soon after infection. To study the impact of the G3BP-N interaction on viral replication and pathogenesis in detail, we generated a mutant SARS-CoV-2 (RATA) that specifically lacks the G3BP-binding motif in the N protein. RATA triggers a stronger and more persistent SG response in infected cells, showing reduced replication across various cell lines, and greatly reduced pathogenesis in K18-hACE2 transgenic mice. At early times of infection, G3BP and WT N protein strongly colocalise with dsRNA and with non-structural protein 3 (nsp3), a component of the pore complex in double membrane vesicles (DMVs) from which nascent viral RNA emerges. Furthermore, G3BP-N complexes promote highly localized translation of viral mRNAs in the immediate vicinity of the DMVs and thus contribute to efficient viral gene expression and replication. In contrast, G3BP is absent from the DMVs in cells infected with RATA and translation of viral mRNAs is less efficient. This work provides a fuller understanding of the multifunctional roles of G3BP in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Siwen Long
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Mykhailo Guzyk
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Laura Perez Vidakovics
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Xiao Han
- Department of Medicine Solna, Science for Life Laboratory, Karolinska Institute Solna, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Renhua Sun
- Department of Medicine Solna, Science for Life Laboratory, Karolinska Institute Solna, Solna, Sweden
| | - Megan Wang
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Marc D Panas
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Egon Urgard
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Centre, University of Copenhagen, Copenhagen, Denmark
| | - Jonathan M Coquet
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Immunology and Microbiology, Leo Foundation Skin Immunology Research Centre, University of Copenhagen, Copenhagen, Denmark
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Adnane Achour
- Department of Medicine Solna, Science for Life Laboratory, Karolinska Institute Solna, Solna, Sweden
- Division of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Gerald M McInerney
- Division of Virology and Immunology, Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
10
|
Guo J, Huang R, Mei Y, Lu S, Gong J, Wang L, Ding L, Wu H, Pan D, Liu W. Application of stress granule core element G3BP1 in various diseases: A review. Int J Biol Macromol 2024; 282:137254. [PMID: 39515684 DOI: 10.1016/j.ijbiomac.2024.137254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 10/15/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Ras-GTPase-activating protein-binding protein 1 (G3BP1) is a core component and crucial regulatory switch in stress granules (SGs). When the concentration of free RNA within cells increases, it can trigger RNA-dependent liquid-liquid phase separation (LLPS) with G3BP1 as the core, thereby forming SGs that affect cell survival or death. In addition, G3BP1 interacts with various host proteins to regulate the expression of SGs. As a multifunctional binding protein, G3BP1 has diverse biological functions, influencing cell proliferation, differentiation, apoptosis, and RNA metabolism and serving as a crucial regulator in signaling pathways such as Rac1-PAK1, TSC-mTORC1, NF-κB, and STAT3. Therefore, it plays a significant role in the regulation of neurodegenerative diseases, myocardial hypertrophy, and congenital immunity, and is involved in the proliferation, invasion, and metastasis of cancer cells. G3BP1 is an important antiviral factor that interacts with viral proteins, and regulates SG assembly to exert antiviral effects. This article focuses on the recent discoveries and progress of G3BP1 in biology, including its structure and function, regulation of SG formation and dissolution, and its relationships with non-neoplastic diseases, tumors, and viruses.
Collapse
Affiliation(s)
- Jieyu Guo
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China; School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Rongyi Huang
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Yan Mei
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Siao Lu
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China; School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Jun Gong
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Long Wang
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Liqiong Ding
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Hongnian Wu
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Dan Pan
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China
| | - Wu Liu
- School of Basic Medical Sciences, Hubei Key Laboratory of Diabetes and Angiopathy, Xianning Medical College, Hubei University of Science and Technology, Xianning 437000, Hubei, China.
| |
Collapse
|
11
|
Qi X, Zhao R, Yao X, Liu Q, Liu P, Zhu Z, Tu C, Gong W, Li X. Getah virus Nsp3 binds G3BP to block formation of bona fide stress granules. Int J Biol Macromol 2024; 279:135274. [PMID: 39226976 DOI: 10.1016/j.ijbiomac.2024.135274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 08/16/2024] [Accepted: 08/31/2024] [Indexed: 09/05/2024]
Abstract
Stress granules (SGs) are cytoplasmic aggregates of proteins and mRNA that form in response to diverse environmental stressors, including viral infections. Several viruses possess the ability to block the formation of stress granules by targeting the SGs marker protein G3BP. However, the molecular functions and mechanisms underlying the regulation of SGs formation by Getah virus (GETV) remain unclear. In this study, we found that GETV infection triggered the formation of Nsp3-G3BP aggregates, which differed in composition from SGs. Further studies revealed that the presence of these aggregates was dependent on the activation of the PKR/eIF2α signaling pathway. Interestingly, we found that Nsp3 HVD domain blocked the formation of SGs by binding to G3BP NTF2 domain. Moreover, knockout of G3BP in NCI-H1299 cells had no effect on GETV replication, while overexpression of G3BP to form the genuine SGs significantly inhibited GETV replication. Overall, our study elucidates a novel role GETV Nsp3 to change the composition of SG as well as cellular stress response.
Collapse
Affiliation(s)
- Xiaoyi Qi
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Ruihan Zhao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Xiaohui Yao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Qinqiu Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Panrao Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Zhenbang Zhu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
| | - Changchun Tu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun 130122, China
| | - Wenjie Gong
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China; Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
12
|
Jiang Z, Merits A, Qin Y, Xing G, Zhang L, Chen J, Wang N, Varjak M, Zhai X, Li D, Song W, Su S. Attenuated Getah virus confers protection against multiple arthritogenic alphaviruses. PLoS Pathog 2024; 20:e1012700. [PMID: 39556619 PMCID: PMC11630583 DOI: 10.1371/journal.ppat.1012700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 12/10/2024] [Accepted: 10/27/2024] [Indexed: 11/20/2024] Open
Abstract
Alphaviruses are important arthropod-transmitted pathogens of humans and livestock. Getah virus (GETV) is an arthritogenic alphavirus that causes disease in horses and piglets; it also poses a potential threat to humans. A live attenuated vaccine candidate named GETV-3ΔS2-CM1, harbouring a deletion in nonstructural protein 3 and substitutions in the capsid protein, is genetically stable and exhibits robust immunogenicity. It was shown to confer passive protection to piglets born to immunized sows. In mice, a single dose of GETV-3ΔS2-CM1 protected against infection with different strains of GETV, Semliki Forest virus, Ross River virus, o'nyong'nyong virus, chikungunya virus, and Barmah Forest virus. Chimaeras based on the GETV-3ΔS2-CM1 backbone maintained both the attenuated phenotype and high immunogenicity. The safety, efficacy, and ability to induce protection against multiple alphaviruses highlights the potential of GETV-3ΔS2-CM1 and chimaeras using this backbone as promising vaccine candidates. By contributing simultaneously to the wellbeing of animals and humans, our universal next generation vaccine strategy helps to achieve "One Health" goals.
Collapse
Affiliation(s)
- Zhiwen Jiang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Ying Qin
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Gang Xing
- MOA Key Laboratory of Animal Virology, Zhejiang University, Hangzhou, China
| | - Letian Zhang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Jie Chen
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ningning Wang
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Margus Varjak
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Xiaofeng Zhai
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Dongyan Li
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Wanjie Song
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Shuo Su
- Sanya Institute of Nanjing Agricultural University, Academy for Advanced Interdisciplinary Studies, Jiangsu Engineering Laboratory of Animal Immunology, Institute of Immunology and College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
13
|
Liu J, Guan G, Wu C, Wang B, Chu K, Zhang X, He S, Zhang N, Yang G, Jin Z, Zhao T. SARS-CoV-2 Nucleocapsid Protein Antagonizes GADD34-Mediated Innate Immune Pathway through Atypical Foci. Molecules 2024; 29:4792. [PMID: 39459161 PMCID: PMC11510332 DOI: 10.3390/molecules29204792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/22/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
The integrated stress response, especially stress granules (SGs), contributes to host immunity. Typical G3BP1+ stress granules (tSGs) are usually formed after virus infection to restrain viral replication and stimulate innate immunity. Recently, several SG-like foci or atypical SGs (aSGs) with proviral function have been found during viral infection. We have shown that the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) nucleocapsid (N) protein induces atypical N+/G3BP1+ foci (N+foci), leading to the inhibition of host immunity and facilitation of viral infection. However, the precise mechanism has not been well clarified yet. In this study, we showed that the SARS-CoV-2 N (SARS2-N) protein inhibits dsRNA-induced growth arrest and DNA damage-inducible 34 (GADD34) expression. Mechanistically, the SARS2-N protein promotes the interaction between GADD34 mRNA and G3BP1, sequestering GADD34 mRNA into the N+foci. Importantly, we found that GADD34 participates in IRF3 nuclear translocation through its KVRF motif and promotes the transcription of downstream interferon genes. The suppression of GADD34 expression by the SARS2-N protein impairs the nuclear localization of IRF3 and compromises the host's innate immune response, which facilitates viral replication. Taking these findings together, our study revealed a novel mechanism by which the SARS2-N protein antagonized the GADD34-mediated innate immune pathway via induction of N+foci. We think this is a critical strategy for viral pathogenesis and has potential therapeutic implications.
Collapse
Affiliation(s)
- Jie Liu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Guanwen Guan
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Chunxiu Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Bingbing Wang
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Kaifei Chu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Xu Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Su He
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Naru Zhang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Geng Yang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
| | - Zhigang Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| | - Tiejun Zhao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou 310015, China
- College of Life Sciences, Zhejiang Normal University, Jinhua 321004, China
| |
Collapse
|
14
|
Glineburg M, Yildirim E, Gomez N, Rodriguez G, Pak J, Li X, Altheim C, Waksmacki J, McInerney G, Barmada S, Todd P. Stress granule formation helps to mitigate neurodegeneration. Nucleic Acids Res 2024; 52:9745-9759. [PMID: 39106168 PMCID: PMC11381325 DOI: 10.1093/nar/gkae655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 08/09/2024] Open
Abstract
Cellular stress pathways that inhibit translation initiation lead to transient formation of cytoplasmic RNA/protein complexes known as stress granules. Many of the proteins found within stress granules and the dynamics of stress granule formation and dissolution are implicated in neurodegenerative disease. Whether stress granule formation is protective or harmful in neurodegenerative conditions is not known. To address this, we took advantage of the alphavirus protein nsP3, which selectively binds dimers of the central stress granule nucleator protein G3BP and markedly reduces stress granule formation without directly impacting the protein translational inhibitory pathways that trigger stress granule formation. In Drosophila and rodent neurons, reducing stress granule formation with nsP3 had modest impacts on lifespan even in the setting of serial stress pathway induction. In contrast, reducing stress granule formation in models of ataxia, amyotrophic lateral sclerosis and frontotemporal dementia largely exacerbated disease phenotypes. These data support a model whereby stress granules mitigate, rather than promote, neurodegenerative cascades.
Collapse
Affiliation(s)
- M Rebecca Glineburg
- Biological Sciences, Schmid College of Science and Technology, Chapman University, 1 University Drive, Orange, CA 92866, USA
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Evrim Yildirim
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Nicolas Gomez
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
- Cell and Molecular Biology Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Genesis Rodriguez
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Jaclyn Pak
- Biological Sciences, Schmid College of Science and Technology, Chapman University, 1 University Drive, Orange, CA 92866, USA
| | - Xingli Li
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Christopher Altheim
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Jacob Waksmacki
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Gerald M McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm 17165, Sweden
| | - Sami J Barmada
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
| | - Peter K Todd
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, BSRB48109-2200, Ann Arbor, MI 4005, USA
- Veterans Affairs Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Omler A, Mutso M, Vaher M, Freitas JR, Taylor A, David CT, Moseley GW, Liu X, Merits A, Mahalingam S. Exploring Barmah Forest virus pathogenesis: molecular tools to investigate non-structural protein 3 nuclear localization and viral genomic determinants of replication. mBio 2024; 15:e0099324. [PMID: 38953633 PMCID: PMC11323547 DOI: 10.1128/mbio.00993-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 05/03/2024] [Indexed: 07/04/2024] Open
Abstract
Barmah Forest virus (BFV) is a mosquito-borne virus that causes arthralgia with accompanying rash, fever, and myalgia in humans. The virus is mainly found in Australia and has caused outbreaks associated with significant health concerns. As the sole representative of the Barmah Forest complex within the genus Alphavirus, BFV is not closely related genetically to other alphaviruses. Notably, basic knowledge of BFV molecular virology has not been well studied due to a lack of critical investigative tools such as an infectious clone. Here we describe the construction of an infectious BFV cDNA clone based on Genbank sequence and demonstrate that the clone-derived virus has in vitro and in vivo properties similar to naturally occurring virus, BFV field isolate 2193 (BFV2193-FI). A substitution in nsP4, V1911D, which was identified in the Genbank reference sequence, was found to inhibit virus rescue and replication. T1325P substitution in nsP2 selected during virus passaging was shown to be an adaptive mutation, compensating for the inhibitory effect of nsP4-V1911D. The two mutations were associated with changes in viral non-structural polyprotein processing and type I interferon (IFN) induction. Interestingly, a nuclear localization signal, active in mammalian but not mosquito cells, was identified in nsP3. A point mutation abolishing nsP3 nuclear localization attenuated BFV replication. This effect was more prominent in the presence of type I interferon signaling, suggesting nsP3 nuclear localization might be associated with IFN antagonism. Furthermore, abolishing nsP3 nuclear localization reduced virus replication in mice but did not significantly affect disease.IMPORTANCEBarmah Forest virus (BFV) is Australia's second most prevalent arbovirus, with approximately 1,000 cases reported annually. The clinical symptoms of BFV infection include rash, polyarthritis, arthralgia, and myalgia. As BFV is not closely related to other pathogenic alphaviruses or well-studied model viruses, our understanding of its molecular virology and mechanisms of pathogenesis is limited. There is also a lack of molecular tools essential for corresponding studies. Here we describe the construction of an infectious clone of BFV, variants harboring point mutations, and sequences encoding marker protein. In infected mammalian cells, nsP3 of BFV was located in the nuclei. This finding extends our understanding of the diverse mechanisms used by alphavirus replicase proteins to interact with host cells. Our novel observations highlight the complex synergy through which the viral replication machinery evolves to correct mutation errors within the viral genome.
Collapse
Affiliation(s)
- Ailar Omler
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Margit Mutso
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Mihkel Vaher
- The Institute of Molecular and Cell Biology, University of Tartu, Tartu, Estonia
| | - Joseph R. Freitas
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Adam Taylor
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Cassandra T. David
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gregory W. Moseley
- Department of Microbiology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Xiang Liu
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| | - Andres Merits
- Institute of Bioengineering, University of Tartu, Tartu, Estonia
| | - Suresh Mahalingam
- Emerging Viruses, Inflammation and Therapeutics Group, Menzies Health Institute Queensland, Griffith University, Gold Coast, Queensland, Australia
- Global Virus Network (GVN) Centre for Excellence in Arboviruses, Griffith University, Gold Coast, Queensland, Australia
- School of Pharmacy and Medical Sciences, Griffith University, Gold Coast, Queensland, Australia
| |
Collapse
|
16
|
den Boon JA, Nishikiori M, Zhan H, Ahlquist P. Positive-strand RNA virus genome replication organelles: structure, assembly, control. Trends Genet 2024; 40:681-693. [PMID: 38724328 DOI: 10.1016/j.tig.2024.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 08/09/2024]
Abstract
Positive-strand RNA [(+)RNA] viruses include pandemic SARS-CoV-2, tumor-inducing hepatitis C virus, debilitating chikungunya virus (CHIKV), lethal encephalitis viruses, and many other major pathogens. (+)RNA viruses replicate their RNA genomes in virus-induced replication organelles (ROs) that also evolve new viral species and variants by recombination and mutation and are crucial virus control targets. Recent cryo-electron microscopy (cryo-EM) reveals that viral RNA replication proteins form striking ringed 'crowns' at RO vesicle junctions with the cytosol. These crowns direct RO vesicle formation, viral (-)RNA and (+)RNA synthesis and capping, innate immune escape, and transfer of progeny (+)RNA genomes into translation and encapsidation. Ongoing studies are illuminating crown assembly, sequential functions, host factor interactions, etc., with significant implications for control and beneficial uses of viruses.
Collapse
Affiliation(s)
- Johan A den Boon
- Rowe Center for Virology, Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI; McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI
| | - Masaki Nishikiori
- Rowe Center for Virology, Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI; McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI
| | - Hong Zhan
- Rowe Center for Virology, Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI; McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI
| | - Paul Ahlquist
- Rowe Center for Virology, Morgridge Institute for Research, Madison, WI, USA; Institute for Molecular Virology, University of Wisconsin-Madison, Madison, WI; McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI.
| |
Collapse
|
17
|
Drago L, Pennati A, Rothbächer U, Ashita R, Hashimoto S, Saito R, Fujiwara S, Ballarin L. Stress granule-related genes during embryogenesis of an invertebrate chordate. Front Cell Dev Biol 2024; 12:1414759. [PMID: 39149517 PMCID: PMC11324471 DOI: 10.3389/fcell.2024.1414759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 06/20/2024] [Indexed: 08/17/2024] Open
Abstract
Controlling global protein synthesis through the assembly of stress granules represents a strategy adopted by eukaryotic cells to face various stress conditions. TIA 1-related nucleolysin (TIAR), tristetraprolin (TTP), and Ras-GTPase-activating protein SH3-domain-binding protein (G3BP) are key components of stress granules, allowing the regulation of mRNA stability, and thus controlling not only stress responses but also cell proliferation and differentiation. In this study, we aimed at investigating the roles of tiar, ttp, and g3bp during embryogenesis of the solitary ascidian Ciona robusta under both physiological and stress conditions. We carried out CRISPR/Cas9 to evaluate the effects of gene knockout on normal embryonic development, and gene reporter assay to study the time and tissue specificity of gene transcription, together with whole-mount in situ hybridization and quantitative real time PCR. To induce acute stress conditions, we used iron and cadmium as "essential" and "non-essential" metals, respectively. Our results highlight, for the first time, the importance of tiar, ttp, and g3bp in controlling the development of mesendodermal tissue derivatives during embryogenesis of an invertebrate chordate.
Collapse
Affiliation(s)
- Laura Drago
- Department of Biology, University of Padova, Padua, Italy
| | | | - Ute Rothbächer
- Institute of Zoology, University of Innsbruck, Innsbruck, Austria
| | - Ryuji Ashita
- Department of Chemistry and Biotechnology, University of Kochi, Kochi, Japan
| | - Seika Hashimoto
- Department of Chemistry and Biotechnology, University of Kochi, Kochi, Japan
| | - Ryota Saito
- Department of Chemistry and Biotechnology, University of Kochi, Kochi, Japan
| | - Shigeki Fujiwara
- Department of Chemistry and Biotechnology, University of Kochi, Kochi, Japan
| | | |
Collapse
|
18
|
Peng Q, Jiao B, Cheng Y, Yuan B, Zhou J, Cai J, Jiang N, Lin H, Xi D. NtG3BPL1 confers resistance to chilli veinal mottle virus through promoting the degradation of 6K2 in tobacco. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2024; 119:720-734. [PMID: 38713838 DOI: 10.1111/tpj.16789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 03/01/2024] [Accepted: 04/10/2024] [Indexed: 05/09/2024]
Abstract
The RNA regulatory network is a complex and dynamic regulation in plant cells involved in mRNA modification, translation, and degradation. Ras-GAP SH3 domain-binding protein (G3BP) is a scaffold protein for the assembly of stress granules (SGs) and is considered an antiviral component in mammals. However, the function of G3BP during virus infection in plants is still largely unknown. In this study, four members of the G3BP-like proteins (NtG3BPLs) were identified in Nicotiana tabacum and the expression levels of NtG3BPL1 were upregulated during chilli veinal mottle virus (ChiVMV) infection. NtG3BPL1 was localized in the nucleus and cytoplasm, forming cytoplasmic granules under transient high-temperature treatment, whereas the abundance of cytoplasmic granules was decreased under ChiVMV infection. Overexpression of NtG3BPL1 inhibited ChiVMV infection and delayed the onset of symptoms, whereas knockout of NtG3BPL1 promoted ChiVMV infection. In addition, NtG3BPL1 directly interacted with ChiVMV 6K2 protein, whereas 6K2 protein had no effect on NtG3BPL1-derived cytoplasmic granules. Further studies revealed that the expression of NtG3BPL1 reduced the chloroplast localization of 6K2-GFP and the NtG3BPL1-6K2 interaction complex was localized in the cytoplasm. Furthermore, NtG3BPL1 promoted the degradation of 6K2 through autophagy pathway, and the accumulation of 6K2 and ChiVMV was affected by autophagy activation or inhibition in plants. Taken together, our results demonstrate that NtG3BPL1 plays a positive role in tobacco resistance against ChiVMV infection, revealing a novel mechanism of plant G3BP in antiviral strategy.
Collapse
Affiliation(s)
- Qiding Peng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Bolei Jiao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Yongchao Cheng
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Bowen Yuan
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Jingya Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Jingliu Cai
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Ning Jiang
- Agronomic Research Center, Yunnan Academy of Tobacco Agricultural Sciences, Kunming, 650021, Yunnan, P.R. China
| | - Honghui Lin
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| | - Dehui Xi
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, Sichuan, P.R. China
| |
Collapse
|
19
|
Li M, Hou Y, Zhou Y, Yang Z, Zhao H, Jian T, Yu Q, Zeng F, Liu X, Zhang Z, Zhao YG. LLPS of FXR proteins drives replication organelle clustering for β-coronaviral proliferation. J Cell Biol 2024; 223:e202309140. [PMID: 38587486 PMCID: PMC11001562 DOI: 10.1083/jcb.202309140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/16/2024] [Accepted: 02/26/2024] [Indexed: 04/09/2024] Open
Abstract
β-Coronaviruses remodel host endomembranes to form double-membrane vesicles (DMVs) as replication organelles (ROs) that provide a shielded microenvironment for viral RNA synthesis in infected cells. DMVs are clustered, but the molecular underpinnings and pathophysiological functions remain unknown. Here, we reveal that host fragile X-related (FXR) family proteins (FXR1/FXR2/FMR1) are required for DMV clustering induced by expression of viral non-structural proteins (Nsps) Nsp3 and Nsp4. Depleting FXRs results in DMV dispersion in the cytoplasm. FXR1/2 and FMR1 are recruited to DMV sites via specific interaction with Nsp3. FXRs form condensates driven by liquid-liquid phase separation, which is required for DMV clustering. FXR1 liquid droplets concentrate Nsp3 and Nsp3-decorated liposomes in vitro. FXR droplets facilitate recruitment of translation machinery for efficient translation surrounding DMVs. In cells depleted of FXRs, SARS-CoV-2 replication is significantly attenuated. Thus, SARS-CoV-2 exploits host FXR proteins to cluster viral DMVs via phase separation for efficient viral replication.
Collapse
Affiliation(s)
- Meng Li
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Yali Hou
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Yuzheng Zhou
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Zhenni Yang
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Hongyu Zhao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, P.R. China
| | - Tao Jian
- Division of Life Science, State Key Laboratory of Molecular Neuroscience, Hong Kong University of Science and Technology, Kowloon, P.R. China
| | - Qianxi Yu
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Fuxing Zeng
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
- Institute for Biological Electron Microscopy, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Xiaotian Liu
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Zheng Zhang
- Institute for Hepatology, National Clinical Research Center for Infectious Disease, Shenzhen Third People’s Hospital, The Second Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, P.R. China
| | - Yan G. Zhao
- Shenzhen Key Laboratory of Biomolecular Assembling and Regulation, School of Life Sciences, Southern University of Science and Technology, Shenzhen, P.R. China
| |
Collapse
|
20
|
Nie X, Wang D, Pan Y, Hua Y, Lü P, Yang Y. Discovery, classification and application of the CPISPR-Cas13 system. Technol Health Care 2024; 32:525-544. [PMID: 37545273 DOI: 10.3233/thc-230258] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND The clustered regularly interspaced short palindromic repeats (CRISPR)-Cas system is an acquired immune system of bacteria and archaea. Continued research has resulted in the identification of other Cas13 proteins. OBJECTIVE This review briefly describes the discovery, classification, and application of the CRISPR-Cas13 system, including recent technological advances in addition to factors affecting system performance. METHODS Cas13-based molecular therapy of human, animal, and plant transcriptomes was discussed, including regulation of gene expression to combat pathogenic RNA viruses. In addition, the latest progress, potential shortcomings, and challenges of the CRISPR-Cas system for treatment of animal and plant diseases are reviewed. RESULTS The CRISPR-Cas system VI is characterized by two RNA-guided higher eukaryotes and prokaryotes nucleotide-binding domains. CRISPR RNA can cleave specific RNA through the interaction between the stem-loop rich chain of uracil residues and the Cas13a protein. The CRISPR-Cas13 system has been applied for gene editing in animal and plant cells, in addition to biological detection via accurate targeting of single-stranded RNA. CONCLUSION The CRISPR-Cas13 system offers a high-throughput and convenient technology for detection of viruses and potentially the development of anti-cancer drugs in the near future.
Collapse
Affiliation(s)
- Xiaojuan Nie
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Dandan Wang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ye Pan
- School of Experimental Animal Center, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ye Hua
- Institute of Oncology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Peng Lü
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Yanhua Yang
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
21
|
He S, Gou H, Zhou Y, Wu C, Ren X, Wu X, Guan G, Jin B, Huang J, Jin Z, Zhao T. The SARS-CoV-2 nucleocapsid protein suppresses innate immunity by remodeling stress granules to atypical foci. FASEB J 2023; 37:e23269. [PMID: 37889852 DOI: 10.1096/fj.202201973rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 08/10/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023]
Abstract
Viruses deploy multiple strategies to suppress the host innate immune response to facilitate viral replication and pathogenesis. Typical G3BP1+ stress granules (SGs) are usually formed in host cells after virus infection to restrain viral translation and to stimulate innate immunity. Thus, viruses have evolved various mechanisms to inhibit SGs or to repurpose SG components such as G3BP1. Previous studies showed that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection inhibited host immunity during the early stage of COVID-19. However, the precise mechanism is not yet well understood. Here we showed that the SARS-CoV-2 nucleocapsid (SARS2-N) protein suppressed the double-stranded RNA (dsRNA)-induced innate immune response, concomitant with inhibition of SGs and the induction of atypical SARS2-N+ /G3BP1+ foci (N+ foci). The SARS2-N protein-induced formation of N+ foci was dependent on the ability of its ITFG motif to hijack G3BP1, which contributed to suppress the innate immune response. Importantly, SARS2-N protein facilitated viral replication by inducing the formation of N+ foci. Viral mutations within SARS2-N protein that impair the formation of N+ foci are associated with the inability of the SARS2-N protein to suppress the immune response. Taken together, our study has revealed a novel mechanism by which SARS-CoV-2 suppresses the innate immune response via induction of atypical N+ foci. We think that this is a critical strategy for viral pathogenesis and has potential therapeutic implications.
Collapse
Affiliation(s)
- Su He
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Hongwei Gou
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Yulin Zhou
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Chunxiu Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Xinxin Ren
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xiajunpeng Wu
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Guanwen Guan
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Boxing Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Jinhua Huang
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Zhigang Jin
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
| | - Tiejun Zhao
- College of Life Sciences, Zhejiang Normal University, Jinhua, Zhejiang, China
- School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| |
Collapse
|
22
|
Glineburg MR, Yildirim E, Gomez N, Li X, Pak J, Altheim C, Waksmacki J, McInerney G, Barmada SJ, Todd PK. Stress granule formation helps to mitigate neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566060. [PMID: 37986813 PMCID: PMC10659376 DOI: 10.1101/2023.11.07.566060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Cellular stress pathways that inhibit translation initiation lead to transient formation of cytoplasmic RNA/protein complexes known as stress granules. Many of the proteins found within stress granules and the dynamics of stress granule formation and dissolution are implicated in neurodegenerative disease. Whether stress granule formation is protective or harmful in neurodegenerative conditions is not known. To address this, we took advantage of the alphavirus protein nsP3, which selectively binds dimers of the central stress granule nucleator protein G3BP (rin in Drosophila) and markedly reduces stress granule formation without directly impacting the protein translational inhibitory pathways that trigger stress granule formation. In Drosophila and rodent neurons, reducing stress granule formation with nsP3 had modest impacts on lifespan even in the setting of serial stress pathway induction. In contrast, reducing stress granule formation in models of ataxia, amyotrophic lateral sclerosis and frontotemporal dementia largely exacerbated disease phenotypes. These data support a model whereby stress granules mitigate, rather than promote, neurodegenerative cascades.
Collapse
Affiliation(s)
- M. Rebecca Glineburg
- Biological Sciences, Schmid College of Science and Technology, Chapman University, 450 N. Center St, Orange, CA 92866
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Evrim Yildirim
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Nicolas Gomez
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Xingli Li
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Jaclyn Pak
- Biological Sciences, Schmid College of Science and Technology, Chapman University, 450 N. Center St, Orange, CA 92866
| | - Christopher Altheim
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Jacob Waksmacki
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Gerald McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 17165, Sweden
| | - Sami J. Barmada
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
| | - Peter K. Todd
- Department of Neurology, University of Michigan, 109 Zina Pitcher Place, Ann Arbor, MI, 4005 BSRB48109-2200, USA
- Veterans Affairs Medical Center, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Mukhopadhyay C, Zhou P. Role(s) of G3BPs in Human Pathogenesis. J Pharmacol Exp Ther 2023; 387:100-110. [PMID: 37468286 PMCID: PMC10519580 DOI: 10.1124/jpet.122.001538] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 06/28/2023] [Accepted: 07/11/2023] [Indexed: 07/21/2023] Open
Abstract
Ras-GTPase-activating protein (SH3 domain)-binding proteins (G3BP) are RNA binding proteins that play a critical role in stress granule (SG) formation. SGs protect critical mRNAs from various environmental stress conditions by regulating mRNA stability and translation to maintain regulated gene expression. Recent evidence suggests that G3BPs can also regulate mRNA expression through interactions with RNA outside of SGs. G3BPs have been associated with a number of disease states, including cancer progression, invasion, metastasis, and viral infections, and may be useful as a cancer therapeutic target. This review summarizes the biology of G3BP including their structure, function, localization, role in cancer progression, virus replication, mRNA stability, and SG formation. We will also discuss the potential of G3BPs as a therapeutic target. SIGNIFICANCE STATEMENT: This review will discuss the molecular mechanism(s) and functional role(s) of Ras-GTPase-activating protein (SH3 domain)-binding proteins in the context of stress granule formation, interaction with viruses, stability of RNA, and tumorigenesis.
Collapse
Affiliation(s)
- Chandrani Mukhopadhyay
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York
| | - Pengbo Zhou
- Department of Pathology and Laboratory Medicine, Weill Medical College of Cornell University, New York
| |
Collapse
|
24
|
Rademacher DJ, Bello AI, May JP. CASC3 Biomolecular Condensates Restrict Turnip Crinkle Virus by Limiting Host Factor Availability. J Mol Biol 2023; 435:167956. [PMID: 36642157 PMCID: PMC10338645 DOI: 10.1016/j.jmb.2023.167956] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 12/15/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
The exon-junction complex (EJC) plays a role in post-transcriptional gene regulation and exerts antiviral activity towards several positive-strand RNA viruses. However, the spectrum of RNA viruses that are targeted by the EJC or the underlying mechanisms are not well understood. EJC components from Arabidopsis thaliana were screened for antiviral activity towards Turnip crinkle virus (TCV, Tombusviridae). Overexpression of the accessory EJC component CASC3 inhibited TCV accumulation > 10-fold in Nicotiana benthamiana while knock-down of endogenous CASC3 resulted in a > 4-fold increase in TCV accumulation. CASC3 forms cytoplasmic condensates and deletion of the conserved SELOR domain reduced condensate size 7-fold and significantly decreased antiviral activity towards TCV. Mass spectrometry of CASC3 complexes did not identify endogenous stress granule or P-body markers and CASC3 failed to co-localize with an aggresome-specific dye suggesting that CASC3 condensates are distinct from well-established membraneless compartments. Mass spectrometry and bimolecular fluorescence complementation assays revealed that CASC3 sequesters Heat shock protein 70 (Hsp70-1) and Glyceraldehyde-3-phosphate dehydrogenase (GAPDH), two host factors with roles in tombusvirus replication. Overexpression of Hsp70-1 or GAPDH reduced the antiviral activity of CASC3 2.1-fold and 2.8-fold, respectively, and suggests that CASC3 inhibits TCV by limiting host factor availability. Unrelated Tobacco mosaic virus (TMV) also depends on Hsp70-1 and CASC3 overexpression restricted TMV accumulation 4-fold and demonstrates that CASC3 antiviral activity is not TCV-specific. Like CASC3, Auxin response factor 19 (ARF19) forms poorly dynamic condensates but ARF19 overexpression failed to inhibit TCV accumulation and suggests that CASC3 has antiviral activities that are not ubiquitous among cytoplasmic condensates.
Collapse
Affiliation(s)
- Dana J Rademacher
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA
| | - Abudu I Bello
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA
| | - Jared P May
- Division of Biological and Biomedical Systems, School of Science and Engineering, University of Missouri-Kansas City, 5009 Rockhill Road, Kansas City, MO 64110, USA.
| |
Collapse
|
25
|
Yin P, Jian X, Liu Y, Liu Y, Lv L, Cui H, Zhang L. Elucidating cellular interactome of chikungunya virus identifies host dependency factors. Virol Sin 2023; 38:497-507. [PMID: 37182691 PMCID: PMC10436055 DOI: 10.1016/j.virs.2023.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 05/11/2023] [Indexed: 05/16/2023] Open
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-transmitted RNA virus causing joint and muscle pain. To better understand how CHIKV rewires the host cell and usurps host cell functions, we generated a systematic CHIKV-human protein-protein interaction map and revealed several novel connections that will inform further mechanistic studies. One of these novel interactions, between the viral protein E1 and STIP1 homology and U-box containing protein 1 (STUB1), was found to mediate ubiquitination of E1 and degrade E1 through the proteasome. Capsid associated with G3BP1, G3BP2 and AAA+ ATPase valosin-containing protein (VCP). Furthermore, VCP inhibitors blocked CHIKV infection, suggesting VCP could serve as a therapeutic target. Further work is required to fully understand the functional consequences of these interactions. Given that CHIKV proteins are conserved across alphaviruses, many virus-host protein-protein interactions identified in this study might also exist in other alphaviruses. Construction of interactome of CHIKV provides the basis for further studying the function of alphavirus biology.
Collapse
Affiliation(s)
- Peiqi Yin
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250013, China; NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100176, China
| | - Xia Jian
- NHC Key Laboratory of Systems Biology of Pathogens, Institute of Pathogen Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100176, China
| | - Yihan Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Yuwen Liu
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Lu Lv
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Haoran Cui
- Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Leiliang Zhang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250013, China; Department of Pathogen Biology, School of Clinical and Basic Medical Sciences, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
26
|
Abstract
RNA granules are mesoscale assemblies that form in the absence of limiting membranes. RNA granules contain factors for RNA biogenesis and turnover and are often assumed to represent specialized compartments for RNA biochemistry. Recent evidence suggests that RNA granules assemble by phase separation of subsoluble ribonucleoprotein (RNP) complexes that partially demix from the cytoplasm or nucleoplasm. We explore the possibility that some RNA granules are nonessential condensation by-products that arise when RNP complexes exceed their solubility limit as a consequence of cellular activity, stress, or aging. We describe the use of evolutionary and mutational analyses and single-molecule techniques to distinguish functional RNA granules from "incidental condensates."
Collapse
Affiliation(s)
- Andrea Putnam
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Laura Thomas
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | - Geraldine Seydoux
- Howard Hughes Medical Institute, Department of Molecular Biology and Genetics, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
27
|
Schulte T, Panas MD, Han X, Williams L, Kedersha N, Fleck JS, Tan TJC, Dopico XC, Olsson A, Morro AM, Hanke L, Nilvebrant J, Giang KA, Nygren PÅ, Anderson P, Achour A, McInerney GM. Caprin-1 binding to the critical stress granule protein G3BP1 is influenced by pH. Open Biol 2023; 13:220369. [PMID: 37161291 PMCID: PMC10170197 DOI: 10.1098/rsob.220369] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 03/28/2023] [Indexed: 05/11/2023] Open
Abstract
G3BP is the central node within stress-induced protein-RNA interaction networks known as stress granules (SGs). The SG-associated proteins Caprin-1 and USP10 bind mutually exclusively to the NTF2 domain of G3BP1, promoting and inhibiting SG formation, respectively. Herein, we present the crystal structure of G3BP1-NTF2 in complex with a Caprin-1-derived short linear motif (SLiM). Caprin-1 interacts with His-31 and His-62 within a third NTF2-binding site outside those covered by USP10, as confirmed using biochemical and biophysical-binding assays. Nano-differential scanning fluorimetry revealed reduced thermal stability of G3BP1-NTF2 at acidic pH. This destabilization was counterbalanced significantly better by bound USP10 than Caprin-1. The G3BP1/USP10 complex immunoprecipated from human U2OS cells was more resistant to acidic buffer washes than G3BP1/Caprin-1. Acidification of cellular condensates by approximately 0.5 units relative to the cytosol was detected by ratiometric fluorescence analysis of pHluorin2 fused to G3BP1. Cells expressing a Caprin-1/FGDF chimera with higher G3BP1-binding affinity had reduced Caprin-1 levels and slightly reduced condensate sizes. This unexpected finding may suggest that binding of the USP10-derived SLiM to NTF2 reduces the propensity of G3BP1 to enter condensates.
Collapse
Affiliation(s)
- Tim Schulte
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Division of Infectious Diseases, Karolinska University Hospital, Stockholm, 171 77, Sweden
| | - Marc D. Panas
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Xiao Han
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Division of Infectious Diseases, Karolinska University Hospital, Stockholm, 171 77, Sweden
| | - Lucy Williams
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Nancy Kedersha
- Division of Rheumatology, Immunity, and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Jonas Simon Fleck
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Division of Infectious Diseases, Karolinska University Hospital, Stockholm, 171 77, Sweden
| | - Timothy J. C. Tan
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Xaquin Castro Dopico
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Anders Olsson
- Protein Expression and Characterization, AlbaNova University Center, Royal Institute of Technology, 114 21, Stockholm
| | - Ainhoa Moliner Morro
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Leo Hanke
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| | - Johan Nilvebrant
- Division of Protein Engineering, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Center, Royal Institute of Technology, 114 21, Stockholm
- Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Sweden
| | - Kim Anh Giang
- Division of Protein Engineering, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Center, Royal Institute of Technology, 114 21, Stockholm
- Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Sweden
| | - Per-Åke Nygren
- Division of Protein Engineering, Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, AlbaNova University Center, Royal Institute of Technology, 114 21, Stockholm
- Science for Life Laboratory, Tomtebodavägen 23A, 171 65, Sweden
| | - Paul Anderson
- Division of Rheumatology, Immunity, and Inflammation, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
- Harvard Medical School Initiative for RNA Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Adnane Achour
- Science for Life Laboratory, Department of Medicine Solna, Karolinska Institutet, and Division of Infectious Diseases, Karolinska University Hospital, Stockholm, 171 77, Sweden
| | - Gerald M. McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, 171 77, Sweden
| |
Collapse
|
28
|
Kim DK, Weller B, Lin CW, Sheykhkarimli D, Knapp JJ, Dugied G, Zanzoni A, Pons C, Tofaute MJ, Maseko SB, Spirohn K, Laval F, Lambourne L, Kishore N, Rayhan A, Sauer M, Young V, Halder H, la Rosa NMD, Pogoutse O, Strobel A, Schwehn P, Li R, Rothballer ST, Altmann M, Cassonnet P, Coté AG, Vergara LE, Hazelwood I, Liu BB, Nguyen M, Pandiarajan R, Dohai B, Coloma PAR, Poirson J, Giuliana P, Willems L, Taipale M, Jacob Y, Hao T, Hill DE, Brun C, Twizere JC, Krappmann D, Heinig M, Falter C, Aloy P, Demeret C, Vidal M, Calderwood MA, Roth FP, Falter-Braun P. A proteome-scale map of the SARS-CoV-2-human contactome. Nat Biotechnol 2023; 41:140-149. [PMID: 36217029 PMCID: PMC9849141 DOI: 10.1038/s41587-022-01475-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 08/15/2022] [Indexed: 01/22/2023]
Abstract
Understanding the mechanisms of coronavirus disease 2019 (COVID-19) disease severity to efficiently design therapies for emerging virus variants remains an urgent challenge of the ongoing pandemic. Infection and immune reactions are mediated by direct contacts between viral molecules and the host proteome, and the vast majority of these virus-host contacts (the 'contactome') have not been identified. Here, we present a systematic contactome map of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with the human host encompassing more than 200 binary virus-host and intraviral protein-protein interactions. We find that host proteins genetically associated with comorbidities of severe illness and long COVID are enriched in SARS-CoV-2 targeted network communities. Evaluating contactome-derived hypotheses, we demonstrate that viral NSP14 activates nuclear factor κB (NF-κB)-dependent transcription, even in the presence of cytokine signaling. Moreover, for several tested host proteins, genetic knock-down substantially reduces viral replication. Additionally, we show for USP25 that this effect is phenocopied by the small-molecule inhibitor AZ1. Our results connect viral proteins to human genetic architecture for COVID-19 severity and offer potential therapeutic targets.
Collapse
Affiliation(s)
- Dae-Kyum Kim
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.240614.50000 0001 2181 8635Department of Cancer Genetics and Genomics, Roswell Park Comprehensive Cancer Center, Buffalo, NY USA
| | - Benjamin Weller
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Chung-Wen Lin
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Dayag Sheykhkarimli
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Jennifer J. Knapp
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Guillaume Dugied
- grid.428999.70000 0001 2353 6535Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, Institut Pasteur, Paris, France ,grid.4444.00000 0001 2112 9282UMR3569, Centre National de la Recherche Scientifique, Paris, France ,grid.5842.b0000 0001 2171 2558Université de Paris, Paris, France
| | - Andreas Zanzoni
- grid.5399.60000 0001 2176 4817Aix-Marseille Université, Inserm, TAGC, Marseille, France
| | - Carles Pons
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute for Science and Technology, Barcelona, Spain
| | - Marie J. Tofaute
- grid.4567.00000 0004 0483 2525Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Sibusiso B. Maseko
- grid.4861.b0000 0001 0805 7253Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Kerstin Spirohn
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Florent Laval
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.4861.b0000 0001 0805 7253Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA ,grid.4861.b0000 0001 0805 7253TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium ,grid.4861.b0000 0001 0805 7253Laboratory of Molecular and Cellular Epigenetics, GIGA Institute, University of Liège, Liège, Belgium
| | - Luke Lambourne
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Nishka Kishore
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Ashyad Rayhan
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Mayra Sauer
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Veronika Young
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Hridi Halder
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Nora Marín-de la Rosa
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Oxana Pogoutse
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Alexandra Strobel
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Patrick Schwehn
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Roujia Li
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Simin T. Rothballer
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Melina Altmann
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Patricia Cassonnet
- grid.428999.70000 0001 2353 6535Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, Institut Pasteur, Paris, France ,grid.4444.00000 0001 2112 9282UMR3569, Centre National de la Recherche Scientifique, Paris, France ,grid.5842.b0000 0001 2171 2558Université de Paris, Paris, France
| | - Atina G. Coté
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Lena Elorduy Vergara
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Isaiah Hazelwood
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Betty B. Liu
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Maria Nguyen
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Ramakrishnan Pandiarajan
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Bushra Dohai
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Patricia A. Rodriguez Coloma
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Juline Poirson
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.440050.50000 0004 0408 2525Molecular Architecture of Life Program, Canadian Institute for Advanced Research (CIFAR), Toronto, ON Canada
| | - Paolo Giuliana
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA
| | - Luc Willems
- grid.4861.b0000 0001 0805 7253TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium ,grid.4861.b0000 0001 0805 7253Laboratory of Molecular and Cellular Epigenetics, GIGA Institute, University of Liège, Liège, Belgium
| | - Mikko Taipale
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.4861.b0000 0001 0805 7253Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium
| | - Yves Jacob
- grid.428999.70000 0001 2353 6535Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, Institut Pasteur, Paris, France ,grid.4444.00000 0001 2112 9282UMR3569, Centre National de la Recherche Scientifique, Paris, France ,grid.5842.b0000 0001 2171 2558Université de Paris, Paris, France
| | - Tong Hao
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - David E. Hill
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Christine Brun
- grid.5399.60000 0001 2176 4817Aix-Marseille Université, Inserm, TAGC, Marseille, France ,grid.4444.00000 0001 2112 9282CNRS, Marseille, France
| | - Jean-Claude Twizere
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.4861.b0000 0001 0805 7253Laboratory of Viral Interactomes, GIGA Institute, University of Liège, Liège, Belgium ,grid.4861.b0000 0001 0805 7253TERRA Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Daniel Krappmann
- grid.4567.00000 0004 0483 2525Research Unit Cellular Signal Integration, Institute of Molecular Toxicology and Pharmacology, Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Matthias Heinig
- grid.4567.00000 0004 0483 2525Institute of Computational Biology (ICB), Computational Health Center, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany ,grid.6936.a0000000123222966Department of Informatics, Technische Universität München, Munich, Germany
| | - Claudia Falter
- grid.4567.00000 0004 0483 2525Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
| | - Patrick Aloy
- grid.7722.00000 0001 1811 6966Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute for Science and Technology, Barcelona, Spain ,grid.425902.80000 0000 9601 989XInstitució Catalana de Recerca I Estudis Avaçats (ICREA), Barcelona, Spain
| | - Caroline Demeret
- Unité de Génétique Moléculaire des Virus à ARN, Département de Virologie, Institut Pasteur, Paris, France. .,UMR3569, Centre National de la Recherche Scientifique, Paris, France. .,Université de Paris, Paris, France.
| | - Marc Vidal
- Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA, USA. .,Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
| | - Michael A. Calderwood
- grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.38142.3c000000041936754XDepartment of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA USA ,grid.65499.370000 0001 2106 9910Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA USA
| | - Frederick P. Roth
- grid.17063.330000 0001 2157 2938Donnelly Centre for Cellular and Biomolecular Research (CCBR), University of Toronto, Toronto, Ontario Canada ,grid.17063.330000 0001 2157 2938Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada ,grid.250674.20000 0004 0626 6184Lunenfeld-Tanenbaum Research Institute (LTRI), Sinai Health System, Toronto, Ontario Canada ,grid.65499.370000 0001 2106 9910Center for Cancer Systems Biology (CCSB), Dana-Farber Cancer Institute, Boston, MA USA ,grid.17063.330000 0001 2157 2938Department of Computer Science, University of Toronto, Toronto, Ontario Canada
| | - Pascal Falter-Braun
- Institute of Network Biology (INET), Molecular Targets and Therapeutics Center (MTTC), Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany. .,Microbe-Host Interactions, Faculty of Biology, Ludwig-Maximilians-Universität (LMU) München, Planegg-Martinsried, Germany.
| |
Collapse
|
29
|
Wang S, Merits A. G3BP/Rin-Binding Motifs Inserted into Flexible Regions of nsP2 Support RNA Replication of Chikungunya Virus. J Virol 2022; 96:e0127822. [PMID: 36226983 PMCID: PMC9645214 DOI: 10.1128/jvi.01278-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/16/2022] [Indexed: 11/20/2022] Open
Abstract
Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus. In infected cells, its positive-sense RNA genome is translated into polyproteins that are subsequently processed into four nonstructural proteins (nsP1 to 4), the virus-encoded subunits of the RNA replicase. However, for RNA replication, interactions between nsPs and host proteins are also needed. These interactions are mostly mediated through the intrinsically disordered C-terminal hypervariable domain (HVD) in nsP3. Duplicate FGDF motifs in the HVD are required for interaction with mammalian RasGAP SH3-binding proteins (G3BPs) and their mosquito homolog Rin; these interactions are crucial for CHIKV RNA replication. In this study, we inactivated G3BP/Rin-binding motifs in the HVD and inserted peptides containing either native or inactivated G3BP/Rin-binding motifs into flexible regions of nsP1, nsP2, or nsP4. Insertion of native motifs into nsP1 or nsP2 but not into the C terminus of nsP4 activated CHIKV RNA replication in human cells in a G3BP-dependent manner. In mosquito cells, activation also resulted from the insertion of inactive motifs after residue 8 or 466 in nsP2; however, the effect was significantly larger when the inserted sequence contained native motifs. Nonetheless, CHIKV mutants harboring mutations in the HVD and containing insertions of native motifs in nsP2 were not viable in mosquito cells. In contrast, mutant genomes containing native motifs after residue 466 or 618 in nsP2 replicated in BHK-21 cells, with the latter mutant forming infectious progeny. Thus, the binding of G3BPs to nsP2 can support CHIKV RNA replication and restore the infectivity of viruses lacking G3BP-binding motifs in the HVD of nsP3. IMPORTANCE CHIKV is a reemerging alphavirus that has spread throughout more than 60 countries and is the causative agent of chikungunya fever. No approved drugs or vaccines are available for the treatment or prevention of CHIKV infection. CHIKV replication depends on the ability of its replicase proteins to interact with host cell factors, and a better understanding of host cell factor roles in viral infection will increase our understanding of CHIKV RNA replication and provide new strategies for viral infection attenuation. Here, we demonstrate that the motifs required for the binding of host G3BP/Rin proteins remain functional when transferred from their natural location in nsP3 to different replicase proteins and may enable mutant viruses to complete a full replication cycle. To our knowledge, this is the first demonstration of interaction motifs for crucial host factors being successfully transferred from one replicase protein to another subunit of alphavirus replicase.
Collapse
Affiliation(s)
- Sainan Wang
- Institute of Technology, University of Tartu, Tartu, Estonia
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia
| |
Collapse
|
30
|
Yang E, Huang S, Jami-Alahmadi Y, McInerney GM, Wohlschlegel JA, Li MMH. Elucidation of TRIM25 ubiquitination targets involved in diverse cellular and antiviral processes. PLoS Pathog 2022; 18:e1010743. [PMID: 36067236 PMCID: PMC9481182 DOI: 10.1371/journal.ppat.1010743] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 09/16/2022] [Accepted: 07/15/2022] [Indexed: 11/19/2022] Open
Abstract
The tripartite motif (TRIM) family of E3 ubiquitin ligases is well known for its roles in antiviral restriction and innate immunity regulation, in addition to many other cellular pathways. In particular, TRIM25-mediated ubiquitination affects both carcinogenesis and antiviral response. While individual substrates have been identified for TRIM25, it remains unclear how it regulates diverse processes. Here we characterized a mutation, R54P, critical for TRIM25 catalytic activity, which we successfully utilized to "trap" substrates. We demonstrated that TRIM25 targets proteins implicated in stress granule formation (G3BP1/2), nonsense-mediated mRNA decay (UPF1), nucleoside synthesis (NME1), and mRNA translation and stability (PABPC4). The R54P mutation abolishes TRIM25 inhibition of alphaviruses independently of the host interferon response, suggesting that this antiviral effect is a direct consequence of ubiquitination. Consistent with that, we observed diminished antiviral activity upon knockdown of several TRIM25-R54P specific interactors including NME1 and PABPC4. Our findings highlight that multiple substrates mediate the cellular and antiviral activities of TRIM25, illustrating the multi-faceted role of this ubiquitination network in modulating diverse biological processes.
Collapse
Affiliation(s)
- Emily Yang
- Molecular Biology Institute, University of California, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
| | - Serina Huang
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Gerald M. McInerney
- Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - James A. Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
| | - Melody M. H. Li
- Molecular Biology Institute, University of California, Los Angeles, California, United States of America
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, California, United States of America
- AIDS Institute, David Geffen School of Medicine, University of California, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
31
|
Neyret A, Bernard E, Aïqui-Reboul-Paviet O, Bakhache W, Eldin P, Chaloin L, Briant L. Identification of a non-canonical G3BP-binding sequence in a Mayaro virus nsP3 hypervariable domain. Front Cell Infect Microbiol 2022; 12:958176. [PMID: 36034716 PMCID: PMC9403187 DOI: 10.3389/fcimb.2022.958176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022] Open
Abstract
Ras-GTPase-activating SH3 domain-binding-proteins 1 (G3BP1) and 2 (G3BP2) are multifunctional RNA-binding proteins involved in stress granule nucleation, previously identified as essential cofactors of Old World alphaviruses. They are recruited to viral replication complexes formed by the Chikungunya virus (CHIKV), Semliki Forest virus (SFV), and Sindbis virus (SINV) via an interaction with a duplicated FGxF motif conserved in the hypervariable domain (HVD) of virus-encoded nsP3. According to mutagenesis studies, this FGxF duplication is strictly required for G3BP binding and optimal viral growth. Contrasting with this scenario, nsP3 encoded by Mayaro virus (MAYV), an arthritogenic virus grouped with Old World alphaviruses, contains a single canonical FGxF sequence. In light of this unusual feature, we questioned MAYV nsP3/G3BPs relationships. We report that G3BP1 and G3BP2 are both required for MAYV growth in human cells and bind nsP3 protein. In infected cells, they are recruited to nsP3-containing cytosolic foci and active replication complexes. Unexpectedly, deletion of the single FGxF sequence in MAYV nsP3 did not abolish these phenotypes. Using mutagenesis and in silico modeling, we identify an upstream FGAP amino acid sequence as an additional MAYV nsP3/G3BP interaction motif required for optimal viral infectivity. These results, therefore, highlight a non-conventional G3BP binding sequence in MAYV nsP3.
Collapse
|
32
|
Lasswitz L, Zapatero-Belinchón FJ, Moeller R, Hülskötter K, Laurent T, Carlson LA, Goffinet C, Simmons G, Baumgärtner W, Gerold G. The Tetraspanin CD81 Is a Host Factor for Chikungunya Virus Replication. mBio 2022; 13:e0073122. [PMID: 35612284 PMCID: PMC9239085 DOI: 10.1128/mbio.00731-22] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 05/04/2022] [Indexed: 11/20/2022] Open
Abstract
Chikungunya virus (CHIKV) is an arthritogenic reemerging virus replicating in plasma membrane-derived compartments termed "spherules." Here, we identify the human transmembrane protein CD81 as host factor required for CHIKV replication. Ablation of CD81 results in decreased CHIKV permissiveness, while overexpression enhances infection. CD81 is dispensable for virus uptake but critically required for viral genome replication. Likewise, murine CD81 is crucial for CHIKV permissiveness and is expressed in target cells such as dermal fibroblasts, muscle and liver cells. Whereas related alphaviruses, including Ross River virus (RRV), Semliki Forest virus (SFV), Sindbis virus (SINV) and Venezuelan equine encephalitis virus (VEEV), also depend on CD81 for infection, RNA viruses from other families, such as coronaviruses, replicate independently of CD81. Strikingly, the replication-enhancing function of CD81 is linked to cholesterol binding. These results define a mechanism exploited by alphaviruses to hijack the membrane microdomain-modeling protein CD81 for virus replication through interaction with cholesterol. IMPORTANCE In this study, we discover the tetraspanin CD81 as a host factor for the globally emerging chikungunya virus and related alphaviruses. We show that CD81 promotes replication of viral genomes in human and mouse cells, while virus entry into cells is independent of CD81. This provides novel insights into how alphaviruses hijack host proteins to complete their life cycle. Alphaviruses replicate at distinct sites of the plasma membrane, which are enriched in cholesterol. We found that the cholesterol-binding ability of CD81 is important for its function as an alphavirus host factor. This discovery thus broadens our understanding of the alphavirus replication process and the use of host factors to reprogram cells into virus replication factories.
Collapse
Affiliation(s)
- Lisa Lasswitz
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany
- Institute for Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hanover, Hanover, Germany
| | - Francisco J. Zapatero-Belinchón
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany
- Institute for Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hanover, Hanover, Germany
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
- Department of Clinical Microbiology, Virology, Umeå University, Umeå, Sweden
| | - Rebecca Moeller
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany
- Institute for Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hanover, Hanover, Germany
| | - Kirsten Hülskötter
- Department of Pathology, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Timothée Laurent
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Lars-Anders Carlson
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden
| | - Christine Goffinet
- Charité–Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Graham Simmons
- Vitalant Research Institute, University of California, San Francisco, California, USA
- Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - Wolfgang Baumgärtner
- Department of Pathology, University of Veterinary Medicine Hanover, Hanover, Germany
| | - Gisa Gerold
- Institute for Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research, a joint venture between the Medical School Hannover and the Helmholtz Centre for Infection Research, Hanover, Germany
- Institute for Biochemistry & Research Center for Emerging Infections and Zoonoses (RIZ), University of Veterinary Medicine Hanover, Hanover, Germany
- Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
- Department of Clinical Microbiology, Virology, Umeå University, Umeå, Sweden
| |
Collapse
|
33
|
Ge Y, Jin J, Li J, Ye M, Jin X. The roles of G3BP1 in human diseases (review). Gene X 2022; 821:146294. [PMID: 35176431 DOI: 10.1016/j.gene.2022.146294] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/24/2022] [Accepted: 02/03/2022] [Indexed: 11/04/2022] Open
Abstract
Ras-GTPase-activating protein binding protein 1 (G3BP1) is a multifunctional binding protein involved in a variety of biological functions, including cell proliferation, metastasis, apoptosis, differentiation and RNA metabolism. It has been revealed that G3BP1, as an antiviral factor, can interact with viral proteins and regulate the assembly of stress granules (SGs), which can inhibit viral replication. Furthermore, several viruses have the ability to hijack G3BP1 as a cofactor, recruiting translation initiation factors to promote viral proliferation. However, many functions of G3BP1 are associated with other diseases. In various cancers, G3BP1 is a cancer-promoting factor, which can promote the proliferation, invasion and metastasis of cancer cells. Moreover, compared with normal tissues, G3BP1 expression is higher in tumor tissues, indicating that it can be used as an indicator for cancer diagnosis. In this review, the structure of G3BP1 and the regulation of G3BP1 in multiple dimensions are described. In addition, the effects and potential mechanisms of G3BP1 on various carcinomas, viral infections, nervous system diseases and cardiovascular diseases are elucidated, which may provide a direction for clinical applications of G3BP1 in the future.
Collapse
Affiliation(s)
- Yidong Ge
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jiabei Jin
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Jinyun Li
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China
| | - Meng Ye
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China.
| | - Xiaofeng Jin
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo 315020, China; Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathphysiology, Medical School of Ningbo University, Ningbo 315211, China.
| |
Collapse
|
34
|
Abstract
Alphaviruses are positive-strand RNA viruses, typically transmitted by mosquitoes between vertebrate hosts. They encode four essential replication proteins, the non-structural proteins nsP1-4, which possess the enzymatic activities of RNA capping, RNA helicase, site-specific protease, ADP-ribosyl removal and RNA polymerase. Alphaviruses have been key models in the study of membrane-associated RNA replication, which is a conserved feature among the positive-strand RNA viruses of animals and plants. We review new structural and functional information on the nsPs and their interaction with host proteins and membranes, as well as with viral RNA sequences. The dodecameric ring structure of nsP1 is likely to be one of the evolutionary innovations that facilitated the success of the progenitors of current positive-strand RNA viruses.
Collapse
Affiliation(s)
- Tero Ahola
- Department of Microbiology, Faculty of Agriculture and Forestry, University of Helsinki, Helsinki, Finland
| | - Gerald McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Andres Merits
- Institute of Technology, University of Tartu, Tartu, Estonia.
| |
Collapse
|
35
|
Kang W, Wang Y, Yang W, Zhang J, Zheng H, Li D. Research Progress on the Structure and Function of G3BP. Front Immunol 2021; 12:718548. [PMID: 34526993 PMCID: PMC8435845 DOI: 10.3389/fimmu.2021.718548] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 08/10/2021] [Indexed: 01/10/2023] Open
Abstract
Ras-GTPase-activating protein (SH3 domain)-binding protein (G3BP) is an RNA binding protein. G3BP is a key component of stress granules (SGs) and can interact with many host proteins to regulate the expression of SGs. As an antiviral factor, G3BP can interact with viral proteins to regulate the assembly of SGs and thus exert antiviral effects. However, many viruses can also use G3BP as a proximal factor and recruit translation initiation factors to promote viral proliferation. G3BP regulates mRNA translation and attenuation to regulate gene expression; therefore, it is closely related to diseases, such as cancer, embryonic death, arteriosclerosis, and neurodevelopmental disorders. This review discusses the important discoveries and developments related G3BP in the biological field over the past 20 years, which includes the formation of SGs, interaction with viruses, stability of RNA, and disease progression.
Collapse
Affiliation(s)
- Weifang Kang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Yue Wang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wenping Yang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Jing Zhang
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Haixue Zheng
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Dan Li
- State Key Laboratory of Veterinary Etiological Biology and OIE/National Foot and Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
36
|
Brown SL, Garrison DJ, May JP. Phase separation of a plant virus movement protein and cellular factors support virus-host interactions. PLoS Pathog 2021; 17:e1009622. [PMID: 34543360 PMCID: PMC8483311 DOI: 10.1371/journal.ppat.1009622] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/30/2021] [Accepted: 09/13/2021] [Indexed: 12/27/2022] Open
Abstract
Both cellular and viral proteins can undergo phase separation and form membraneless compartments that concentrate biomolecules. The p26 movement protein from single-stranded, positive-sense Pea enation mosaic virus 2 (PEMV2) separates into a dense phase in nucleoli where p26 and related orthologues must interact with fibrillarin (Fib2) as a pre-requisite for systemic virus movement. Using in vitro assays, viral ribonucleoprotein complexes containing p26, Fib2, and PEMV2 genomic RNAs formed droplets that may provide the basis for self-assembly in planta. Mutating basic p26 residues (R/K-G) blocked droplet formation and partitioning into Fib2 droplets or the nucleolus and prevented systemic movement of a Tobacco mosaic virus (TMV) vector in Nicotiana benthamiana. Mutating acidic residues (D/E-G) reduced droplet formation in vitro, increased nucleolar retention 6.5-fold, and prevented systemic movement of TMV, thus demonstrating that p26 requires electrostatic interactions for droplet formation and charged residues are critical for nucleolar trafficking and virus movement. p26 readily partitioned into stress granules (SGs), which are membraneless compartments that assemble by clustering of the RNA binding protein G3BP following stress. G3BP is upregulated during PEMV2 infection and over-expression of G3BP restricted PEMV2 RNA accumulation >20-fold. Deletion of the NTF2 domain that is required for G3BP condensation restored PEMV2 RNA accumulation >4-fold, demonstrating that phase separation enhances G3BP antiviral activity. These results indicate that p26 partitions into membraneless compartments with either proviral (Fib2) or antiviral (G3BP) factors.
Collapse
Affiliation(s)
- Shelby L. Brown
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Dana J. Garrison
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| | - Jared P. May
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, Missouri, United States of America
| |
Collapse
|
37
|
Kril V, Aïqui-Reboul-Paviet O, Briant L, Amara A. New Insights into Chikungunya Virus Infection and Pathogenesis. Annu Rev Virol 2021; 8:327-347. [PMID: 34255544 DOI: 10.1146/annurev-virology-091919-102021] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Chikungunya virus (CHIKV) is a re-emerging mosquito-borne alphavirus responsible for major outbreaks of disease since 2004 in the Indian Ocean islands, South east Asia, and the Americas. CHIKV causes debilitating musculoskeletal disorders in humans that are characterized by fever, rash, polyarthralgia, and myalgia. The disease is often self-limiting and nonlethal; however, some patients experience atypical or severe clinical manifestations, as well as a chronic rheumatic syndrome. Unfortunately, no efficient antivirals against CHIKV infection are available so far, highlighting the importance of deepening our knowledge of CHIKV host cell interactions and viral replication strategies. In this review, we discuss recent breakthroughs in the molecular mechanisms that regulate CHIKV infection and lay down the foundations to understand viral pathogenesis. We describe the role of the recently identified host factors co-opted by the virus for infection and pathogenesis, and emphasize the importance of CHIKV nonstructural proteins in both replication complex assembly and host immune response evasion. Expected final online publication date for the Annual Review of Virology, Volume 8 is September 2021. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Vasiliya Kril
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| | - Olivier Aïqui-Reboul-Paviet
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Laurence Briant
- RNA Viruses and Metabolism Team, CNRS UMR 9004, Institut de Recherche en Infectiologie de Montpellier, University of Montpellier, 34293 Montpellier, France;
| | - Ali Amara
- Biology of Emerging Virus Team, INSERM U944, CNRS UMR 7212, Institut de Recherche Saint-Louis, Université de Paris, Hôpital Saint-Louis, 75010 Paris, France;
| |
Collapse
|
38
|
Arabidopsis thaliana G3BP Ortholog Rescues Mammalian Stress Granule Phenotype across Kingdoms. Int J Mol Sci 2021; 22:ijms22126287. [PMID: 34208100 PMCID: PMC8230867 DOI: 10.3390/ijms22126287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/08/2021] [Accepted: 06/08/2021] [Indexed: 11/16/2022] Open
Abstract
Stress granules (SGs) are dynamic RNA–protein complexes localized in the cytoplasm that rapidly form under stress conditions and disperse when normal conditions are restored. The formation of SGs depends on the Ras-GAP SH3 domain-binding protein (G3BP). Formations, interactions and functions of plant and human SGs are strikingly similar, suggesting a conserved mechanism. However, functional analyses of plant G3BPs are missing. Thus, members of the Arabidopsis thaliana G3BP (AtG3BP) protein family were investigated in a complementation assay in a human G3BP knock-out cell line. It was shown that two out of seven AtG3BPs were able to complement the function of their human homolog. GFP-AtG3BP fusion proteins co-localized with human SG marker proteins Caprin-1 and eIF4G1 and restored SG formation in G3BP double KO cells. Interaction between AtG3BP-1 and -7 and known human G3BP interaction partners such as Caprin-1 and USP10 was also demonstrated by co-immunoprecipitation. In addition, an RG/RGG domain exchange from Arabidopsis G3BP into the human G3BP background showed the ability for complementation. In summary, our results support a conserved mechanism of SG function over the kingdoms, which will help to further elucidate the biological function of the Arabidopsis G3BP protein family.
Collapse
|
39
|
Abulfaraj AA, Hirt H, Rayapuram N. G3BPs in Plant Stress. FRONTIERS IN PLANT SCIENCE 2021; 12:680710. [PMID: 34177995 PMCID: PMC8222905 DOI: 10.3389/fpls.2021.680710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/14/2021] [Indexed: 05/24/2023]
Abstract
The sessile nature of plants enforces highly adaptable strategies to adapt to different environmental stresses. Plants respond to these stresses by a massive reprogramming of mRNA metabolism. Balancing of mRNA fates, including translation, sequestration, and decay is essential for plants to not only coordinate growth and development but also to combat biotic and abiotic environmental stresses. RNA stress granules (SGs) and processing bodies (P bodies) synchronize mRNA metabolism for optimum functioning of an organism. SGs are evolutionarily conserved cytoplasmic localized RNA-protein storage sites that are formed in response to adverse conditions, harboring mostly but not always translationally inactive mRNAs. SGs disassemble and release mRNAs into a translationally active form upon stress relief. RasGAP SH3 domain binding proteins (G3BPs or Rasputins) are "scaffolds" for the assembly and stability of SGs, which coordinate receptor mediated signal transduction with RNA metabolism. The role of G3BPs in the formation of SGs is well established in mammals, but G3BPs in plants are poorly characterized. In this review, we discuss recent findings of the dynamics and functions of plant G3BPs in response to environmental stresses and speculate on possible mechanisms such as transcription and post-translational modifications that might regulate the function of this important family of proteins.
Collapse
Affiliation(s)
- Aala A. Abulfaraj
- Department of Biological Sciences, Science and Arts College, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Heribert Hirt
- King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Max Perutz Laboratories, University of Vienna, Vienna, Austria
| | - Naganand Rayapuram
- King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| |
Collapse
|
40
|
Contu L, Balistreri G, Domanski M, Uldry AC, Mühlemann O. Characterisation of the Semliki Forest Virus-host cell interactome reveals the viral capsid protein as an inhibitor of nonsense-mediated mRNA decay. PLoS Pathog 2021; 17:e1009603. [PMID: 34019569 PMCID: PMC8174725 DOI: 10.1371/journal.ppat.1009603] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 06/03/2021] [Accepted: 05/03/2021] [Indexed: 01/08/2023] Open
Abstract
The positive-sense, single-stranded RNA alphaviruses pose a potential epidemic threat. Understanding the complex interactions between the viral and the host cell proteins is crucial for elucidating the mechanisms underlying successful virus replication strategies and for developing specific antiviral interventions. Here we present the first comprehensive protein-protein interaction map between the proteins of Semliki Forest Virus (SFV), a mosquito-borne member of the alphaviruses, and host cell proteins. Among the many identified cellular interactors of SFV proteins, the enrichment of factors involved in translation and nonsense-mediated mRNA decay (NMD) was striking, reflecting the virus' hijacking of the translation machinery and indicating viral countermeasures for escaping NMD by inhibiting NMD at later time points during the infectious cycle. In addition to observing a general inhibition of NMD about 4 hours post infection, we also demonstrate that transient expression of the SFV capsid protein is sufficient to inhibit NMD in cells, suggesting that the massive production of capsid protein during the SFV reproduction cycle is responsible for NMD inhibition.
Collapse
Affiliation(s)
- Lara Contu
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Giuseppe Balistreri
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
- Queensland Brain Institute, The University of Queensland, Brisbane, Australia
| | - Michal Domanski
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
| | - Anne-Christine Uldry
- Proteomics & Mass Spectrometry Core Facility, Department for BioMedical Research, University of Bern, Bern, Switzerland
| | - Oliver Mühlemann
- Department of Chemistry and Biochemistry, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
41
|
Embarc-Buh A, Francisco-Velilla R, Martinez-Salas E. RNA-Binding Proteins at the Host-Pathogen Interface Targeting Viral Regulatory Elements. Viruses 2021; 13:952. [PMID: 34064059 PMCID: PMC8224014 DOI: 10.3390/v13060952] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 05/10/2021] [Accepted: 05/17/2021] [Indexed: 12/16/2022] Open
Abstract
Viral RNAs contain the information needed to synthesize their own proteins, to replicate, and to spread to susceptible cells. However, due to their reduced coding capacity RNA viruses rely on host cells to complete their multiplication cycle. This is largely achieved by the concerted action of regulatory structural elements on viral RNAs and a subset of host proteins, whose dedicated function across all stages of the infection steps is critical to complete the viral cycle. Importantly, not only the RNA sequence but also the RNA architecture imposed by the presence of specific structural domains mediates the interaction with host RNA-binding proteins (RBPs), ultimately affecting virus multiplication and spreading. In marked difference with other biological systems, the genome of positive strand RNA viruses is also the mRNA. Here we focus on distinct types of positive strand RNA viruses that differ in the regulatory elements used to promote translation of the viral RNA, as well as in the mechanisms used to evade the series of events connected to antiviral response, including translation shutoff induced in infected cells, assembly of stress granules, and trafficking stress.
Collapse
Affiliation(s)
| | | | - Encarnacion Martinez-Salas
- Centro de Biología Molecular Severo Ochoa, CSIC-UAM, Nicolás Cabrera 1, 28049 Madrid, Spain; (A.E.-B.); (R.F.-V.)
| |
Collapse
|
42
|
The Paradoxes of Viral mRNA Translation during Mammalian Orthoreovirus Infection. Viruses 2021; 13:v13020275. [PMID: 33670092 PMCID: PMC7916891 DOI: 10.3390/v13020275] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 02/06/2023] Open
Abstract
De novo viral protein synthesis following entry into host cells is essential for viral replication. As a consequence, viruses have evolved mechanisms to engage the host translational machinery while at the same time avoiding or counteracting host defenses that act to repress translation. Mammalian orthoreoviruses are dsRNA-containing viruses whose mRNAs were used as models for early investigations into the mechanisms that underpin the recognition and engagement of eukaryotic mRNAs by host cell ribosomes. However, there remain many unanswered questions and paradoxes regarding translation of reoviral mRNAs in the context of infection. This review summarizes the current state of knowledge about reovirus translation, identifies key unanswered questions, and proposes possible pathways toward a better understanding of reovirus translation.
Collapse
|
43
|
Liu W, Wang S, Lin B, Zhang W, Ji G. Applications of CRISPR/Cas9 in the research of malignant musculoskeletal tumors. BMC Musculoskelet Disord 2021; 22:149. [PMID: 33546657 PMCID: PMC7866880 DOI: 10.1186/s12891-021-04020-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 01/26/2021] [Indexed: 12/05/2022] Open
Abstract
Background Malignant tumors of the musculoskeletal system, especially osteosarcoma, Ewing sarcoma and rhabdomyosarcoma, pose a major threat to the lives and health of adolescents and children. Current treatments for musculoskeletal tumors mainly include surgery, chemotherapy, and radiotherapy. The problems of chemotherapy resistance, poor long-term outcome of radiotherapy, and the inherent toxicity and side effects of chemical drugs make it extremely urgent to seek new treatment strategies. Main text As a potent gene editing tool, the rapid development of CRISPR/Cas9 technology in recent years has prompted scientists to apply it to the study of musculoskeletal tumors. This review summarizes the application of CRISPR/Cas9 technology for the treatment of malignant musculoskeletal tumors, focusing on its essential role in the field of basic research. Conclusion CRISPR, has demonstrated strong efficacy in targeting tumor-related genes, and its future application in the clinical treatment of musculoskeletal tumors is promising.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Shubin Wang
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Binhui Lin
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Guangrong Ji
- Department of Orthopaedics, Xiang'an Hospital, School of Medicine, Xiamen University, No. 2000 East Xiang'an Road, Xiang'an District, Xiamen, 361102, China.
| |
Collapse
|
44
|
May JP, Simon AE. Targeting of viral RNAs by Upf1-mediated RNA decay pathways. Curr Opin Virol 2020; 47:1-8. [PMID: 33341474 DOI: 10.1016/j.coviro.2020.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 12/25/2022]
Abstract
Viral RNAs are susceptible to co-translational RNA decay pathways mediated by the RNA helicase Upstream frameshift 1 (Upf1). Upf1 is a key component in nonsense-mediated decay (NMD), Staufen1-mediated mRNA decay (SMD), and structure-mediated RNA decay (SRD) pathways, among others. Diverse families of viruses have features that predispose them to Upf1 targeting, but have evolved means to escape decay through the action of cis-acting or trans-acting viral factors. Studies aimed at understanding how viruses are subjected to and circumvent NMD have increased our understanding of NMD target selection of host mRNAs. This review focuses on the knowledge gained from studying NMD in viral systems as well as related Upf1-dependent pathways and how these pathways restrict virus replication.
Collapse
Affiliation(s)
- Jared P May
- Department of Cell and Molecular Biology and Biochemistry, School of Biological and Chemical Sciences, University of Missouri-Kansas City, Kansas City, MO 64110, USA.
| | - Anne E Simon
- Department of Cell Biology and Molecular Genetics, University of Maryland - College Park, College Park, MD, USA.
| |
Collapse
|
45
|
Structural and Functional Characterization of Host FHL1 Protein Interaction with Hypervariable Domain of Chikungunya Virus nsP3 Protein. J Virol 2020; 95:JVI.01672-20. [PMID: 33055253 DOI: 10.1128/jvi.01672-20] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 10/10/2020] [Indexed: 11/20/2022] Open
Abstract
Decades of insufficient control have resulted in unprecedented spread of chikungunya virus (CHIKV) around the globe, and millions have suffered from the highly debilitating disease. Nevertheless, the current understanding of CHIKV-host interactions and adaptability of the virus to replication in mosquitoes and mammalian hosts is still elusive. Our new study shows that four-and-a-half LIM domain protein (FHL1) is one of the host factors that interact with the hypervariable domain (HVD) of CHIKV nsP3. Unlike G3BPs, FHL1 is not a prerequisite of CHIKV replication, and many commonly used cell lines do not express FHL1. However, its expression has a detectable stimulatory effect(s) on CHIKV replication, and Fhl1 knockout (KO) cell lines demonstrate slower infection spread. Nuclear magnetic resonance (NMR)-based studies revealed that the binding site of FHL1 in CHIKV nsP3 HVD overlaps that of another proviral host factor, CD2AP. The structural data also demonstrated that FHL1-HVD interaction is mostly determined by the LIM1 domain of FHL1. However, it does not mirror binding of the entire protein, suggesting that other LIM domains are involved. In agreement with previously published data, our biological experiments showed that interactions of CHIKV HVD with CD2AP and FHL1 have additive effects on the efficiency of CHIKV replication. This study shows that CHIKV mutants with extensive modifications of FHL1- or both FHL1- and CD2AP-binding sites remain viable and develop spreading infection in multiple cell types. Our study also demonstrated that other members of the FHL family can bind to CHIKV HVD and thus may be involved in viral replication.IMPORTANCE Replication of chikungunya virus (CHIKV) is determined by a wide range of host factors. Previously, we have demonstrated that the hypervariable domain (HVD) of CHIKV nsP3 contains linear motifs that recruit defined families of host proteins into formation of functional viral replication complexes. Now, using NMR-based structural and biological approaches, we have characterized the binding site of the cellular FHL1 protein in CHIKV HVD and defined the biological significance of this interaction. In contrast to previously described binding of G3BP to CHIKV HVD, the FHL1-HVD interaction was found to not be a prerequisite of viral replication. However, the presence of FHL1 has a stimulatory effect on CHIKV infectivity and, subsequently, the infection spread. FHL1 and CD2AP proteins were found to have overlapping binding sites in CHIKV HVD and additive proviral functions. Elimination of the FHL1-binding site in the nsP3 HVD can be used for the development of stable, attenuated vaccine candidates.
Collapse
|
46
|
Legrand N, Dixon DA, Sobolewski C. Stress granules in colorectal cancer: Current knowledge and potential therapeutic applications. World J Gastroenterol 2020; 26:5223-5247. [PMID: 32994684 PMCID: PMC7504244 DOI: 10.3748/wjg.v26.i35.5223] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/12/2020] [Accepted: 09/03/2020] [Indexed: 02/06/2023] Open
Abstract
Stress granules (SGs) represent important non-membrane cytoplasmic compartments, involved in cellular adaptation to various stressful conditions (e.g., hypoxia, nutrient deprivation, oxidative stress). These granules contain several scaffold proteins and RNA-binding proteins, which bind to mRNAs and keep them translationally silent while protecting them from harmful conditions. Although the role of SGs in cancer development is still poorly known and vary between cancer types, increasing evidence indicate that the expression and/or the activity of several key SGs components are deregulated in colorectal tumors but also in pre-neoplastic conditions (e.g., inflammatory bowel disease), thus suggesting a potential role in the onset of colorectal cancer (CRC). It is therefore believed that SGs formation importantly contributes to various steps of colorectal tumorigenesis but also in chemoresistance. As CRC is the third most frequent cancer and one of the leading causes of cancer mortality worldwide, development of new therapeutic targets is needed to offset the development of chemoresistance and formation of metastasis. Abolishing SGs assembly may therefore represent an appealing therapeutic strategy to re-sensitize colon cancer cells to anti-cancer chemotherapies. In this review, we summarize the current knowledge on SGs in colorectal cancer and the potential therapeutic strategies that could be employed to target them.
Collapse
Affiliation(s)
- Noémie Legrand
- Department of Medicine, Faculty of Medicine, University of Geneva, Geneva CH-1211, Switzerland
| | - Dan A Dixon
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, and University of Kansas Cancer Center, Lawrence, KS 66045, United States
| | - Cyril Sobolewski
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva CH-1211, Switzerland
| |
Collapse
|
47
|
Erasmus JH, Archer J, Fuerte-Stone J, Khandhar AP, Voigt E, Granger B, Bombardi RG, Govero J, Tan Q, Durnell LA, Coler RN, Diamond MS, Crowe JE, Reed SG, Thackray LB, Carnahan RH, Van Hoeven N. Intramuscular Delivery of Replicon RNA Encoding ZIKV-117 Human Monoclonal Antibody Protects against Zika Virus Infection. Mol Ther Methods Clin Dev 2020; 18:402-414. [PMID: 32695842 PMCID: PMC7363633 DOI: 10.1016/j.omtm.2020.06.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 05/15/2020] [Indexed: 12/31/2022]
Abstract
Monoclonal antibody (mAb) therapeutics are an effective modality for the treatment of infectious, autoimmune, and cancer-related diseases. However, the discovery, development, and manufacturing processes are complex, resource-consuming activities that preclude the rapid deployment of mAbs in outbreaks of emerging infectious diseases. Given recent advances in nucleic acid delivery technology, it is now possible to deliver exogenous mRNA encoding mAbs for in situ expression following intravenous (i.v.) infusion of lipid nanoparticle-encapsulated mRNA. However, the requirement for i.v. administration limits the application to settings where infusion is an option, increasing the cost of treatment. As an alternative strategy, and to enable intramuscular (IM) administration of mRNA-encoded mAbs, we describe a nanostructured lipid carrier for delivery of an alphavirus replicon encoding a previously described highly neutralizing human mAb, ZIKV-117. Using a lethal Zika virus challenge model in mice, our studies show robust protection following alphavirus-driven expression of ZIKV-117 mRNA when given by IM administration as pre-exposure prophylaxis or post-exposure therapy.
Collapse
Affiliation(s)
- Jesse H. Erasmus
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
- HDT Biocorp, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Jacob Archer
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
- HDT Biocorp, Seattle, WA, USA
- Department of Microbiology, University of Washington, Seattle, WA, USA
| | - Jasmine Fuerte-Stone
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
| | - Amit P. Khandhar
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
- HDT Biocorp, Seattle, WA, USA
| | - Emily Voigt
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
| | - Brian Granger
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
| | - Robin G. Bombardi
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 27232, USA
| | - Jennifer Govero
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Qing Tan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lorellin A. Durnell
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rhea N. Coler
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
| | - Michael S. Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James E. Crowe
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 27232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 27232, USA
- Department of Pathology Microbiology & Immunology, Vanderbilt University Medical Center, Nashville, TN 27232, USA
| | - Steven G. Reed
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
- HDT Biocorp, Seattle, WA, USA
| | - Larissa B. Thackray
- Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Robert H. Carnahan
- The Vanderbilt Vaccine Center, Vanderbilt University Medical Center, Nashville, TN 27232, USA
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN 27232, USA
| | - Neal Van Hoeven
- Pre-Clinical Vaccine Development, Infectious Disease Research Institute, Seattle, WA, USA
| |
Collapse
|
48
|
Zhao D, Li J, Wang Y, Li X, Gao L, Cao H, Zheng SJ. Critical role for G3BP1 in infectious bursal disease virus (IBDV)-induced stress granule formation and viral replication. Vet Microbiol 2020; 248:108806. [PMID: 32827928 DOI: 10.1016/j.vetmic.2020.108806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 07/18/2020] [Indexed: 12/24/2022]
Abstract
Stress granules (SGs), complexes for mRNA storage, are formed in host cellular response to stress stimuli and play an important role in innate immune response. GTPase-activating protein (SH3 domain)-binding protein 1 (G3BP1) is a key component of SGs. However, whether IBDV infection induces SG formation in host cells and what role of G3BP1 plays in this process are unclear. We report here that IBDV infection initiated typical stress granule formation and enhanced G3BP1 expression in DF-1 cells. Our data show that knockdown of G3BP1 by RNAi markedly inhibited IBDV-induced SG formation and viral replication in DF-1 cells. Conversely, ectopic expression of G3BP1 enhanced IBDV-induced SG formation and significantly promoted IBDV replication in host cells. Thus, G3BP1 plays a critical role in IBDV-induced SG formation and viral replication, providing an important clue to elucidating how IBDV employs cellular SGs for its own benefits.
Collapse
Affiliation(s)
- Dianzheng Zhao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Jiaxin Li
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Yongqiang Wang
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Xiaoqi Li
- College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Li Gao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Hong Cao
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China
| | - Shijun J Zheng
- Key Laboratory of Animal Epidemiology of the Ministry of Agriculture, College of Veterinary Medicine, China Agricultural University, Beijing 100193, China; College of Veterinary Medicine, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
49
|
Activation of the PI3K-AKT Pathway by Old World Alphaviruses. Cells 2020; 9:cells9040970. [PMID: 32326388 PMCID: PMC7226951 DOI: 10.3390/cells9040970] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Alphaviruses can infect a broad range of vertebrate hosts, including birds, horses, primates, and humans, in which infection can lead to rash, fever, encephalitis, and arthralgia or arthritis. They are most often transmitted by mosquitoes in which they establish persistent, asymptomatic infections. Currently, there are no vaccines or antiviral therapies for any alphavirus. Several Old World alphaviruses, including Semliki Forest virus, Ross River virus and chikungunya virus, activate or hyperactivate the phosphatidylinositol-3-kinase (PI3K)-AKT pathway in vertebrate cells, potentially influencing many cellular processes, including survival, proliferation, metabolism and autophagy. Inhibition of PI3K or AKT inhibits replication of several alphaviruses either in vitro or in vivo, indicating the importance for viral replication. In this review, we discuss what is known about the mechanism(s) of activation of the pathway during infection and describe those effects of PI3K-AKT activation which could be of advantage to the alphaviruses. Such knowledge may be useful for the identification and development of therapies.
Collapse
|
50
|
Sensitivity of Alphaviruses to G3BP Deletion Correlates with Efficiency of Replicase Polyprotein Processing. J Virol 2020; 94:JVI.01681-19. [PMID: 31941782 DOI: 10.1128/jvi.01681-19] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 01/02/2020] [Indexed: 11/20/2022] Open
Abstract
We present a comprehensive overview of the dependency of several Old World alphaviruses for the host protein G3BP. Based on their replication ability in G3BP-deleted cells, Old World alphaviruses can be categorized into two groups, being either resistant or sensitive to G3BP deletion. We observed that all sensitive viruses have an Arg residue at the P4 position of the cleavage site between the nonstructural protein P1 (nsP1) and nsP2 regions of the replicase precursor polyprotein (1/2 site), while a different residue is found at this site in viruses resistant to G3BP deletion. Swapping this residue between resistant and sensitive viruses also switches the G3BP deletion sensitivity. In the absence of G3BP, chikungunya virus (CHIKV) replication is at the limit of detection. The P4 Arg-to-His substitution partially rescues this defect. The P4 residue of the 1/2 site is known to play a regulatory role during processing at this site, and we found that if processing is blocked, the influence of the P4 residue on the sensitivity to G3BP deletion is abolished. Immunofluorescence experiments with CHIKV replicase with manipulated processing indicate that the synthesis of double-stranded RNA is defective in the absence of G3BP and suggest a role of G3BP during negative-strand RNA synthesis. This study provides a functional link between the host protein G3BP and the P4 residue of the 1/2 site for viral RNA replication of Old World alphaviruses. While this suggests a link between G3BP proteins and viral replicase polyprotein processing, we propose that G3BP proteins do not have a regulatory role during polyprotein processing.IMPORTANCE Old World alphaviruses comprise several medically relevant viruses, including chikungunya virus and Ross River virus. Recurrent outbreaks and the lack of antivirals and vaccines demand ongoing research to fight the emergence of these infectious diseases. In this context, a thorough investigation of virus-host interactions is critical. Here, we highlight the importance of the host protein G3BP for several Old World alphaviruses. Our data strongly suggest that G3BP plays a crucial role for the activity of the viral replicase and, thus, the amplification of the viral RNA genome. To our knowledge, the present work is the first to provide a functional link between the regulation of viral polyprotein processing and RNA replication and a host factor for alphaviruses. Moreover, the results of this study raise several questions about the fundamental regulatory mechanisms that dictate the activity of the viral replicase, thereby paving the way for future studies.
Collapse
|