Unusual biosynthesis and structure of locillomycins from Bacillus ...

2 downloads 17 Views 1MB Size Report
Jul 10, 2015 - bSchool of Life Science and Chemical Engineering, Huaiyin Institute of Technology, ..... families of lipopeptides are key factors in antagonism of Bacillus subtilis toward .... Peng C, Pu J, Song L, Jian X, Tang M, Tang G. 2012.
AEM Accepted Manuscript Posted Online 10 July 2015 Appl. Environ. Microbiol. doi:10.1128/AEM.01639-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

2

Unusual biosynthesis and structure of locillomycins from Bacillus subtilis 916

3

Chuping Luo a,b*, Xuehui Liuc*, Xian Zhoud, Junyao Guo a, John Truongd, Xiaoyu Wanga ,

4

Huafei Zhou a , Xiangqian Li*b, Zhiyi , Chena*

5

a

6

China

7

b

8

223003, China

9

c

10

d

11

2751, Australia

12

Corresponding Authors:

13

*Chuping Luo, E-mail: [email protected], Tel: +86-0517-89221726,

14

*Xuehui Liu, E-mail: [email protected], Tel: +86-010-64887727

15

*Xiangqian Li, E-mail: [email protected], Tel: +86-0517-89221726

16

*Zhiyi Chen, E-mail: [email protected] , Tel: +86-025-84390393.

1

Institute of Plant Protection, Jiangsu Academy of Agricultural Sciences, Nanjing 210014,

School of Life Science and Chemical Engineering, Huaiyin Institute of Technology, Huaian

Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China The National Institute of Complementary Medicine, University of Western Sydney, Sydney

17

1

18

ABSTRACT: Three families of Bacillus cyclic lipopeptides – surfactins, iturins and fengycins –

19

have well-recognized potential uses in biotechnology and biopharmaceutical applications. This

20

study outlines the isolation and characterization of locillomycins, a novel family of cyclic

21

lipopeptide produced by B. subtilis 916. Structure elucidation of locillomycins revealed several

22

molecular features not observed in other Bacillus lipopeptides, including a unique nonapeptide

23

sequence and macrocyclization. Locillomycins are active against bacteria and viruses.

24

Biochemical analysis and gene deletions studies have supported the assignment of a 38 kb gene

25

cluster as the locillomycins biosynthetic gene cluster. Interestingly, this gene cluster encodes 4

26

proteins (namely LocA, LocB, LocC and LocD) using a hexamodular nonribosomal peptide

27

synthetase to biosynthesize cyclic nonapeptides. Genome analysis and the chemical structures of

28

the end products indicated that the biosynthetic pathway exhibited two distinct features: (i) a

29

non-linear hexamodular assembly line with three modules in the middle utilized twice while the

30

first and last two modules were used only once; and (ii) several domains are skipped or

31

optionally selected.

32

2

33

In the competition for nutrients, members of the Bacillus genus often produce a vast array

34

of biologically active molecules that potentially inhibit the development of competing organisms.

35

The gram-positive bacterium Bacillus subtilis has an average of 4 – 5% of its genome devoted to

36

antibiotic synthesis and is able to produce more than two dozen antibiotics with an amazing

37

variety of structures.(1) Many of these compounds, which have a peptide origin, are synthesized

38

either ribosomally or non-ribosomally. Among the non-ribosomally generated amphipathic

39

cyclic lipopeptides, surfactins, iturins and fengycins have well-recognized potential applications

40

in biotechnology and biopharmaceutical products due to their antagonistic activities and

41

surfactant properties.(2, 3) Furthermore, the mechanisms behind the observed biocontrol efficacy

42

of the different Bacillus strains have also been well described.(4–6) Lipopeptides are able to

43

induce systemic resistance in plants and facilitate the multicellular behaviors of the producing

44

strains such as swarming motility, biofilm formation and colony morphology.(5–7)

45

Surfactins, iturins and fengycins are synthesized by nonribosomal peptide synthetases

46

(NRPSs) which exhibit a distinct modular architecture.(2, 8–10) A module is typically composed

47

of three core domains with each domain responsible for a certain biochemical reaction.(11)

48

Specifically, the amino acid adenylation domain (A domain) controls the entry of the substrates

49

into the peptide by recognizing and activating a specific amino acid. The thiolation domain (T

50

domain), also referred to as the peptidyl carrier protein (PCP), contains an invariant serine

51

residue which is essential for the binding of a 4'-phosphopantetheine cofactor. An N-terminal

3

52

condensation domain (C domain) is required for the coupling of two consecutively bound amino

53

acids.(12, 13) These three domains constitute a minimal elongation module, the basic repetitive

54

unit of a multi-modular NRPS. Furthermore, modules can be supplemented with domains that

55

catalyze the modification of the activated amino acid, such as N-methylation and epimerization.

56

In some cases, when the first module of a NRPS complex lacks a C-domain, the last module

57

contains a termination thioesterase domain (TE domain) to release the end product.(14) Order

58

and specificity of the modules within the protein template determines the sequence of the product

59

(type A, linear NRPS).(8, 11) Genetic and biochemical analyses have revealed that the modular

60

arrangement of most lipopeptide synthetases is colinear with the amino acid sequences of

61

lipopeptides.(1, 2) This assembly-line arrangement of the conserved catalytic modules and

62

domains provide the means to construct hybrid NRPS used to synthesize new lipopeptide

63

compounds.(15–18) The prospect of creating numerous bioactive lipopeptides by engineering

64

existing lipopeptide synthetases has stimulated the search for new NRPS responsible for

65

lipopeptide synthesis.(19–24)

66

To date, only two reported kinds of biosynthetic machinery within the NRPS assembly line

67

do not conform to the rule of colinearity. These include the type B and type C NRPS which

68

iteratively use all of their modules or certain domains during the assembly of a product,

69

respectively.(25–28) While our understanding of nonlinear NRPS biosynthetic mechanism is

70

limited, it is clear that this mechanism has great potential to introduce structural diversity to the

4

71

secondary metabolites through combinatorial biosynthesis.(25, 33, 36) It is necessary to propose

72

nonlinear NRPS biosynthetic models that can further unravel the details of this biosynthetic

73

mechanism. The gram-positive bacterium B. subtilis serves as a model organism and is

74

intensively used in the heterologous expression of commercial metabolites.(36–38) However,

75

apart from this study, no non-linear NRPS assembly line has been observed in this well-

76

characterized strain.

77

The commercial strain of B. subtilis 916 was isolated from paddy soils of Jurong County,

78

Jiangsu and has been reported to be effective in the biocontrol of plant diseases (39). While

79

NRPSs for LPs production in B. subtilis 916 have been reported recently and the locillomycin

80

also described briefly (40), in this study, we describe in detail how to elucidate the novel

81

structure and unique biosynthesis of this new family of nonribosomal lipopeptides locillomycins

82

and its biological function. In this study, our results strongly suggest that locillomycins revealed

83

molecular features not observed in the three families of surfactins, iturins and fengycins,

84

including a unique nonapeptide sequence and macrocyclization. Locillomycins are active against

85

bacteria and viruses with low hemolytic activity and can be used for therapeutic applications as

86

natural health products. While the end products were straightforward, the proposed biosynthetic

87

pathway of locillomycins contained several atypical aspects that were not predictable by

88

bioinformatics analysis. Our study thus adds to the knowledge regarding non-linear NRPS

5

89

biosynthesis used by B. subtilis and further broadens the potential of strain 916 as a model

90

system for nonlinear NRPS studies.

91

6

92

MATERIALS AND METHODS

93

Cultivation, Isolation and Purification. Luria-Bertani (LB) broth was inoculated with B.

94

subtilis 916 to a concentration of 5.5 × 105 cells/mL. One hundred cultures of 330 mL each in 2-

95

liter Erlenmeyer flasks were incubated with 180 rpm agitation at 28 ºC for three days. After

96

removing the biomass by centrifugation, the broth was titrated to pH 2.8 with concentrated

97

hydrochloric acid and the resulting grey precipitate was extracted with 150 mL MeOH. The

98

MeOH extraction was added with 300 mL H2O and titrated to pH 7 with 5 M NaOH. The

99

extracted impurities were added to an Agilent amino solid phase extraction column and

100

sequentially washed with 50% (v/v) MeOH-H2O, 100% MeOH, 1% (v/v) formic acid-MeOH

101

and 2% (v/v) formic acid-MeOH. The 2% (v/v) formic acid-MeOH elution was concentrated by

102

nitrogen drying to 10 mg/mL and loaded on an Agilent C18 solid phase extraction column and

103

sequentially washed with 40, 42, 46, and 48% (v/v) MeOH-H2O. The 44, 46, and 48% (v/v)

104

MeOH-H2O elutions were concentrated by nitrogen drying to 20 mg/mL. These fractions were

105

further processed by HPLC by repetitive 50 µL injections using a reversed-phase column (RP-

106

18, 5 µM, 4 × 250 mm; Merck) with a 0.5 mL/min flow rate and monitored at 230 nm. Fraction I

107

yielded derivatives A which was collected at an average peak retention time of 9 min. Fraction II

108

yielded derivatives B at a retention time of 13 min. Fraction III yielded derivatives C at a

109

retention time of 18 min. Both eluted with 50% (v/v) CH3CN, 0.5% (v/v) trifluoroacetic acid in

7

110

H2O. All collections were concentrated by nitrogen and vacuum drying and resulted in the

111

purified locillomycin A, B and C derivatives.

112

NMR Structure Determination. All NMR spectra were acquired at 25 ºC on a Bruker

113

AVANCE III 600MHz, an Agilent DD2 500MHz (for 13C detected spectra) or an Agilent DD2

114

600MHz spectrometer equipped with a cold-probe. Locillomycin A (~8 mg) were dissolved in

115

500 µL aqueous solvent (20 mM phosphate buffer, pH 6.5, D2O/H2O, (9:1, v/v)) while ~8 mg of

116

Locillomycin B and C respectively was dissolved in 500 µL CD3OH to make the NMR samples.

117

For each sample, a series of 1D and 2D spectra, including 1H, 13C, DEPT, 1H-1H COSY, 1H-1H

118

TOCSY, 1H-1H ROESY, 1H-13C HSQC, 1H-13C HMBC, were acquired for the structure

119

elucidation. The spin-lock time for each TOCSY experiment was 80 ms and the mixing time for

120

each ROESY was 200 ms. All NMR data were processed and analyzed with MestReNova. The

121

chemical shifts in the aqueous solvent were referenced to the HOD peak at 4.77 ppm while those

122

in the CD3OH were referenced to the peak of the methyl group at 3.30 ppm. Chemical shifts of

123

13

124

for the peptide part was done using the protocol developed by Wuethrich.(46)

125

Gene Disruption in B. subtilis 916 and mutant analysis. In vivo generation of targeted

126

mutations in B. subtilis 916 were achieved by a modified protocol originally developed for B.

127

subtilis 168.9 The knockout plasmids were constructed from vector pUC19 by inserting

128

homologous fragments from B. subtilis 916 genomic DNA and a neomycin cassette from

C were referenced indirectly(48) or to the peak of CD3OH at 49 ppm. Sequential assignment

8

129

pBEST501. An example for disruption of LocD (ΔlocD) is detailed below. A 2.5 kb fragment

130

was amplified by PCR using the primers LocDF and LocDR and cloned into vector pUC19,

131

generating pLocD1. The latter was digested with xbaI, which cuts in the middle of the PCR

132

fragment. Simultaneously, pBEST501 with the same enzyme to obtain the neomycin cassette

133

(~1.3 kb), which was then ligated to pLocD1, resulting in pLocD2. This was subsequently

134

transformed into the naturally competent B. subtilis 916, where it was introduced into the

135

genome via double-crossover homologous recombination. The disruption locD gene was

136

demonstrated in the resistant colonies obtained by PCR with appropriate primers and by

137

Southern hybridization. The locillomycins were extracts from mutants and further analyzed by

138

HPLC/MS as described previously.

139

Heterologous expression and purification of internal adenylation domains. The pET28a

140

expression system was used to clone PCR products in which Nco I and Xho I sites were

141

introduced to by the PCR with 5’-modified primers (Table 1). PCR from chromosomal B. subtilis

142

916 was carried out with 50 µL reaction volumes containing 2.5 U

143

µL 10× Mg-free reaction buffer, 200 µM dNTP, 2.5 mM MgCl2 and 1 µM each of primers.

144

Amplification conditions were: 30 cycles of 95 °C for 30 s, 55 °C for 30 s and 72°C for 3 min,

145

followed by a final extension at 72 °C for 10 min. PCR products were digested with NcoI and

146

XhoI, and cloned into the digested pET28a vector. PCR products cloned into the XhoI site of

147

pET28a results in appending the poly-Histag at the carboxyl end of recombinant proteins.

EXTaq

DNA polymerase, 5

9

148

Transformants of E. coli BL21(DE3) were selected by growth at 28 °C in LB media containing

149

100 µg/mL ampicillin. Cells were induced with 1mM IPTG at an OD600 of 0.6 and allowed to

150

grow for an ad

151

ditional 4 h before being harvested. Purification of the His6-tagged proteins was carried out by

152

Ni2+-affinity chromatography. They were separated using 10% (w/v) SDS–PAGE, stained with

153

Coomassie blue, and analyzed by density scanning using an image analysis system (Bio-Rad).

154

Pyrophosphate Exchange Assay. Amino acid-dependent ATP-sodium pyrophosphate assays

155

were performed by using the spectrophotometric assay furnished by the EnzChek Pyrophosphate

156

Assay Kit (Molecular Probes). A 200 µL reaction contained 75 mM Tris•HCl (pH 7.5), 10 mM

157

MgCl2, 5 mM DTT, 5 mM ATP, 400 mM MesG, 0.2 units purine nucleoside phosphorylase, and

158

0.2 units inorganic pyrophosphatase, 2 mM amino acid. Reaction without amino acid was as the

159

control. Reaction was initiated by addition of enzyme and incubated at 30 °C for 30 min.

160

Reading of absorbance at 360 nm was measured in a Perkin-Elmer Lambda 6-vis

161

spectrophotometer. All reactions were performed in triplicate.

162

Nucleotide sequence accession number. The complete genome sequence of B. subtilis 916 and

163

the gene cluster for biosynthesis locillomycins described in this work have been submitted to

164

GenBank under accession numbers CP009611 and KF866134, respectively.

165

RESULTS AND DISCUSSION

10

166

Identification and in vivo gene disruption of loc gene cluster. Identification and

167

bioinformatics analyses of the loc gene cluster were employed as described in our previous work.

168

(40) The draft genome and complete genome sequence of B. subtilis 916 was recently analyzed

169

(GenBank no. AFSU00000000.1 and CP009611) and four NRPS gene clusters were identified

170

(Figure S1).(41) In addition to the three conventional gene clusters, srf (for surfactins), bmy (for

171

bacillomycin Ls) and fen (for fengycins), the B. subtilis 916 also contains a fourth NRPSs gene

172

cluster, loc (GenBank no. KF866134) (40). Further analysis of the loc open reading frames led to

173

the conclusion that the loc gene cluster was a potential candidate for the biosynthesis of an

174

unknown family of lipopepetides called locillomycins. Interestingly, this particular locus in B.

175

subtilis 916 shares a high degree of homology (>95% identity) with the same locus found in B.

176

subtilis 168 and B. amyloquefaciens FZB42 strains (Figure 1).

177

Four NRPS genes designated locD, A, B and C are contiguous in the loc gene cluster and

178

correspond to the six peptide extension modules (Figure 1). The first gene, locD, encodes a 145.8

179

kDa protein containing the polyketide synthase module, and includes domains with homology to

180

the proteins involved in the synthesis of fatty acids and polyketides: fatty acid acyl-CoA

181

synthetase domain (ACS), acyl carrier protein/thiolation domain (T) and β-keto acyl synthetase

182

domain (KAS). The second gene, locA, encodes a 267.4 kDa protein with only one extension

183

module containing the core elongation domains: C domain, A domain and T domain. Two C

184

domains exist independently in the upstream of the A domain. The third gene, locB, encodes a

11

185

443.1 kDa protein containing three extension modules. The first module contains a predicted

186

epimerization (E) domain, suggesting that the corresponding amino acid appears in the D-

187

configuration. The fourth gene, locC, encoding a 243.9 kDa protein, contains two extension

188

modules and a termination module, which contains a C-terminal TE domain. A similarity

189

analysis also showed that the nucleotide sequence of loc gene cluster has a low similarity ( B > A) ( Table S3 – S5 and Figure S6 – S8). This

251

difference is derived from the different lengths of the long-chain acyl group shown in Figure S10

252

and the method of structural determination will not be detailed here.

253

Unlike the surfactin and iturin lipopeptide families, in which the peptide residues are

254

interlinked with a β-hydroxy or a β-amino fatty acid to form a cyclic lactone or lactam, the

255

locillomycins form an internal lactone ring within the peptidic moiety. This construction is more

256

similar to the fengycin family which also forms a lactone without the participation of a β-

257

hydroxyl fatty acid. Importantly, however, the number of residues in locillomycins is nine rather

258

than ten as observed in the fengycin family (eight out of ten residues form the lactone ring)

259

(Figure 4). These nine-membered cyclic lipopeptides are also different from the iturin and

260

surfactin families, which are seven-membered rings. Furthermore, only one D-amino acid (D-

261

Gln2) is incorporated in the locillomycin family, while all the members of the other three

15

262

families possess more than two D-amino acid residues. The length of the fatty acid chains is

263

similar to other Bacillus lipopeptides.

264

Biochemical investigation of internal adenylation domains. To further elucidate the

265

biosynthetic process of locillomycin, DNA fragments coding the adenylation domains of

266

modules LocA1 (A1), LocB1 (A2), LocB2 (A3), LocB3 (A4), LocC1 (A5), LocC2 (A6) (Figure

267

6) were amplified from chromosomal B. subtilis 916 DNA and cloned into IPTG-inducible

268

expression as described in Experimental section. The constructs were confirmed by sequencing

269

fusion sites. The enzyme fragments were overexpressed in Escherichia coli (E. coli) as His6-

270

tagged proteins and purified by Ni2+-affinity chromatography. All proteins were found within the

271

soluble fraction after French press lysis of E. coli cells, as confirmed from Coomassie blue-

272

stained SDS-polyacrylamide gels (Figure 5A) with calculated molecular masses of 60.0 to 65.0

273

kDa. The purified adenylation domains were biochemically investigated with respect to their

274

activity and specificity in the ATP-PPi-exchange reaction. In order to determine amino acid

275

specificity, each protein was incubated with all constituent amino acids of locillomycins (control

276

without amino acid). The six adenylation domains were found to activate the amino acids

277

corresponding to the constituent amino acids of locillomycins. Specifically, A1 exclusively

278

activated L-Thr; A2 activated L-Gln (100%), L-Asn (80%) and L-Asp (10%); A3 activated L-

279

Asp (100%), L-Asn (15%) and L-Gln (10%); A4 exclusively activated Gly; A5 activated L-Tyr

280

(100%) and L-Val (15%); and A6 activated L-Val (100%) and L-Asn (10%). (The highest

16

281

activity was set at 100%, Fig. 5B) A1, the only adenylation domain of LocA, exclusively

282

activated L-Thr and is likely to be involved in the position 1 amino acid biosynthesis. A2, A3

283

and A4, the three adenylation domains of LocB, are likely to iteratively act to incorporate the

284

2nd-7th amino acids. Particularly, the A2 was able to efficiently activate both L-Gln and L-Asn.

285

A5 and A6, the last two adenylation domains of LocC, optimally activated L-Tyr and L-Val,

286

respectively, which is in agreement with the last two amino acids of locillomycins.

287

Proposed locillomycins biosynthesis pathway. The gene cluster for locillomycin

288

biosynthesis that was identified in this study sets the stage for further delineating the intricate

289

chemical assembly of this family of lipopeptide antibiotics. Based on the chemical structures of

290

locillomycins, the analysis of in vivo gene disruptions, internal adenylation domains experiments,

291

amino acid sequence homology of loc proteins to enzymes and previous knowledge of

292

lipopeptide biosynthetic pathways led to the proposal of a locillomycins biosynthetic pathway

293

(Figure 6). In brief, it’s a pathway where a cyclic lipo-nonapeptide is assembled by

294

multifunctional enzymes of a hexamodular NRPS. In the initial step, the ACS domain couples

295

coenzymeA (CoA) to a long-chain fatty acid, presumably tridecanoic to pentadecanoic, in an

296

ATP-dependent reaction. The activated fatty acid is then transferred to the 4-phosphopantetheine

297

cofactor of the T domain. In the second step, the fatty acid is coupled to the activated threonine

298

directly, catalyzed by one of the condensation domains preceding the A1 domain of the peptide

299

synthetases, with the KAS, two T domains and one C domain skipped (Figure 6). In subsequent

17

300

condensation reactions, the donor part containing the fatty acid and thiolated threonine is

301

connected to the acceptor part containing the A2 domain activated glutamine which is

302

epimerized by the E domain following the A2 before the next extension step (Figure 6). The

303

process continues in a canonical fashion until the A4 domain activated glycine is connected.

304

When this occurs, the whole LocB is used a second time for further extension (Figure 6). This

305

time the A2 activates asparagine rather than glutamine, which is connected to the donor without

306

epimerization. Extension then proceeds through LocB and LocC all the way down to the TE

307

domain, which catalyzes the cyclization of the synthesized linear peptide by connecting the

308

carbonyl group of Val9 and the hydroxyl group of Thr1 sidechain, before releasing the mature

309

cyclic lipopeptide of locillomycins (Figure 6).(47) Particular emphasis should be noted that the

310

first A domain (A1) and the last two A domains (A5, A6) were used only once, whereas the three

311

A domains (A2, A3, A4) in the middle were iteratively used during the biosynthesis (Figure 6).

312

Moreover, the A2 domain in LocB somehow exhibits different specificity in the iteration. Also,

313

the epimerization domain following the A2 domain which is responsible for conversion of L-Gln

314

to D-Gln in the first time is skipped in the second time (Figure 6). The different selectivity of

315

these catalytic domains within the same NRPS assembly line is quite unusual compared to other

316

NRPS assembly lines. Thus, compared to the iterative uses in type B and C NRPS, loc NRPS

317

was designated as a type D NRPS(Figure 6).(25–28)

18

318

Typical lipopeptide biosynthesis involves the KAS domain catalyzing the formation of a β-

319

ketoacyl thioester, which is subsequently converted into a β-substituted fatty acid by a

320

transamination or transhydoxylation reaction. This is followed by a transfer of the fatty acid to a

321

T domain and is coupled to the first activated amino acid. In contrast, with regards to the

322

synthesis of locillomycin, only the ACS domain, not the KAS domain, is responsible for the

323

activation of the fatty acid. The reason for skipping the KAS domain may be due to the lack of

324

any functional domains such as amino transferase or hydroxyl transferase in the downstream of

325

the KAS domain. The function of the redundant T and C domains in the front of the LocA are

326

unclear since one condensation domain preceding the first A domain of the peptide synthetase is

327

enough to couple the fatty acid to the first activated amino acid.(2) While a few of these atypical

328

biochemical features of single module or multiple modules iteration have been previously

329

observed, the locillomycins represent a rare nonlinear NRPS biosynthetic model with a

330

combination of iteration, skipping, alternating and specificity changes (Figure 6).

331

Biological functions of locillomycin. Biological activity assays revealed that locillomycins

332

have moderate antibacterial activities (Figure S11). The antibacterial minimum inhibitory

333

concentrations (MICs) of locillomycin A, B and C were 24.3, 18.8, and 17.6 µg/mL, respectively,

334

for methicillin-resistant Staphylococcus aureus (MRSA), and were 6.3, 5.8, and 5.4 µg/mL,

335

respectively, for Xanthomonas oryzae pv. oryzae. The antiviral results at different locillomycins

336

concentrations revealed that the Porcine Epidemic Diarrhea Virus (PEDV) infection could be

19

337

effectively inhibited using the compound. Particularly, at the concentration of 10 µg/mL,

338

locillomycins reduced the virus copies by 300 fold and was about ten times as efficient as

339

surfactins (Figure S12). Locillomycins also have lower hemolytic activities than surfactins and

340

are presumed to reduce toxicity against eukaryotic cells, which could improve their possible

341

therapeutic application (Figure S12).(3)

342

20

343

Acknowledgements

344

This work was supported by the National Natural Science Foundation of China (grants

345

no.30900929), National High-tech R&D Program of China (2011AA10A201), and Jiangsu

346

Independent Innovation Fund (CX(12)5001) . We are also grateful to L. Du (Department of

347

Chemistry, University of Nebraska-Lincoln) for his critical review and revision.

348

Author Contributions

349

The authors declare no competing financial interest. The manuscript was written through

350

contributions of all authors. All authors have given approval to the final version of the

351

manuscript. §Chuping Luo and Xuehui Liu contributed equally to this work.

352 353

21

354

References

355 356

1.

Stein T. 2005. Bacillus subtilis antibiotics: structures, syntheses and specific functions. Molecular microbiology 56:845–857.

357 358

2.

Ongena M, Jacques P. 2008. Bacillus lipopeptides: versatile weapons for plant disease biocontrol. Trends in microbiology 16:115–125.

359 360

3.

Rodrigues L, Banat IM, Teixeira J, Oliveira R. 2006. Biosurfactants: potential applications in medicine. The Journal of antimicrobial chemotherapy 57:609–618.

361 362 363 364

4.

Romero D, de Vicente A, Rakotoaly RH, Dufour SE, Veening JW, Arrebola E, Cazorla FM, Kuipers OP, Paquot M, Pérez-García A. 2007. The iturin and fengycin families of lipopeptides are key factors in antagonism of Bacillus subtilis toward Podosphaera fusca. Molecular plant-microbe interactions: MPMI 20:430–440.

365 366 367

5.

Ongena M, Jourdan E, Adam A, Paquot M, Brans A, Joris B, Arpigny JL, Thonart P. 2007. Surfactin and fengycin lipopeptides of Bacillus subtilis as elicitors of induced systemic resistance in plants. Environmental microbiology 9:1084–1090.

368 369 370

6.

Zeriouh H, de Vicente A, Pérez-García A, Romero D. 2014. Surfactin triggers biofilm formation of Bacillus subtilis in melon phylloplane and contributes to the biocontrol activity. Environmental microbiology 16:2196–2211.

371 372 373 374

7.

Han Q, Wu F, Wang X, Qi H, Shi L, Ren A, Liu Q, Zhao M, Tang C. 2014. The bacterial lipopeptide iturins induce Verticillium dahliae cell death by affecting fungal signalling pathways and mediate plant defence responses involved in pathogen-associated molecular pattern-triggered immunity. Environmental microbiology 17:1166-1188.

375 376 377 378 379 380

8.

Duitman EH, Hamoen LW, Rembold M, Venema G, Seitz H, Saenger W, Bernhard F, Reinhardt R, Schmidt M, Ullrich C, Stein T, Leenders F, Vater J. 1999. The mycosubtilin synthetase of Bacillus subtilis ATCC6633: A multifunctional hybrid between a peptide synthetase , an amino transferase , and a fatty acid synthase. Proceedings of the National Academy of Sciences of the United States of America 96:13294–13299.

381 382

9.

Shoda M. 2001. Cloning , sequencing , and characterization of the iturin A operon. Journal of bacteriology 183:6265–6273.

22

383 384 385

10.

Tosato V, Albertinij AM, Zotti M, Sondal S, Bruschi CV. 1997. Sequence completion , identification and definition of the fengycin operon in Bacillus subtilis 168. Microbiology 143:3443–3450.

386 387 388 389

11.

Kessler N, Schuhmann H, Morneweg S, Linne U, Marahiel MA. 2004. The linear pentadecapeptide gramicidin is assembled by four multimodular nonribosomal peptide synthetases that comprise 16 modules with 56 catalytic domains. The Journal of biological chemistry 279:7413–7419.

390 391

12.

Role C, The OF, Domain C. 1998. Peptide Bond Formation in Nonribosomal Peptide Biosynthesis. The Journal of biological chemistry 273:22773–22781.

392 393 394

13.

Belshaw PJ, Walsh CT, Stachelhaus T. 1999. Aminoacyl-CoAs as probes of condensation domain selectivity in nonribosomal peptide synthesis. Science (New York, N.Y.) 284:486–489.

395 396

14.

Sieber SA, Marahiel MA. 2003. Learning from Nature’s Drug Factories: Nonribosomal Synthesis of Macrocyclic Peptides. Journal of bacteriology 185:7036–7043.

397 398 399

15.

Mootz HD, Schwarzer D, Marahiel MA. 2000. Construction of hybrid peptide synthetases by module and domain fusions. Proceedings of the National Academy of Sciences of the United States of America 97:5848–5853.

400 401 402 403

16.

Hoe BC, Gorzelnik KV, Yang JY, Hendricks N, Dorrestein PC. 2012. Enzymatic resistance to the lipopeptide surfactin as identified through imaging mass spectrometry of bacterial competition. Proceedings of the National Academy of Sciences of the United States of America 109:13082–13087.

404 405

17.

Zaman M, Toth I. 2013. Immunostimulation by Synthetic Lipopeptide-Based Vaccine Candidates: Structure-Activity Relationships. Frontiers in immunology 4:1–12.

406 407

18.

Sieber SA, Marahiel MA. 2005. Molecular mechanisms underlying nonribosomal peptide synthesis: approaches to new antibiotics. Chemical reviews 105:715–738.

408 409

19.

Robbel L, Marahiel MA. 2010. Daptomycin, a bacterial lipopeptide synthesized by a nonribosomal machinery. The Journal of biological chemistry 285:27501–27508.

410 411 412

20.

Magarvey NA, Haltli B, He M, Greenstein M, Hucul JA. 2006. Biosynthetic pathway for mannopeptimycins, lipoglycopeptide antibiotics active against drug-resistant grampositive pathogens. Antimicrobial agents and chemotherapy 50:2167–2177.

23

413 414 415 416

21.

Mascio CTM, Mortin LI, Howland KT, Van Praagh ADG, Zhang S, Arya A, Chuong CL, Kang C, Li T, Silverman JA. 2012. In vitro and in vivo characterization of CB183,315, a novel lipopeptide antibiotic for treatment of Clostridium difficile. Antimicrobial agents and chemotherapy 56:5023–5030.

417 418 419 420

22.

Miao V, Coëffet-Legal M-F, Brian P, Brost R, Penn J, Whiting A, Martin S, Ford R, Parr I, Bouchard M, Silva CJ, Wrigley SK, Baltz RH. 2005. Daptomycin biosynthesis in Streptomyces roseosporus: cloning and analysis of the gene cluster and revision of peptide stereochemistry. Microbiology 151:1507–1523.

421 422 423 424

23.

Owen JG, Reddy BVB, Ternei MA, Charlop-Powers Z, Calle PY, Kim JH, Brady SF. 2013. Mapping gene clusters within arrayed metagenomic libraries to expand the structural diversity of biomedically relevant natural products. Proceedings of the National Academy of Sciences of the United States of America 110:11797–11802.

425 426 427 428

24.

Rosconi F, Davyt D, Martínez V, Martínez M, Abin-Carriquiry JA, Zane H, Butler A, de Souza EM, Fabiano E. 2013. Identification and structural characterization of serobactins, a suite of lipopeptide siderophores produced by the grass endophyte Herbaspirillum seropedicae. Environmental microbiology 15:916–927.

429 430 431

25.

Felnagle EA, Jackson EE, Chan YA, Podevels AM, Berti AD, Mcmahon MD, Thomas MG. 2008. Nonribosomal Peptide Synthetases Involved in the Production of Medically Relevant Natural Products. molecular pharmaceutics 5:191–211.

432 433 434 435

26.

Tang MC, Fu CY, Tang GL. 2012. Characterization of SfmD as a Heme peroxidase that catalyzes the regioselective hydroxylation of 3-methyltyrosine to 3-hydroxy-5methyltyrosine in saframycin A biosynthesis. The Journal of biological chemistry 287:5112–5121.

436 437 438 439

27.

Dimise EJ, Widboom PF, Bruner SD. 2008. Structure elucidation and biosynthesis of fuscachelins, peptide siderophores from the moderate thermophile Thermobifida fusca. Proceedings of the National Academy of Sciences of the United States of America 105:15311–15316.

440 441 442

28.

Peng C, Pu J, Song L, Jian X, Tang M, Tang G. 2012. Hijacking a hydroxyethyl unit from a central metabolic ketose into a nonribosomal peptide assembly line. Proceedings of the National Academy of Sciences of the United States of America 109:8540–8545.

24

443 444 445

29.

Zhang W, Ostash B, Walsh CT. 2010. Identification of the biosynthetic gene cluster for the pacidamycin group of peptidyl nucleoside antibiotics. Proceedings of the National Academy of Sciences of the United States of America 107:1–6.

446 447 448

30.

Ross AC, Xu Y, Lu L, Kersten RD, Shao Z, Al-Suwailem AM, Dorrestein PC, Qian PY, Moore BS. 2013. Biosynthetic multitasking facilitates thalassospiramide structural diversity in marine bacteria. Journal of the American Chemical Society 135:1155–1162.

449 450

31.

Lautru S, Deeth RJ, Bailey LM, Challis GL. 2005. Discovery of a new peptide natural product by Streptomyces coelicolor genome mining. Nature chemical biology 1:265–269.

451 452

32.

Zhang Q, Pang B, Ding W, Liu W. 2013. Aromatic Polyketides Produced by Bacterial Iterative Type I Polyketide Synthases. ACS Catalysis 3:1439–1447.

453 454 455

33.

Moss SJ, Martin CJ, Wilkinson B. 2004. Loss of co-linearity by modular polyketide synthases: a mechanism for the evolution of chemical deversity. Natural product reports 21:575–593.

456 457 458

34.

Busch B, Ueberschaar N, Sugimoto Y, Hertweck C. 2012. Interchenar retrotransfer of aureothin intermediates in an iterative polyketide synthase module. Journal of the American Chemical Society 134:12382–12385.

459 460 461

35.

Sugimoto Y, Ding L, Ishida K, Hertweck C. 2014. Rational design of modular polyketide synthases: morphing the aureothin pathway into a luteoreticulin assembly line. angewandte chemie-international edition 53:1560–1564.

462 463 464

36.

Gruenewald S, Mootz HD, Stehmeier P, Stachelhaus T. 2004. In vivo Production of Artificial Nonribosomal Peptide Products in the Heterologous Host Escherichia coli. Applied and environmental microbiology 70:3282–3291.

465 466 467

37.

Tsuge K, Inoue S, Ano T, Itaya M, Shoda M. 2005. Horizontal Transfer of Iturin A Operon , itu , to Bacillus subtilis 168 and Conversion into an Iturin A Producer. Antimicrobial agents and chemotherapy 49:4641–4648.

468 469 470

38.

Park SY, Choi SK, Kim J, Oh TK, Park SH. 2012. Efficient production of polymyxin in the surrogate host Bacillus subtilis by introducing a foreign ectB gene and disrupting the abrB gene. Applied and environmental microbiology 78:4194–4199.

471 472

39.

Luo C, Zhou H, Zou J, Wang X, Zhang R, Xiang Y. 2015. Bacillomycin L and surfactin contribute synergistically to the phenotypic features of Bacillus subtilis 916 and

25

473 474

the biocontrol of rice sheath blight induced by Rhizoctonia solani. Applied Microbiology and Biotechnology 99:1897–1910.

475 476 477

40.

Luo C, Liu X, Zhou H, Wang X, Chen Z. 2015. Nonribosomal peptide synthase gene cluster for lipopeptide biosynthesis in Bacillus subtilis 916 and their phenotypic functions. Applied and Environmental Microbiology 81:422–431.

478 479

41.

Wang X, Luo C, Chen Z. 2012. Genome sequence of the plant growth-promoting rhizobacterium Bacillus sp. strain 916. Journal of bacteriology 194:5467–5468.

480 481 482

42.

Borisova SA, Circello BT, Zhang JK, van der Donk WA, Metcalf WW. 2010. Biosynthesis of rhizocticins, antifungal phosphonate oligopeptides produced by Bacillus subtilis ATCC6633. Chemistry & biology 17:28–37.

483 484

43.

Lautru S, Challis GL. 2004. Substrate recognition by nonribosomal peptide synthetase multi-enzymes. Microbiology 150:1629–1636.

485 486 487

44.

Challis GL, Ravel J, Townsend CA. 2000. Predictive, structure-based model of amino acid recognition by nonribosomal peptide synthetase adenylation domains. Chemistry & biology 7:211–24.

488 489

45.

Kunst F, Rapoport G. 1995. Salt stress is an environmental signal affecting degradative enzyme synthesis in Bacillus subtilis. Journal of bacteriology 177:2403–2407.

490

46.

Wuthrich K. 1986. NMR of Proteins and Nucleic Acids.

491 492

47.

Du L, Lou L. 2010. PKS and NRPS release mechanisms. Natural product reports 27:255– 78.

493 494 495 496 497

48.

Markley J, Bax A, Arata Y, Hibers C, Kaptein R, Sykes B, Wright PE, Wuthrich K. 1998. Recommendations for the presentation of NMR structures of proteins and nucleic acids--IUPAC-IUBMB-IUPAB Inter-Union Task Group on the standardization of data bases of protein and nucleic acid structures determined by NMR spectroscopy. Eur J Biochem 256:1–15.

498 499

26

500

Figure legends:

501

Figure 1. Organization of the locillomycins gene cluster and surrounding genes on the B.

502

subtilis 916 chromosome. The same locus of B. subtilis 168 and B. amyloquefaciens FZB42 are

503

also shown for comparison. Genes with high degree of homology between the three strains

504

(>95% identity) are shown in yellow. The biosynthetic clusters for locillomycins and sporulation

505

killing factor are shown in blue and purple, respectively. Other unrelated genes are also shown in

506

green, red and orange, respectively.

507

508

Figure 2. HPLC-MS chromatograms for locillomycins produced by wild-type B. subtilis 916

509

and its mutant ΔlocD. (A) locillomycins A-C were detected in B. subtilis 916 broth cultures, but

510

were below detectable level in ΔlocD. (B) B(a-c) and B(d-f) are the MS spectra of the extracts

511

from mutant ΔlocD and wild-type B. subtilis 916 respectively. Molecular weights of

512

locillomycin-A, -B and -C were detected in B. subtilis 916 broth cultures, but not observed in

513

mutant ΔlocD culture broth.

514

515

Figure 3. Important NOE and HMBC interactions for determination of the attaching site of the

516

acyl group and the cyclic structure of the peptide moiety were illustrated in the structure (upper

517

right) and in the spectra (upper left and lower panel). The ROESY (upper left) and HMBC

27

518

(lower) spectra are also labeled with assigned signals along the sides. The assigned nucleic

519

names of the key residues are underlined and displayed in red, and the red circles showed

520

relevant NOE and HMBC interactions.

521

522

Figure 4. Structural comparison of locillomycins with surfactins, iturins and fengycins. Some of

523

the key bonds are colored red and blue for clarity. The residue numbers are labeled next to the α-

524

carbon. Statistical sequences and configuration for each residue are listed below the formulas.

525

526

Figure 5. Expression, purification and relative substrate specificities of internal adenylation

527

domains (A domains). (A) Expression and purification of the A domains were analyzed on 10%

528

SDS-polyacrylamide slab gels. (B) A domains were investigated in terms of activity in the ATP-

529

PPi exchange reaction with the amino acids of locillomycin and a control without amino acid.

530

The highest activities were set at 100%. The background was below 5%. The specificities of the

531

different domains coincide with the primary structures of the locillomycins.

532

533

Figure 6. Proposed biosynthetic pathway for locillomycins. Nonribosomal peptide synthetase

534

genes (locA, locB and locC) are represented by arrows, and domains encoded by the respective

535

genes are shown underneath; the domains encoded by locB are proposed to act iteratively. The 28

536

FA (fatty acid chain) couples to the peptide by formation of an amide bond with the amino group

537

of the threonine residue, and a nonapeptide is cyclized by formation of a macrolactone between

538

the threonine hydroxyl and the valine carboxylate. The KAS domain is skipped during the

539

synthesis and is labeled with dashed line box while domains optionally used are labeled by a

540

purple box. LocB appeared twice because it is iteratively used and the A2 domain exhibits

541

different specificity in these two rounds. The E domain following the A2 domain also loses its

542

function in the second round.

543

544

29

Table 1: Primers used in this study Primer

Size

Sequence (5’-3’ )

Note

LocDF

30

TTTGCATGCATGAACTATGATTTATCACAT

Used for ΔlocD mutant

LocDR

29

TTTGAGCTCAGTAAAAATGAGAGGCAATT

LocCF

30

TTTAAGCTTAATTCAGCTCTTTATGAAGAA

LocCR

30

TTTGAGCTCATGAAGTTTACTAATGAATAC

Orf1UF

30

TTTAAGCTTGAATAAATAATTCACGGTAAA

Orf1UR

30

TTTTCTAGAATTCAGCTGCTTTATCGTAAG

Orf1DF

30

TTTTCTAGACTGTTGCTTTGTCGCATAATG

Orf1DR

30

TTTGCATGCAAGAGTGAGTTATCCAGTTGA

Orf3F

30

TTTAAGCTTCATTGAACTGAATAAAATGTA

Orf3R

30

TTTGAGCTCTTAGTCTCAATCTCAATGTTT

Orf7F

30

TTTGCATGCATACGATTAATAAAAGATATG

Orf7R

30

TTTGAATTCTTAAATTGTTATATCATCTTT

LocAA1F

32

TTTCCATGGTATCTGAAATAGAAATGATTACG

LocAA1R

30

TTTCTCGAGTTCTTTCTGAATAGCTGTTTG

LocBA2F

33

TTTCCATGGTAAAAGATGTAGAAATTATTACAG

LocBA2R

33

TTTCTCGAGCATTTGTTCTTTTTTATTATTTGG

LocBA3F

29

TTTCCATGGTTTCGGAGATTGATATCACG

LocBA3R

29

TTTCTCGAGGGTTTCCTGAACATCATTCT

LocBA4F

31

TTTGGATCCATCTCTTCTATAGATATCATGA

construction

Used for ΔlocC mutant construction

Used for Δorf1 mutant construction

Used for Δorf3 mutant construction

Used for Δorf1 mutant construction

Used for A1 domain expression in E.coli

Used for A2 domain expression in E.coli

Used for A3 domain expression in E.coli

Used for A4 domain

LocBA4R

32

TTTCTCGAGTTATGTTTCCTGCAAAACTGTTT

expression in E.coli

LocCA5F

28

TTTCCATGGGAGATGTAGGTTTGCTGAC

Used for A5 domain

LocCA5R

33

TTTCTCGAGAACTAATTCTTTTTCAATGTTATT

LocCA6F

32

AAACCATGGTATATCAAATTAACATGATGACT

LocCA6R

30

AAACTCGAGTTTTGTCTCTGTTTCGTTTCT

PEDF

24

CGCAAAGACTGAACCCACTAATTT

expression in E.coli

Used for A6 domain expression in E.coli

Used

for

quantitative

analysis of PEDV PEDR

24

TTGCCTCTGTTGTTACTTGGAGAT