Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1346 2023-08-23 06:40:31 |
2 format correct Meta information modification 1346 2023-08-23 07:40:46 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Tam, D.; Lorenzo-Leal, A.C.; Hernández, L.R.; Bach, H. NSPs Present in the SARS-CoV-2 Genome. Encyclopedia. Available online: https://encyclopedia.pub/entry/48349 (accessed on 01 July 2024).
Tam D, Lorenzo-Leal AC, Hernández LR, Bach H. NSPs Present in the SARS-CoV-2 Genome. Encyclopedia. Available at: https://encyclopedia.pub/entry/48349. Accessed July 01, 2024.
Tam, Donald, Ana C. Lorenzo-Leal, Luis Ricardo Hernández, Horacio Bach. "NSPs Present in the SARS-CoV-2 Genome" Encyclopedia, https://encyclopedia.pub/entry/48349 (accessed July 01, 2024).
Tam, D., Lorenzo-Leal, A.C., Hernández, L.R., & Bach, H. (2023, August 23). NSPs Present in the SARS-CoV-2 Genome. In Encyclopedia. https://encyclopedia.pub/entry/48349
Tam, Donald, et al. "NSPs Present in the SARS-CoV-2 Genome." Encyclopedia. Web. 23 August, 2023.
NSPs Present in the SARS-CoV-2 Genome
Edit

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped respiratory β coronavirus that causes coronavirus disease (COVID-19), leading to a deadly pandemic that has claimed millions of lives worldwide. Like other coronaviruses, the SARS-CoV-2 genome also codes for non-structural proteins (NSPs). These NSPs are found within open reading frame 1a (ORF1a) and open reading frame 1ab (ORF1ab) of the SARS-CoV-2 genome and encode NSP1 to NSP11 and NSP12 to NSP16, respectively. 

SARS-CoV-2 non-structural protein virulence factors

1. NSP1

NSP1 inhibits the translation of host proteins by binding to the mRNA binding pocket of the 40S ribosome via its C-terminal domain [1][2]. This prevents the synthesis of important proteins involved in the innate immune response, including IFN-b, IFN-g1, IL-8, and retinoic acid-inducible gene 1, which trigger apoptosis [3][4]. The N-terminal region of NSP1 upregulates the translation of viral mRNAs by binding to the 5′ UTR stem-loop 1 region [3][5]. NSP1 recruits exonucleases that cleave host mRNA and uncapped mRNA transcripts at their internal ribosome entry sites [6]. NSP1 has also been shown to block host mRNA transcripts from exiting the nucleus by binding to the NXF1-NXT1 heterodimer docking complex involved in the nuclear export of host mRNAs through the nuclear pore complex [7]. Altogether, this virulence factor incapacitates the ability of the host cell to mount an innate immune response while enhancing viral replication [3].

2. NSP2

Attempts to determine the function of NSP2 in the host were performed by in situ biotinylation [8]. However, although results showed that NSP2 interacted with the host proteins prohibitin 1 (PHB1) and prohibitin 2 (PHB2), a validation of the results using immunoblotting of the immunoprecipitated proteins with an anti-NSP2 antibody (not available) could not be proven. PHB 1 and 2 are involved in different cell activities, such as the morphology of mitochondria [9] and transcription factor regulation [10]. Interestingly, PHB1 has been reported as a receptor facilitating the entry of Chikungunya and Dengue 2 viruses [11][12].

3. NSP3

This papain-like protease (PLpro) protein cleaves the N-terminal region of pp1a to release NSP1 to NSP3 from the polyprotein [13]. It is also involved in the cleavage of proteinaceous post-translational modifications on host proteins involved in the innate antiviral responses, further dampening the immune response Field [13]. PLpro can also regulate the innate immune response by cleaving ubiquitin-like interferon-stimulated gene 15 (ISG15) from interferon response factor 3 (IRF3) to inhibit the IFN I pathway [13][14][15].

4. NSP4

NSP4 is a transmembrane protein that forms part of the viral replication complex within the virally infected cell [16]. It is involved in modifying the ER membranes to form DMV in conjunction with NSP6 to compartmentalize viral replication and viral assembly, acting as an additional barrier against intracellular antiviral responses [16].

5. NSP5

NSP5 is a 3-cysteine-like protease (3Clpro), the main viral protease of SARS-CoV-2 [17]. It cleaves at 11 specific sites after glutamine residues along pp1a and pp1ab to release NSP4 to NSP16 in their intermediate or mature forms [17][18].

6. NSP6

NSP6 is a transmembrane protein that interacts with NSP3 and NSP4 to induce the formation of endoplasmic reticulum (ER)-derived autophagosomes and DMVs [19]. These autophagosomes and DMVs serve as a site for replication transcription complexes (RTCs) to form, which are necessary for viral replication [16][19]. It is also involved in modulating ER stress by binding to the host sigma receptor that is involved in limiting the production of autophagosomes and autolysosomes to disrupt viral replication [20]. NSP6 has also been implicated in activating host autophagy pathways through the omegasome pathway to promote the assembly of viral replicase proteins and the degradation of immunomodulatory proteins [19]. NSP6 also directly inhibits the cleavage-mediated activation of vacuolar proton pump components such as ATP6AP1 to impair lysosomal acidification and trigger inflammasome-mediated pyroptosis [21].

7. NSP7

NSP7 is an accessory factor protein that forms a heterotetramer with NSP8 and NSP12 to assemble the RTC needed for RNA synthesis [22].

8. NSP8

NSP8 is another accessory factor protein that forms a homodimer with itself and forms a part of the larger RTC complex alongside NSP7 and NSP12, which is necessary for RNA synthesis [22]. It has also been shown to play a role in suppressing IFN-a signaling pathways to further progress disease pathogenesis [23].

9. NSP9

NSP9 is a single-stranded RNA-binding dimeric replicase protein that binds to and activates NSP8 as a cofactor [24]. NSP9 also plays a significant role in RNA synthesis by acting as a primer for NSP12 after being modified by the NiRAN domain of NSP12 by transferring a nucleotide monophosphate to the N-terminus of NSP9 [24][25]. In the host cell, NSP9 localizes around the ER membrane, which associates with the nucleoporin 62 (NUP62), a structural protein that forms part of the nuclear pore complex, to prevent the import of p65 into the nucleus [26]. This mislocalization of p65 leads to decreased NF-kb-regulated gene expression, resulting in a dampened immune response [26].

10. NSP10

NSP10 is a growth-factor-like protein with two zinc-binding motifs that allow it to act as a cofactor for NSP16 to aid in capping viral mRNA transcripts [27]. This capping of viral mRNA transcripts increases the rate of replication by the RTC and allows the transcript to evade the host RNases that cleave uncapped mRNA transcripts [28]. NSP10 also acts as a costimulatory factor to NSP14 to enhance its exonuclease activity to remove mismatched bases, and it also allosterically activates NSP14 to aid in mRNA capping [28][29].

11. NSP11

NSP11 is a short protein that shares identical homology to the first segment of NSP12 [30]. It is an intrinsically disordered protein that sits near the junction of pp1a and pp1b. It may be involved in host cytosolic membrane affinity interactions [30]. It is also suggested that NSP11 may play a role in ribosomal frameshifting to adjust the reading frame by −1 as the reading frame for the NSPs in pp1a and pp1ab are different [30]. NSP11 is also shown to regulate endoribonuclease activity and is necessary for viral replication [30].

12. NSP12

NSP12 is an RNA-dependent RNA polymerase (RdRP) protein that acts as the core of the RTC, which binds one NSP7 and two NSP8 molecules to stabilize the RNA binding region [31], allowing viral replication to proceed [32]. The primary RNA polymerase produces nascent ssRNA from the viral RNA template [33]. NSP12 also possesses an N-terminal nidovirus RdRP-associated nucleotidyltransferase (NiRAN) domain that transfers a GMP moiety to the 5′ pp-RNA to form a 5′ Gppp-RNA cap that NSP14 can methylate in the second step of mRNA capping [28].

13. NSP13

NSP13 is an ATP-dependent RdRP containing an ATP-binding helicase core domain and a zinc-binding domain involved in unwinding the RNA during replication and the transcription of complex RNAs with secondary and tertiary structures [34][35]. It also acts as an RTPase that removes the 5′ gamma phosphate from the mRNA to produce a 5′ pp-RNA in the first step of mRNA capping [36].

14. NSP14

NSP14 is a Zn-dependent exoribonuclease that proofreads and removes mismatched nucleotides in a 3′-5′ direction incorrectly added by the RNA polymerase during genome replication [28][29][37]. This exoribonuclease domain is stabilized by the zinc-finger domain of NSP10, which increases its ability to excise nucleotides [38]. NSP14 also possesses an N7-guanine methyltransferase domain that adds a methyl group to the N7 position of the 5′ Gppp-RNA intermediate in the presence of an S-adenosylmethionine methyl donor during the second step of mRNA capping [39].

15. NSP15

NSP15 is an Mn2+-dependent nidoviral RNA uridylate-specific endoribonuclease (NendoU) with 3 domains: an N-terminal oligomerization domain, a central domain, and a C-terminal catalytic domain [40][41]. The catalytic domain cleaves the nucleotides of single-stranded and double-stranded RNA molecules at the 3′ end of uridylates to produce 2′-3′ phosphodiester and 5′-hydroxyl termini [40][41]. NendoUs are highly conserved in coronaviruses, and they avoid immune detection by cleaving the poly-U sequences on their negative-sense viral RNA templates to avoid detection by MDA5 pattern recognition receptors [40][42]. They also act against the innate immune system by suppressing IFN-IFN-α/β-associated pathways that lead to the formation of certain cytoplasmic stress granules [43].

16. NSP16

NSP16 is a 2′-O-ribose methyltransferase that requires an S-adenosylmethionine (SAM) methyl donor that promotes the association of NSP16 to its cofactor, NSP10 [13][39][44]. This increases the affinity of the NSP16-NSP10 complex to mRNA transcripts and allows the complex to add a methyl group to the 2′-O-ribose of the 5′ cap in the final step of mRNA capping [45]. This allows the viral mRNA to evade being detected by host MDA5 and RIG-I receptors that recognize and degrade unprocessed mRNA transcripts.

References

  1. Thoms, M.; Buschauer, R.; Ameismeier, M.; Koepke, L.; Denk, T.; Hirschenberger, M.; Kratzat, H.; Hayn, M.; Mackens-Kiani, T.; Cheng, J.; et al. Structural basis for translational shutdown and immune evasion by the Nsp1 protein of SARS-CoV-2. Science 2020, 369, 1249–1255.
  2. Schubert, K.; Karousis, E.D.; Jomaa, A.; Scaiola, A.; Echeverria, B.; Gurzeler, L.-A.; Leibundgut, M.; Thiel, V.; Mühlemann, O.; Ban, N. SARS-CoV-2 Nsp1 binds the ribosomal mRNA channel to inhibit translation. Nat. Struct. Mol. Biol. 2020, 27, 959–966.
  3. Afsar, M.; Narayan, R.; Akhtar, M.N.; Das, D.; Rahil, H.; Nagaraj, S.K.; Eswarappa, S.M.; Tripathi, S.; Hussain, T. Drug targeting Nsp1-ribosomal complex shows antiviral activity against SARS-CoV-2. eLife 2022, 11, e74877.
  4. Kao, H.-T.; Orry, A.; Palfreyman, M.G.; Porton, B. Synergistic interactions of repurposed drugs that inhibit Nsp1, a major virulence factor for COVID-19. Sci. Rep. 2022, 12, 10174.
  5. Vora, S.M.; Fontana, P.; Mao, T.; Leger, V.; Zhang, Y.; Fu, T.-M.; Lieberman, J.; Gehrke, L.; Shi, M.; Wang, L.; et al. Targeting stem-loop 1 of the SARS-CoV-2 5′ UTR to suppress viral translation and Nsp1 evasion. Proc. Natl. Acad. Sci USA 2022, 119, e2117198119.
  6. Gaglia, M.M.; Covarrubias, S.; Wong, W.; Glaunsinger, B.A. A Common Strategy for Host RNA Degradation by Divergent Viruses. J. Virol. 2012, 86, 9527–9530.
  7. Zhang, K.; Miorin, L.; Makio, T.; Dehghan, I.; Gao, S.; Xie, Y.; Zhong, H.; Esparza, M.; Kehrer, T.; Kumar, A.; et al. Nsp1 protein of SARS-CoV-2 disrupts the mRNA export machinery to inhibit host gene expression. Sci. Adv. 2021, 7, eabe7386.
  8. Cornillez-Ty, C.T.; Liao, L.; Yates, J.R.; Kuhn, P.; Buchmeier, M.J. Severe Acute Respiratory Syndrome Coronavirus Nonstructural Protein 2 Interacts with a Host Protein Complex Involved in Mitochondrial Biogenesis and Intracellular Signaling. J. Virol. 2009, 83, 10314–10318.
  9. Tatsuta, T.; Model, K.; Langer, T. Formation of membrane-bound ring complexes by prohibitins in mitochondria. Mol. Biol. Cell. 2005, 16, 248–259.
  10. Gamble, S.C.; Chotai, D.; Odontiadis, M.; Dart, D.A.; Brooke, G.N.; Powell, S.M.; Reebye, V.; Varela-Carver, A.; Kawano, Y.; Waxman, J.; et al. Prohibitin, a protein downregulated by androgens, represses androgen receptor activity. Oncogene 2007, 26, 1757–1768.
  11. Wintachai, P.; Wikan, N.; Kuadkitkan, A.; Jaimipuk, T.; Ubol, S.; Pulmanausahakul, R.; Auewarakul, P.; Kasinrerk, W.; Weng, W.-Y.; Panyasrivanit, M.; et al. Identification of prohibitin as a Chikungunya virus receptor protein. J. Med. Virol. 2012, 84, 1757–1770.
  12. Kuadkitkan, A.; Wikan, N.; Fongsaran, C.; Smith, D.R. Identification and characterization of prohibitin as a receptor protein mediating DENV-2 entry into insect cells. Virology 2010, 406, 149–161.
  13. Shin, D.; Mukherjee, R.; Grewe, D.; Bojkova, D.; Baek, K.; Bhattacharya, A.; Schulz, L.; Widera, M.; Mehdipour, A.R.; Tascher, G.; et al. Papain-like protease regulates SARS-CoV-2 viral spread and innate immunity. Nature 2020, 587, 657–662.
  14. Pitsillou, E.; Liang, J.; Ververis, K.; Lim, K.W.; Hung, A.; Karagiannis, T.C. Identification of small molecule inhibitors of the deubiquitinating activity of the SARS-CoV-2 papain-like protease: In silico molecular docking studies and in vitro enzymatic activity assay. Front. Chem. 2020, 8, 623971.
  15. Rao, P.; Patel, R.; Shukla, A.; Parmar, P.; Rawal, R.M.; Saraf, M.; Goswami, D. Identifying structural-functional analogue of GRL0617, the only well-established inhibitor for papain-like protease (PLpro) of SARS-CoV2 from the pool of fungal metabolites using docking and molecular dynamics simulation. Mol. Divers. 2022, 26, 309–329.
  16. Angelini, M.M.; Akhlaghpour, M.; Neuman, B.W.; Buchmeier, M.J. Severe Acute Respiratory Syndrome Coronavirus Nonstructural Proteins 3, 4, and 6 Induce Double-Membrane Vesicles. mBio 2013, 4, e00524-13.
  17. Roe, M.K.; Junod, N.A.; Young, A.R.; Beachboard, D.C.; Stobart, C.C. Targeting novel structural and functional features of coronavirus protease nsp5 (3CLpro, Mpro) in the age of COVID-19. J. Gen. Virol. 2021, 102, 001558.
  18. Milligan, J.C.; Zeisner, T.U.; Papageorgiou, G.; Joshi, D.; Soudy, C.; Ulferts, R.; Wu, M.; Lim, C.T.; Tan, K.W.; Weissmann, F.; et al. Identifying SARS-CoV-2 antiviral compounds by screening for small molecule inhibitors of Nsp5 main protease. Biochem. J. 2021, 478, 2499–2515.
  19. Cottam, E.M.; Maier, H.J.; Manifava, M.; Vaux, L.C.; Chandra-Schoenfelder, P.; Gerner, W.; Britton, P.; Ktistakis, N.T.; Wileman, T. Coronavirus nsp6 proteins generate autophagosomes from the endoplasmic reticulum via an omegasome intermediate. Autophagy 2011, 7, 1335–1347.
  20. Cottam, E.M.; Whelband, M.C.; Wileman, T. Coronavirus NSP6 restricts autophagosome expansion. Autophagy 2014, 10, 1426–1441.
  21. Sun, X.; Liu, Y.; Huang, Z.; Xu, W.; Hu, W.; Yi, L.; Liu, Z.; Chan, H.; Zeng, J.; Liu, X.; et al. SARS-CoV-2 non-structural protein 6 triggers NLRP3-dependent pyroptosis by targeting ATP6AP1. Cell Death Differ. 2022, 29, 1240–1254.
  22. Te Velthuis, A.J.W.; van den Worm, S.H.E.; Snijder, E.J. The SARS-coronavirus nsp7+nsp8 complex is a unique multimeric RNA polymerase capable of both de novo initiation and primer extension. Nucleic Acids Res. 2012, 40, 1737–1747.
  23. Xia, H.; Cao, Z.; Xie, X.; Zhang, X.; Chen, J.Y.-C.; Wang, H.; Menachery, V.D.; Rajsbaum, R.; Shi, P.-Y. Evasion of Type I Interferon by SARS-CoV-2. Cell Rep. 2020, 33, 108234.
  24. Chandel, V.; Sharma, P.P.; Raj, S.; Choudhari, R.; Rathi, B.; Kumar, D. Structure-based drug repurposing for targeting Nsp9 replicase and spike proteins of severe acute respiratory syndrome coronavirus 2. J. Biomol. Struct. Dyn. 2022, 40, 249–262.
  25. Wang, B.; Svetlov, D.; Artsimovitch, I. NMPylation and de-NMPylation of SARS-CoV-2 nsp9 by the NiRAN domain. Nucleic Acids Res. 2021, 49, 8822–8835.
  26. Makiyama, K.; Hazawa, M.; Kobayashi, A.; Lim, K.; Voon, D.C.; Wong, R.W. NSP9 of SARS-CoV-2 attenuates nuclear transport by hampering nucleoporin 62 dynamics and functions in host cells. Biochem. Biophys. Res Commun. 2022, 586, 137–142.
  27. Decroly, E.; Debarnot, C.; Ferron, F.; Bouvet, M.; Coutard, B.; Imbert, I.; Gluais, L.; Papageorgiou, N.; Sharff, A.; Bricogne, G.; et al. Crystal Structure and Functional Analysis of the SARS-Coronavirus RNA Cap 2′-O-Methyltransferase nsp10/nsp16 Complex. PLoS Pathog. 2011, 7, e1002059.
  28. Bouvet, M.; Imbert, I.; Subissi, L.; Gluais, L.; Canard, B.; Decroly, E. RNA 3′-end mismatch excision by the severe acute respiratory syndrome coronavirus nonstructural protein nsp10/nsp14 exoribonuclease complex. Proc. Natl. Acad. Sci. USA 2012, 109, 9372–9377.
  29. Ma, Y.; Wu, L.; Shaw, N.; Gao, Y.; Wang, J.; Sun, Y.; Lou, Z.; Yan, L.; Zhang, R.; Rao, Z. Structural basis and functional analysis of the SARS coronavirus nsp14–nsp10 complex. Proc. Natl. Acad. Sci. USA 2015, 112, 9436–9441.
  30. Gadhave, K.; Kumar, P.; Kumar, A.; Bhardwaj, T.; Garg, N.; Giri, R. Conformational dynamics of 13 amino acids long NSP11 of SARS-CoV-2 under membrane mimetics and different solvent conditions. Microb. Pathog. 2021, 158, 105041.
  31. Peng, Q.; Peng, R.; Yuan, B.; Zhao, J.; Wang, M.; Wang, X.; Wang, Q.; Sun, Y.; Fan, Z.; Qi, J.; et al. Structural and Biochemical Characterization of the nsp12-nsp7-nsp8 Core Polymerase Complex from SARS-CoV-2. Cell Rep. 2020, 31, 107774.
  32. Hillen, H.S.; Kokic, G.; Farnung, L.; Dienemann, C.; Tegunov, D.; Cramer, P. Structure of replicating SARS-CoV-2 polymerase. Nature 2020, 584, 154–156.
  33. Subissi, L.; Posthuma, C.C.; Collet, A.; Zevenhoven-Dobbe, J.C.; Gorbalenya, A.E.; Decroly, E.; Snijder, E.J.; Canard, B.; Imbert, I. One severe acute respiratory syndrome coronavirus protein complex integrates processive RNA polymerase and exonuclease activities. Proc. Natl. Acad. Sci. USA 2014, 111, E3900–E3909.
  34. Chen, J.; Malone, B.; Llewellyn, E.; Grasso, M.; Shelton, P.M.M.; Olinares, P.D.B.; Maruthi, K.; Eng, E.T.; Vatandaslar, H.; Chait, B.T.; et al. Structural Basis for Helicase-Polymerase Coupling in the SARS-CoV-2 Replication-Transcription Complex. Cell 2020, 182, 1560–1573.e13.
  35. Yan, L.; Zhang, Y.; Ge, J.; Zheng, L.; Gao, Y.; Wang, T.; Jia, Z.; Wang, H.; Huang, Y.; Li, M.; et al. Architecture of a SARS-CoV-2 mini replication and transcription complex. Nat. Commun. 2020, 11, 5874.
  36. Ivanov, K.A.; Ziebuhr, J. Human Coronavirus 229E Nonstructural Protein 13: Characterization of Duplex-Unwinding, Nucleoside Triphosphatase, and RNA 5′-Triphosphatase Activities. J. Virol. 2004, 78, 7833–7838.
  37. Chen, Y.; Cai, H.; Pan, J.; Xiang, N.; Tien, P.; Ahola, T.; Guo, D. Functional screen reveals SARS coronavirus nonstructural protein nsp14 as a novel cap N7 methyltransferase. Proc. Natl. Acad. Sci. USA 2009, 106, 3484–3489.
  38. Chen, T.; Fei, C.-Y.; Chen, Y.-P.; Sargsyan, K.; Chang, C.-P.; Yuan, H.S.; Lim, C. Synergistic Inhibition of SARS-CoV-2 Replication Using Disulfiram/Ebselen and Remdesivir. ACS Pharmacol. Transl. Sci. 2021, 4, 898–907.
  39. Ahmed-Belkacem, R.; Sutto-Ortiz, P.; Guiraud, M.; Canard, B.; Vasseur, J.-J.; Decroly, E.; Debart, F. Synthesis of adenine dinucleosides SAM analogs as specific inhibitors of SARS-CoV nsp14 RNA cap guanine-N7-methyltransferase. Eur. J. Med. Chem. 2020, 201, 112557.
  40. Hong, S.; Seo, S.H.; Woo, S.-J.; Kwon, Y.; Song, M.; Ha, N.-C. Epigallocatechin gallate inhibits the uridylate-specific endoribonuclease Nsp15 and efficiently neutralizes the SARS-CoV-2 strain. J. Agric. Food Chem. 2021, 69, 5948–5954.
  41. Khan, A.R.; Misdary, C.; Yegya-Raman, N.; Kim, S.; Narayanan, N.; Siddiqui, S.; Salgame, P.; Radbel, J.; Groote, F.D.; Michel, C.; et al. Montelukast in hospitalized patients diagnosed with COVID-19. J. Asthma 2022, 59, 780–786.
  42. Kim, Y.; Wower, J.; Maltseva, N.; Chang, C.; Jedrzejczak, R.; Wilamowski, M.; Kang, S.; Nicolaescu, V.; Randall, G.; Michalska, K.; et al. Tipiracil binds to uridine site and inhibits Nsp15 endoribonuclease NendoU from SARS-CoV-2. Commun. Biol. 2021, 4, 193.
  43. Nakagawa, K.; Narayanan, K.; Wada, M.; Makino, S. Inhibition of Stress Granule Formation by Middle East Respiratory Syndrome Coronavirus 4a Accessory Protein Facilitates Viral Translation, Leading to Efficient Virus Replication. J. Virol. 2018, 92, e00902-18.
  44. Tazikeh-Lemeski, E.; Moradi, S.; Raoufi, R.; Shahlaei, M.; Janlou, M.A.M.; Zolghadri, S. Targeting SARS-CoV-2 non-structural protein 16: A virtual drug repurposing study. J. Biomol. Struct. Dyn. 2021, 39, 4633–4646.
  45. Zeng, C.; Wu, A.; Wang, Y.; Xu, S.; Tang, Y.; Jin, X.; Wang, S.; Qin, L.; Sun, Y.; Fan, C.; et al. Identification and Characterization of a Ribose 2′-O-Methyltransferase Encoded by the Ronivirus Branch of Nidovirales. J. Virol. 2016, 90, 6675–6685.
More
Information
Subjects: Virology
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , ,
View Times: 395
Entry Collection: COVID-19
Revisions: 2 times (View History)
Update Date: 23 Aug 2023
1000/1000
Video Production Service