Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2022, Volume 16, Issue 9 doi: 10.1016/j.eng.2021.05.010

Multi-Omics-Guided Discovery of Omicsynins Produced by Streptomyces sp. 1647: Pseudo-Tetrapeptides Active Against Influenza A Viruses and Coronavirus HCoV-229E

a NHC Key Laboratory of Biotechnology of Antibiotics, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
b CAMS Key Laboratory of Synthetic Biology for Drug Innovation, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
c CAMS Key Laboratory of Antiviral Drug Research, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China

Received: 2020-10-30 Revised: 2021-05-06 Accepted: 2021-05-16 Available online: 2021-06-26

Next Previous

Abstract

Many microorganisms have mechanisms that protect cells against attack from viruses. The fermentation components of Streptomyces sp. 1647 exhibit potent anti-influenza A virus (IAV) activity. This strain was isolated from soil in southern China in the 1970s, but the chemical nature of its antiviral substance(s) has remained unknown until now. We used an integrated multi-omics strategy to identify the antiviral agents from this streptomycete. The antibiotics and Secondary Metabolite Analysis Shell (antiSMASH) analysis of its genome sequence revealed 38 biosynthetic gene clusters (BGCs) for secondary metabolites, and the target BGCs possibly responsible for the production of antiviral components were narrowed down to three BGCs by bioactivity-guided comparative transcriptomics analysis. Through bioinformatics analysis and genetic manipulation of the regulators and a biosynthetic gene, cluster 36 was identified as the BGC responsible for the biosynthesis of the antiviral compounds. Bioactivity-based molecular networking analysis of mass spectrometric data from different recombinant strains illustrated that the antiviral compounds were a class of structural analogues. Finally, 18 pseudo-tetrapeptides with an internal ureido linkage, omicsynins A1–A6, B1–B6, and C1–C6, were identified and/or isolated from fermentation broth. Among them, 11 compounds (omicsynins A1, A2, A6, B1–B3, B5, B6, C1, C2, and C6) are new compounds. Omicsynins B1–B4 exhibited potent antiviral activity against IAV with the 50% inhibitory concentration (IC50) of approximately 1 µmol∙L–1 and a selectivity index (SI) ranging from 100 to 300. Omicsynins B1–B4 also showed significant antiviral activity against human coronavirus HCoV-229E. By integrating multi-omics data, we discovered a number of novel antiviral pseudo-tetrapeptides produced by Streptomyces sp. 1647, indicating that the secondary metabolites of microorganisms are a valuable source of novel antivirals.

SupplementaryMaterials

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

Fig. 6

References

[ 1 ] WHO. Coronavirus disease (COVID-19) weekly epidemiological update and weekly operational update—situation reports [Internet]. Geneva: World Health Organization; [cited 2021 Apr 30]. Available from: https://www.who.int/ emergencies/diseases/novel-coronavirus-2019/situation-reports. link1

[ 2 ] Newman DJ, Cragg GM. Natural products as sources of new drugs over the nearly four decades from 01/1981 to 09/2019. J Nat Prod 2020;83(3):770–803. link1

[ 3 ] Takizawa N, Yamasaki M. Current landscape and future prospects of antiviral drugs derived from microbial products. J Antibiot 2018;71(1):45–52. link1

[ 4 ] Martinez JP, Sasse F, Brönstrup M, Diez J, Meyerhans A. Antiviral drug discovery: broad-spectrum drugs from nature. Nat Prod Rep 2015;32 (1):29–48. link1

[ 5 ] Bérdy J. Bioactive microbial metabolites. J Antibiot 2005;58(1):1–26. link1

[ 6 ] Williams R, Hoehn M, inventors; ELI LILLY AND CO., assignee. Pyrazomycin and process for production. United States patent US 3802999. 1974 Sep 4.

[ 7 ] Sagar S, Kaur M, Minneman KP. Antiviral lead compounds from marine sponges. Mar Drugs 2010;8(10):2619–38. link1

[ 8 ] Yoon JS, Kim G, Jarhad DB, Kim HR, Shin YS, Qu S, et al. Design, synthesis, and anti-RNA virus activity of 60 -fluorinated-aristeromycin analogues. J Med Chem 2019;62(13):6346–62. link1

[ 9 ] Kusaka T, Yamamoto H, Shibata M, Muroi M, Kishi T, Mizuno K. Streptomyces citricolor nov. sp. and a new antibiotic, aristeromycin. J Antibiot 1968;21 (4):255–63. link1

[10] Kieser T, Bibb MJ, Buttner MJ, Chater KF, Hopwood DA. Practical Streptomyces genetics. Norwich: John Innes Foundation; 2000. link1

[11] Hong B, Phornphisutthimas S, Tilley E, Baumberg S, McDowall KJ. Streptomycin production by Streptomyces griseus can be modulated by a mechanism not associated with change in the adpA component of the A-factor cascade. Biotechnol Lett 2006;29(1):57–64. link1

[12] Bierman M, Logan R, O’Brien K, Seno ET, Nagaraja Rao R, Schoner BE. Plasmid cloning vectors for the conjugal transfer of DNA from Escherichia coli to Streptomyces spp. Gene 1992;116(1):43–9. link1

[13] Kohl M, Wiese S, Warscheid B. Cytoscape: software for visualization and analysis of biological networks. Methods Mol Biol 2011;696(696):291–303. link1

[14] Yin J, Ma L, Wang H, Yan H, Hu J, Jiang W, et al. Chinese herbal medicine compound Yi-Zhi-Hao pellet inhibits replication of influenza virus infection through activation of heme oxygenase-1. Acta Pharm Sin B 2017;7(6):630–7. link1

[15] Pizzi M. Sampling variation of the fifty percent end-point, determined by the Reed-Muench (Behrens) method. Hum Biol 1950;22(3):151–90. link1

[16] Zhong M, Wang HQ, Yan HY, Wu S, Gu ZY, Li YH. Santin inhibits influenza A virus replication through regulating MAPKs and NF-jB pathways. J Asian Nat Prod Res 2019;21(12):1205–14. link1

[17] Wang H, Zhang D, Ge M, Li Z, Jiang J, Li Y. Formononetin inhibits enterovirus 71 replication by regulating COX-2/PGE2 expression. Virol J 2015;12(1):35. link1

[18] Jiang Z, Li X, Ren W, Shi Y, Gao R, Li Y, et al. Discovery of siderophore compounds using genome mining strategy. Chin Med Biotechnol 2019;14 (2):97–107. Chinese.

[19] Escolar L, Pe´ rez-Marti´n J, de Lorenzo V. Opening the iron box: transcriptional metalloregulation by the Fur protein. J Bacteriol 1999;181(20):6223–9. link1

[20] Maxson T, Tietz JI, Hudson GA, Guo XR, Tai HC, Mitchell DA. Targeting reactive carbonyls for identifying natural products and their biosynthetic origins. J Am Chem Soc 2016;138(46):15157–66. link1

[21] Wang M, Carver JJ, Phelan VV, Sanchez LM, Garg N, Peng Y, et al. Sharing and community curation of mass spectrometry data with Global Natural Products Social Molecular Networking. Nat Biotechnol 2016;34(8):828–37. link1

[22] Lai CJS, Zha L, Liu DH, Kang L, Ma X, Zhan ZL, et al. Global profiling and rapid matching of natural products using diagnostic product ion network and in silico analogue database: Gastrodia elata as a case study. J Chromatogr A 2016;1456:187–95. link1

[23] Wang Q, Zhang Y, Wang M, Tan Y, Hu X, He H, et al. Neo-actinomycins A and B, natural actinomycins bearing the 5H-oxazolo[4,5-b]phenoxazine chromophore, from the marine-derived Streptomyces sp. IMB094. Sci Rep 2017;7(1):3591.

[24] Fujii K, Ikai Y, Oka H, Suzuki M, Harada KI. A nonempirical method using LC/MS for determination of the absolute configuration of constituent amino acids in a peptide: combination of Marfey’s method with mass spectrometry and its practical application. Anal Chem 1997;69(24):5146–51. link1

[25] Hamano K, Tanzawa K, Takahashi M, Enokida R, Okazaki H, Kinoshita T, et al., inventors; SANKYO Co., Ltd., assignee. Chymostatinols manufacture with Streptomyces for treatment of osteoporosis. Japanese patent JP 08003188 A. 1996 Jan 9.

[26] Konda Y, Takahashi Y, Arima S, Sato N, Takeda K, Dobashi K, et al. First total synthesis of Mer-N5075A and a diastereomeric mixture of a and b-MAPI, new HIV-I protease inhibitors from a species of Streptomyces. Tetrahedron 2001;57 (20):4311–21. link1

[27] Hoebeke J, Busatto-Samsoen C, Davoust D, Lebrun E. 1 H NMR study of the diastereomeric forms of the protease inhibitor antipain. Magn Reson Chem 1994;32(4):220–4. link1

[28] Jiang P. Preliminary research on antiviral drugs and vaccines [dissertation]. Beijing: Peking University; 2005. Chinese.

[29] Nothias LF, Nothias-Esposito M, da Silva R, Wang M, Protsyuk I, Zhang Z, et al. Bioactivity-based molecular networking for the discovery of drug leads in natural product bioassay-guided fractionation. J Nat Prod 2018;81(4):758–67. link1

[30] Kim ES. Midostaurin: first global approval. Drugs 2017;77(11):1251–9. link1

[31] Suda H, Aoyagi T, Hamada M, Takeuchi T, Umezawa H. Antipain, a new protease inhibitor isolated from actinomycetes. J Antibiot 1972;25(4):263–6. link1

[32] Umezawa H, Aoyagi T, Morishima H, Kunimoto S, Matsuzaki M, Hamada M, et al. Chymostatin, a new chymotrypsin inhibitor produced by actinomycetes. J Antibiot 1970;23(8):425–7. link1

[33] Umezawa H, Aoyagi T, Okura A, Morishima H, Takeuchi T, Okami Y. Letter: elastatinal, a new elastase inhibitor produced by actinomycetes. J Antibiot 1973;26(12):787–9. link1

[34] Appleyard G, Tisdale M. Inhibition of the growth of human coronavirus 229E by leupeptin. J Gen Virol 1985;66(2):363–6. link1

[35] Molla A, Hellen CU, Wimmer E. Inhibition of proteolytic activity of poliovirus and rhinovirus 2A proteinases by elastase-specific inhibitors. J Virol 1993;67 (8):4688–95. link1

[36] Tashiro M, Klenk HD, Rott R. Inhibitory effect of a protease inhibitor, leupeptin, on the development of influenza pneumonia, mediated by concomitant bacteria. J Gen Virol 1987;68(7):2039–41. link1

Related Research