Lantibiotic subtilin
Need Assistance?
  • US & Canada:
    +
  • UK: +

Lantibiotic subtilin

* Please kindly note that our products are not to be used for therapeutic purposes and cannot be sold to patients.

Lantibiotic subtilin is an antibacterial peptide isolated from Bacillus subtilis.

Category
Functional Peptides
Catalog number
BAT-012582
CAS number
1393-38-0
Molecular Formula
C148H227N39O38S5
Molecular Weight
3320.94
IUPAC Name
6-amino-2-[2-[[2-[[6-amino-2-[[7-[[5-amino-2-[[2-[[2-[[(6Z)-21-[[2-[[3-[[15-[[6-amino-2-[[2-amino-3-(1H-indol-3-yl)propanoyl]amino]hexanoyl]amino]-12-(2-carboxyethyl)-9-methylidene-6-(2-methylpropyl)-5,8,11,14-tetraoxo-1-thia-4,7,10,13-tetrazacyclohexadecane-3-carbonyl]amino]-4-methyl-2,9,12-trioxo-5-thia-1,8,11-triazabicyclo[11.3.0]hexadecane-7-carbonyl]amino]-3-methylbutanoyl]amino]-9-(3-amino-3-oxopropyl)-6-ethylidene-15,22-dimethyl-12-(2-methylpropyl)-5,8,11,14,17,20-hexaoxo-1-thia-4,7,10,13,16,19-hexazacyclodocosane-3-carbonyl]amino]-3-phenylpropanoyl]amino]-4-methylpentanoyl]amino]-5-oxopentanoyl]amino]-14-(2-amino-2-oxoethyl)-8,20-dimethyl-4-(2-methylpropyl)-3,6,12,15,21-pentaoxo-9,19-dithia-2,5,13,16,22-pentazabicyclo[9.9.2]docosane-17-carbonyl]amino]hexanoyl]amino]-3-methylpentanoyl]amino]prop-2-enoylamino]hexanoic acid
Synonyms
Trp-Lys-Ser-Glu-Ser-Leu-Cys-Thr-Pro-Gly-Cys-Val-Thr-Gly-Ala-Leu-Gln-Thr-Cys-Phe-Leu-Gln-Thr-Leu-Thr-Cys-Asn-Cys-Lys-Ile-Ser-Lys
Appearance
White Powder
Sequence
WKSESLCTPGCVTGALQTCFLQTLTCNCKISK
InChI
InChI=1S/C148H227N39O38S5/c1-21-74(13)114(144(220)161-77(16)120(196)169-93(148(224)225)41-30-33-51-151)182-127(203)89(40-29-32-50-150)165-136(212)103-66-228-80(19)117-146(222)180-105(138(214)174-99(59-109(155)190)133(209)179-103)68-229-79(18)116(145(221)175-97(56-72(9)10)134(210)184-117)183-128(204)91(44-47-108(154)189)167-130(206)96(55-71(7)8)172-132(208)98(57-82-35-24-23-25-36-82)173-137(213)104-67-227-78(17)115(142(218)158-61-110(191)159-75(14)119(195)170-94(53-69(3)4)129(205)166-90(43-46-107(153)188)126(202)163-86(22-2)123(199)178-104)185-143(219)113(73(11)12)181-140(216)102-65-230-81(20)118(147(223)187-52-34-42-106(187)141(217)157-62-111(192)162-102)186-139(215)101-64-226-63-100(135(211)168-92(45-48-112(193)194)124(200)160-76(15)121(197)171-95(54-70(5)6)131(207)177-101)176-125(201)88(39-28-31-49-149)164-122(198)85(152)58-83-60-156-87-38-27-26-37-84(83)87/h22-27,35-38,60,69-75,78-81,85,88-106,113-118,156H,15-16,21,28-34,39-59,61-68,149-152H2,1-14,17-20H3,(H2,153,188)(H2,154,189)(H2,155,190)(H,157,217)(H,158,218)(H,159,191)(H,160,200)(H,161,220)(H,162,192)(H,163,202)(H,164,198)(H,165,212)(H,166,205)(H,167,206)(H,168,211)(H,169,196)(H,170,195)(H,171,197)(H,172,208)(H,173,213)(H,174,214)(H,175,221)(H,176,201)(H,177,207)(H,178,199)(H,179,209)(H,180,222)(H,181,216)(H,182,203)(H,183,204)(H,184,210)(H,185,219)(H,186,215)(H,193,194)(H,224,225)/b86-22-
InChI Key
WPLOVIFNBMNBPD-ATHMIXSHSA-N
Canonical SMILES
CCC(C)C(C(=O)NC(=C)C(=O)NC(CCCCN)C(=O)O)NC(=O)C(CCCCN)NC(=O)C1CSC(C2C(=O)NC(CSC(C(C(=O)NC(C(=O)N2)CC(C)C)NC(=O)C(CCC(=O)N)NC(=O)C(CC(C)C)NC(=O)C(CC3=CC=CC=C3)NC(=O)C4CSC(C(C(=O)NCC(=O)NC(C(=O)NC(C(=O)NC(C(=O)NC(=CC)C(=O)N4)CCC(=O)N)CC(C)C)C)NC(=O)C(C(C)C)NC(=O)C5CSC(C(C(=O)N6CCCC6C(=O)NCC(=O)N5)NC(=O)C7CSCC(C(=O)NC(C(=O)NC(=C)C(=O)NC(C(=O)N7)CC(C)C)CCC(=O)O)NC(=O)C(CCCCN)NC(=O)C(CC8=CNC9=CC=CC=C98)N)C)C)C)C(=O)NC(C(=O)N1)CC(=O)N)C
1. LanI-Mediated Lantibiotic Immunity in Bacillus subtilis: Functional Analysis
Christoph Geiger, Sophie Marianne Korn, Michael Häsler, Oliver Peetz, Janosch Martin, Peter Kötter, Nina Morgner, Karl-Dieter Entian Appl Environ Microbiol. 2019 May 16;85(11):e00534-19. doi: 10.1128/AEM.00534-19. Print 2019 Jun 1.
Lantibiotics subtilin and nisin are produced by Bacillus subtilis and Lactococcus lactis, respectively. To prevent toxicity of their own lantibiotic, both bacteria express specific immunity proteins, called SpaI and NisI. In addition, ABC transporters SpaFEG and NisFEG prevent lantibiotic toxicity by transporting the respective peptides to the extracellular space. Although the three-dimensional structures of SpaI and NisI have been solved, very little is known about the molecular function of either lipoprotein. Using laser-induced liquid bead ion desorption (LILBID)-mass spectrometry, we show here that subtilin interacts with SpaI monomers. The expression of either SpaI or NisI in a subtilin-nonproducing B. subtilis strain resulted in the respective strain being more resistant against either subtilin or nisin. Furthermore, pore formation provided by subtilin and nisin was prevented specifically upon the expression of either SpaI or NisI. As shown with a nisin-subtilin hybrid molecule, the C-terminal part of subtilin but not any particular lanthionine ring was needed for SpaI-mediated immunity. With respect to growth, SpaI provided less immunity against subtilin than is provided by the ABC transporter SpaFEG. However, SpaI prevented pore formation much more efficiently than SpaFEG. Taken together, our data show the physiological function of SpaI as a fast immune response to protect the cellular membrane.IMPORTANCE The two lantibiotics nisin and subtilin are produced by Lactococcus lactis and Bacillus subtilis, respectively. Both peptides have strong antimicrobial activity against Gram-positive bacteria, and therefore, appropriate protection mechanisms are required for the producing strains. To prevent toxicity of their own lantibiotic, both bacteria express immunity proteins, called SpaI and NisI, and in addition, ABC transporters SpaFEG and NisFEG. Whereas it has been shown that the ABC transporters protect the producing strains by transporting the toxic peptides to the extracellular space, the exact mode of action and the physiological function of the lipoproteins during immunity are still unknown. Understanding the exact role of lantibiotic immunity proteins is of major importance for improving production rates and for the design of newly engineered peptide antibiotics. Here, we show (i) the specificity of each lipoprotein for its own lantibiotic, (ii) the specific physical interaction of subtilin with its lipoprotein SpaI, (iii) the physiological function of SpaI in protecting the cellular membrane, and (iv) the importance of the C-terminal part of subtilin for its interaction with SpaI.
2. Specificity of Subtilin-Mediated Activation of Histidine Kinase SpaK
Christoph Geiger, Tobias Spieß, Sophie Marianne Korn, Peter Kötter, Karl-Dieter Entian Appl Environ Microbiol. 2017 Aug 31;83(18):e00781-17. doi: 10.1128/AEM.00781-17. Print 2017 Sep 15.
Autoinduction via two-component systems is a widespread regulatory mechanism that senses environmental and metabolic changes. Although the lantibiotics nisin and subtilin are closely related and share the same lanthionine ring structure, they autoinduce their biosynthesis in a highly specific manner. Subtilin activates only the two-component system SpaRK of Bacillus subtilis, whereas nisin activates solely the two-component system NisRK of Lactococcus lactis To identify components that determine the specificity of subtilin autoinduction, several variants of the respective lantibiotics were analyzed for their autoinductive capacities. Here, we show that amino acid position 20 is crucial for SpaK activation, as an engineered nisin molecule with phenylalanine at position 20 (nisin N20F) was able to activate SpaK in a specific manner. In combination with the N-terminal tryptophan of subtilin (nisin I1W/N20F), SpaK autoinduction reached almost the level of subtilin-mediated autoinduction. Furthermore, the overall structure of subtilin is also important for its association with the histidine kinase. The destruction of the second lanthionine ring (subtilin C11A, ring B), as well as mutations that interfere with the flexibility of the hinge region located between lanthionine rings C and D (subtilin L21P/Q22P), abolished SpaK autoinduction. Although the C-terminal part of subtilin is needed for efficient SpaK autoinduction, the destruction of lanthionine rings D and E had no measurable impact. Based on these findings, a model for the interaction of subtilin with histidine kinase SpaK was established.IMPORTANCE Although two-component systems are important regulatory systems that sense environmental changes, very little information on the molecular mechanism of sensing or the interaction of the sensor with its respective kinase is available. The strong specificity of linear lantibiotics such as subtilin and nisin for their respective kinases provides an excellent model system to unravel the structural needs of these lantibiotics for activating histidine kinases in a specific manner. More than that, the biosyntheses of lantibiotics are autoinduced via two-component systems. Therefore, an understanding of their interactions with histidine kinases is needed for the biosynthesis of newly engineered peptide antibiotics. Using a Bacillus subtilis-based reporter system, we were able to identify the molecular constraints that are necessary for specific SpaK activation and to provide SpaK specificity to nisin with just two point mutations.
3. Regulation of heterologous subtilin production in Bacillus subtilis W168
Qian Zhang, Carolin M Kobras, Susanne Gebhard, Thorsten Mascher, Diana Wolf Microb Cell Fact. 2022 Apr 7;21(1):57. doi: 10.1186/s12934-022-01782-9.
Background: Subtilin is a peptide antibiotic (lantibiotic) natively produced by Bacillus subtilis ATCC6633. It is encoded in a gene cluster spaBTCSIFEGRK (spa-locus) consisting of four transcriptional units: spaS (subtilin pre-peptide), spaBTC (modification and export), spaIFEG (immunity) and spaRK (regulation). Despite the pioneer understanding on subtilin biosynthesis, a robust platform to facilitate subtilin research and improve subtilin production is still a poorly explored spot. Results: In this work, the intact spa-locus was successfully integrated into the chromosome of Bacillus subtilis W168, which is the by far best-characterized Gram-positive model organism with powerful genetics and many advantages in industrial use. Through systematic analysis of spa-promoter activities in B. subtilis W168 wild type and mutant strains, our work demonstrates that subtilin is basally expressed in B. subtilis W168, and the transition state regulator AbrB strongly represses subtilin biosynthesis in a growth phase-dependent manner. The deletion of AbrB remarkably enhanced subtilin gene expression, resulting in comparable yield of bioactive subtilin production as for B. subtilis ATCC6633. However, while in B. subtilis ATCC6633 AbrB regulates subtilin gene expression via SigH, which in turn activates spaRK, AbrB of B. subtilis W168 controls subtilin gene expression in SigH-independent manner, except for the regulation of spaBTC. Furthermore, the work shows that subtilin biosynthesis in B. subtilis W168 is regulated by the two-component regulatory system SpaRK and strictly relies on subtilin itself as inducer to fulfill the autoregulatory circuit. In addition, by incorporating the subtilin-producing system (spa-locus) and subtilin-reporting system (PpsdA-lux) together, we developed "online" reporter strains to efficiently monitor the dynamics of subtilin biosynthesis. Conclusions: Within this study, the model organism B. subtilis W168 was successfully established as a novel platform for subtilin biosynthesis and the underlying regulatory mechanism was comprehensively characterized. This work will not only facilitate genetic (engineering) studies on subtilin, but also pave the way for its industrial production. More broadly, this work will shed new light on the heterologous production of other lantibiotics.
Online Inquiry
Verification code
Inquiry Basket