1887

Abstract

A notable signalling mechanism employed by mammalian innate immune signalling pathways uses nucleotide-based second messengers such as 2′3′-cGAMP and 2′–5′-oligoadenylates (OAs), which bind and activate STING and RNase L, respectively. Interestingly, the involvement of nucleotide second messengers to activate antiviral responses is evolutionarily conserved, as evidenced by the identification of an antiviral cGAMP-dependent pathway in . Using a mass spectrometry approach, we identified several members of the ABCF family in human, mouse and cell lysates as 2′–5′ OA-binding proteins, suggesting an evolutionarily conserved function. Biochemical characterization of these interactions demonstrates high-affinity binding of 2′–5′ OA to ABCF1, dependent on phosphorylated 2′–5′ OA and an intact Walker A/B motif of the ABC cassette of ABCF1. As further support for species-specific interactions with 2′–5′ OA, we additionally identified that the metabolic enzyme Decr1 from mouse, but not human or cells, forms a high-affinity complex with 2′–5′ OA. A 1.4 Å co-crystal structure of the mouse Decr1–2′–5′ OA complex explains high-affinity recognition of 2′–5′ OA and the mechanism of species specificity. Despite clear evidence of physical interactions, we could not identify profound antiviral functions of ABCF1, ABCF3 or Decr1 or 2′–5′ OA-dependent regulation of cellular translation rates, as suggested by the engagement of ABCF proteins. Thus, although the biological consequences of the here identified interactions need to be further studied, our data suggest that 2′–5′ OA can serve as a signalling hub to distribute a signal to different recipient proteins.

Keyword(s): 2′5′OA , ABCF1 , ABCF3 and innate immunity
Funding
This study was supported by the:
  • National Science Foundation (Award Graduate Research Fellowship)
    • Principle Award Recipient: ApurvaA. Govande
  • Richard and Susan Smith Family Foundation
    • Principle Award Recipient: PhilipJ. Kranzusch
  • Bayerisches Staatsministerium für Wissenschaft, Forschung und Kunst (Award Bavarian Research Network FOR-COVID)
    • Principle Award Recipient: AndreasPichlmair
  • Deutsche Forschungsgemeinschaft (Award PI 1084/3, PI 1084/4, PI 1084/5 and TRR179, TRR237)
    • Principle Award Recipient: AndreasPichlmair
  • HORIZON EUROPE European Research Council () (Award ProDAP, 817798)
    • Principle Award Recipient: AndreasPichlmair
  • This is an open-access article distributed under the terms of the Creative Commons Attribution License. This article was made open access via a Publish and Read agreement between the Microbiology Society and the corresponding author’s institution.
Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001890
2023-09-07
2024-05-03
Loading full text...

Full text loading...

/deliver/fulltext/jgv/104/9/jgv001890.html?itemId=/content/journal/jgv/10.1099/jgv.0.001890&mimeType=html&fmt=ahah

References

  1. Kerr IM, Brown RE, Hovanessian AG. Nature of inhibitor of cell-free protein synthesis formed in response to interferon and double-stranded RNA. Nature 1977; 268:540–542 [View Article] [PubMed]
    [Google Scholar]
  2. Hovanessian AG, Brown RE, Kerr IM. Synthesis of low molecular weight inhibitor of protein synthesis with enzyme from interferon-treated cells. Nature 1977; 268:537–540 [View Article] [PubMed]
    [Google Scholar]
  3. Kerr IM, Brown RE. pppA2’p5’A2’p5’A: an inhibitor of protein synthesis synthesized with an enzyme fraction from interferon-treated cells. Proc Natl Acad Sci 1978; 75:256–260 [View Article] [PubMed]
    [Google Scholar]
  4. Donovan J, Dufner M, Korennykh A. Structural basis for cytosolic double-stranded RNA surveillance by human oligoadenylate synthetase 1. Proc Natl Acad Sci U S A 2013; 110:1652–1657 [View Article] [PubMed]
    [Google Scholar]
  5. Donovan J, Whitney G, Rath S, Korennykh A. Structural mechanism of sensing long dsRNA via a noncatalytic domain in human oligoadenylate synthetase 3. Proc Natl Acad Sci U S A 2015; 112:3949–3954 [View Article] [PubMed]
    [Google Scholar]
  6. Chebath J, Benech P, Hovanessian A, Galabru J, Revel M. Four different forms of interferon-induced 2’,5’-oligo(A) synthetase identified by immunoblotting in human cells. J Biol Chem 1987; 262:3852–3857 [PubMed]
    [Google Scholar]
  7. Hartmann R, Olsen HS, Widder S, Jorgensen R, Justesen J. p59OASL, a 2’-5’ oligoadenylate synthetase like protein: a novel human gene related to the 2’-5’ oligoadenylate synthetase family. Nucleic Acids Res 1998; 26:4121–4128 [View Article] [PubMed]
    [Google Scholar]
  8. Zhu J, Zhang Y, Ghosh A, Cuevas RA, Forero A et al. Antiviral activity of human OASL protein is mediated by enhancing signaling of the RIG-I RNA sensor. Immunity 2014; 40:936–948 [View Article] [PubMed]
    [Google Scholar]
  9. Ibsen MS, Gad HH, Andersen LL, Hornung V, Julkunen I et al. Structural and functional analysis reveals that human OASL binds dsRNA to enhance RIG-I signaling. Nucleic Acids Res 2015; 43:5236–5248 [View Article] [PubMed]
    [Google Scholar]
  10. Lohöfener J, Steinke N, Kay-Fedorov P, Baruch P, Nikulin A et al. The activation mechanism of 2’-5’-oligoadenylate synthetase gives new insights into OAS/cGAS triggers of innate immunity. Struct Lond Engl 2015; 23:851–862 [View Article] [PubMed]
    [Google Scholar]
  11. Ghosh A, Sarkar SN, Sen GC. Cell growth regulatory and antiviral effects of the P69 isozyme of 2-5 (A) synthetase. Virology 2000; 266:319–328 [View Article] [PubMed]
    [Google Scholar]
  12. Li Y, Banerjee S, Wang Y, Goldstein SA, Dong B et al. Activation of RNase L is dependent on OAS3 expression during infection with diverse human viruses. Proc Natl Acad Sci U S A 2016; 113:2241–2246 [View Article] [PubMed]
    [Google Scholar]
  13. Kwon Y-C, Kang J-I, Hwang SB, Ahn B-Y. The ribonuclease L-dependent antiviral roles of human 2’,5’-oligoadenylate synthetase family members against hepatitis C virus. FEBS Lett 2013; 587:156–164 [View Article] [PubMed]
    [Google Scholar]
  14. Lin R-J, Yu H-P, Chang B-L, Tang W-C, Liao C-L et al. Distinct antiviral roles for human 2’,5’-oligoadenylate synthetase family members against dengue virus infection. J Immunol 2009; 183:8035–8043 [View Article] [PubMed]
    [Google Scholar]
  15. Morin B, Rabah N, Boretto-Soler J, Tolou H, Alvarez K et al. High yield synthesis, purification and characterisation of the RNase L activators 5’-triphosphate 2’-5’-oligoadenylates. Antiviral Res 2010; 87:345–352 [View Article] [PubMed]
    [Google Scholar]
  16. Silverman RH. Functional analysis of 2-5A-dependent RNase and 2-5A using 2’,5’-oligoadenylate-cellulose. Anal Biochem 1985; 144:450–460 [View Article] [PubMed]
    [Google Scholar]
  17. Dong B, Silverman RH. 2-5A-dependent RNase molecules dimerize during activation by 2-5A. J Biol Chem 1995; 270:4133–4137 [View Article] [PubMed]
    [Google Scholar]
  18. Han Y, Whitney G, Donovan J, Korennykh A. Innate immune messenger 2-5A tethers human RNase L into active high-order complexes. Cell Rep 2012; 2:902–913 [View Article] [PubMed]
    [Google Scholar]
  19. Li G, Xiang Y, Sabapathy K, Silverman RH. An apoptotic signaling pathway in the interferon antiviral response mediated by RNase L and c-Jun NH2-terminal kinase. J Biol Chem 2004; 279:1123–1131 [View Article] [PubMed]
    [Google Scholar]
  20. Manivannan P, Reddy V, Mukherjee S, Clark KN, Malathi K. RNase L induces expression of a novel Serine/Threonine protein kinase, DRAK1, to promote apoptosis. Int J Mol Sci 2019; 20:E3535 [View Article] [PubMed]
    [Google Scholar]
  21. Kuusksalu A, Pihlak A, Müller WE, Kelve M. The (2’-5’)oligoadenylate synthetase is present in the lowest multicellular organisms, the marine sponges. Demonstration of the existence and identification of its reaction products. Eur J Biochem 1995; 232:351–357 [PubMed]
    [Google Scholar]
  22. Scherbik SV, Kluetzman K, Perelygin AA, Brinton MA. Knock-in of the Oas1b(r) allele into a flavivirus-induced disease susceptible mouse generates the resistant phenotype. Virology 2007; 368:232–237 [View Article] [PubMed]
    [Google Scholar]
  23. Peterson E, Shippee E, Brinton MA, Kaur P. Biochemical characterization of the mouse ABCF3 protein, a partner of the flavivirus-resistance protein OAS1B. J Biol Chem 2019; 294:14937–14952 [View Article] [PubMed]
    [Google Scholar]
  24. Madden JC, Cui D, Brinton MA, Heise MT. RNase L antiviral activity is not a critical component of the oas1b-mediated Flavivirus resistance phenotype. J Virol 2019; 93:e00946-19 [View Article] [PubMed]
    [Google Scholar]
  25. Elkhateeb E, Tag-El-Din-Hassan HT, Sasaki N, Torigoe D, Morimatsu M et al. The role of mouse 2’,5’-oligoadenylate synthetase 1 paralogs. Infect Genet Evol 2016; 45:393–401 [View Article] [PubMed]
    [Google Scholar]
  26. Hancks DC, Hartley MK, Hagan C, Clark NL, Elde NC. Overlapping patterns of rapid evolution in the nucleic acid sensors cGAS and OAS1 suggest a common mechanism of pathogen antagonism and escape. PLoS Genet 2015; 11:e1005203 [View Article] [PubMed]
    [Google Scholar]
  27. Zhou W, Whiteley AT, de Oliveira Mann CC, Morehouse BR, Nowak RP et al. Structure of the human cGAS-DNA complex reveals enhanced control of immune surveillance. Cell 2018; 174:300–311 [View Article] [PubMed]
    [Google Scholar]
  28. Lowey B, Whiteley AT, Keszei AFA, Morehouse BR, Mathews IT et al. CBASS immunity uses CARF-related effectors to sense 3’-5’- and 2’-5’-linked cyclic oligonucleotide signals and protect bacteria from phage infection. Cell 2020; 182:38–49 [View Article] [PubMed]
    [Google Scholar]
  29. Turpaev K, Hartmann R, Kisselev L, Justesen J. Ap3A and Ap4A are primers for oligoadenylate synthesis catalyzed by interferon-inducible 2-5A synthetase. FEBS Lett 1997; 408:177–181 [View Article] [PubMed]
    [Google Scholar]
  30. Hartmann R, Justesen J, Sarkar SN, Sen GC, Yee VC. Crystal structure of the 2’-specific and double-stranded RNA-activated interferon-induced antiviral protein 2’-5’-oligoadenylate synthetase. Mol Cell 2003; 12:1173–1185 [View Article] [PubMed]
    [Google Scholar]
  31. Hubel P, Urban C, Bergant V, Schneider WM, Knauer B et al. A protein-interaction network of interferon-stimulated genes extends the innate immune system landscape. Nat Immunol 2019; 20:493–502 [View Article] [PubMed]
    [Google Scholar]
  32. Cox J, Mann M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol 2008; 26:1367–1372 [View Article] [PubMed]
    [Google Scholar]
  33. Tyanova S, Temu T, Sinitcyn P, Carlson A, Hein MY et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat Methods 2016; 13:731–740 [View Article] [PubMed]
    [Google Scholar]
  34. Liebschner D, Afonine PV, Baker ML, Bunkóczi G, Chen VB et al. Macromolecular structure determination using X-rays, neutrons and electrons: recent developments in Phenix. Acta Crystallogr D Struct Biol 2019; 75:861–877 [View Article] [PubMed]
    [Google Scholar]
  35. Alphey MS, Yu W, Byres E, Li D, Hunter WN. Structure and reactivity of human mitochondrial 2,4-dienoyl-CoA reductase: enzyme-ligand interactions in a distinctive short-chain reductase active site. J Biol Chem 2005; 280:3068–3077 [View Article] [PubMed]
    [Google Scholar]
  36. Emsley P, Cowtan K. Coot: model-building tools for molecular graphics. Acta Crystallogr D Biol Crystallogr 2004; 60:2126–2132 [View Article] [PubMed]
    [Google Scholar]
  37. Rebouillat D, Hovnanian A, Marié I, Hovanessian AG. The 100-kDa 2’,5’-oligoadenylate synthetase catalyzing preferentially the synthesis of dimeric pppA2’p5’A molecules is composed of three homologous domains. J Biol Chem 1999; 274:1557–1565 [View Article] [PubMed]
    [Google Scholar]
  38. Tyzack JK, Wang X, Belsham GJ, Proud CG. ABC50 interacts with eukaryotic initiation factor 2 and associates with the ribosome in an ATP-dependent manner. J Biol Chem 2000; 275:34131–34139 [View Article] [PubMed]
    [Google Scholar]
  39. Paytubi S, Wang X, Lam YW, Izquierdo L, Hunter MJ et al. ABC50 promotes translation initiation in mammalian cells. J Biol Chem 2009; 284:24061–24073 [View Article] [PubMed]
    [Google Scholar]
  40. Wilcox SM, Arora H, Munro L, Xin J, Fenninger F et al. The role of the innate immune response regulatory gene ABCF1 in mammalian embryogenesis and development. PLoS One 2017; 12:e0175918 [View Article] [PubMed]
    [Google Scholar]
  41. Lee MN, Roy M, Ong S-E, Mertins P, Villani A-C et al. Identification of regulators of the innate immune response to cytosolic DNA and retroviral infection by an integrative approach. Nat Immunol 2013; 14:179–185 [View Article] [PubMed]
    [Google Scholar]
  42. Helander HM, Koivuranta KT, Horelli-Kuitunen N, Palvimo JJ, Palotie A et al. Molecular cloning and characterization of the human mitochondrial 2,4-dienoyl-CoA reductase gene (DECR). Genomics 1997; 46:112–119 [View Article] [PubMed]
    [Google Scholar]
  43. Blomme A, Ford CA, Mui E, Patel R, Ntala C et al. 2,4-dienoyl-CoA reductase regulates lipid homeostasis in treatment-resistant prostate cancer. Nat Commun 2020; 111:1–17 [View Article] [PubMed]
    [Google Scholar]
  44. Nassar ZD, Mah CY, Dehairs J, Burvenich IJ, Irani S et al. Human DECR1 is an androgen-repressed survival factor that regulates PUFA oxidation to protect prostate tumor cells from ferroptosis. eLife 2020; 9:1–34 [View Article] [PubMed]
    [Google Scholar]
  45. Kohn A, Gitelman J, Inbar M. Unsaturated free fatty acids inactivate animal enveloped viruses. Arch Virol 1980; 66:301–307 [View Article] [PubMed]
    [Google Scholar]
  46. Kohn A, Gitelman J, Inbar M. Interaction of polyunsaturated fatty acids with animal cells and enveloped viruses. Antimicrob Agents Chemother 1980; 18:962–968 [View Article] [PubMed]
    [Google Scholar]
  47. Pichlmair A, Kandasamy K, Alvisi G, Mulhern O, Sacco R et al. Viral immune modulators perturb the human molecular network by common and unique strategies. Nature 2012; 487:486–490 [View Article] [PubMed]
    [Google Scholar]
  48. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science 2013; 339:786–791 [View Article] [PubMed]
    [Google Scholar]
  49. Slavik KM, Morehouse BR, Ragucci AE, Zhou W, Ai X et al. cGAS-like receptors sense RNA and control 3’2’-cGAMP signalling in Drosophila. Nature 2021; 597:109–113 [View Article] [PubMed]
    [Google Scholar]
  50. Holleufer A, Winther KG, Gad HH, Ai X, Chen Y et al. Two cGAS-like receptors induce antiviral immunity in Drosophila. Nature 2021; 597:114–118 [View Article] [PubMed]
    [Google Scholar]
  51. Whiteley AT, Eaglesham JB, de Oliveira Mann CC, Morehouse BR, Lowey B et al. Bacterial cGAS-like enzymes synthesize diverse nucleotide signals. Nature 2019; 567:194–199 [View Article] [PubMed]
    [Google Scholar]
  52. Cohen D, Melamed S, Millman A, Shulman G, Oppenheimer-Shaanan Y et al. Cyclic GMP-AMP signalling protects bacteria against viral infection. Nature 2019; 574:691–695 [View Article] [PubMed]
    [Google Scholar]
  53. Tal N, Morehouse BR, Millman A, Stokar-Avihail A, Avraham C et al. Cyclic CMP and cyclic UMP mediate bacterial immunity against phages. Cell 2021; 184:5728–5739 [View Article] [PubMed]
    [Google Scholar]
  54. Ablasser A, Schmid-Burgk JL, Hemmerling I, Horvath GL, Schmidt T et al. Cell intrinsic immunity spreads to bystander cells via the intercellular transfer of cGAMP. Nature 2013; 503:530–534 [View Article] [PubMed]
    [Google Scholar]
  55. Stewart JD, Cowan JL, Perry LS, Coldwell MJ, Proud CG. ABC50 mutants modify translation start codon selection. Biochem J 2015; 467:217–229 [View Article] [PubMed]
    [Google Scholar]
  56. Pennemann FL, Mussabekova A, Urban C, Stukalov A, Andersen LL et al. Cross-species analysis of viral nucleic acid interacting proteins identifies TAOKs as innate immune regulators. Nat Commun 2021; 12:7009 [View Article] [PubMed]
    [Google Scholar]
  57. Arora H, Wilcox SM, Johnson LA, Munro L, Eyford BA et al. The ATP-binding cassette gene ABCF1 functions as an E2 ubiquitin-conjugating enzyme controlling macrophage polarization to dampen lethal septic shock. Immunity 2019; 50:418–431 [View Article] [PubMed]
    [Google Scholar]
  58. Drappier M, Michiels T. Inhibition of the OAS/RNase L pathway by viruses. Curr Opin Virol 2015; 15:19–26 [View Article] [PubMed]
    [Google Scholar]
  59. Bisbal C, Martinand C, Silhol M, Lebleu B, Salehzada T. Cloning and characterization of A RNAse L inhibitor. A new component of the interferon-regulated 2-5A pathway. J Biol Chem 1995; 270:13308–13317 [View Article] [PubMed]
    [Google Scholar]
  60. Martinand C, Salehzada T, Silhol M, Lebleu B, Bisbal C. The RNase L inhibitor (RLI) is induced by double-stranded RNA. J Interferon Cytokine Res 1998; 18:1031–1038 [View Article] [PubMed]
    [Google Scholar]
  61. Leu G-Z, Lin T-Y, Hsu JTA. Anti-HCV activities of selective polyunsaturated fatty acids. Biochem Biophys Res Commun 2004; 318:275–280 [View Article] [PubMed]
    [Google Scholar]
  62. Heaton NS, Randall G. Microbes and Metabolism Multifaceted roles for lipids in viral infection 2011 10.1016/j.tim.2011.03.007
    [Google Scholar]
  63. Perelygin AA, Scherbik SV, Zhulin IB, Stockman BM, Li Y et al. Positional cloning of the murine flavivirus resistance gene. Proc Natl Acad Sci U S A 2002; 99:9322–9327 [View Article] [PubMed]
    [Google Scholar]
  64. Kakuta S, Shibata S, Iwakura Y. Genomic structure of the mouse 2’,5’-oligoadenylate synthetase gene family 2004 https://home.liebertpub.com/jir 22:981–993
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001890
Loading
/content/journal/jgv/10.1099/jgv.0.001890
Loading

Data & Media loading...

Supplements

Supplementary material 1

EXCEL

Supplementary material 2

EXCEL

Supplementary material 3

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error