De Novo Biosynthesis of β-Valienamine in Engineered Streptomyces

Oct 5, 2015 - The C7N aminocyclitol β-valienamine is a lead compound for the development of new biologically active β-glycosidase inhibitors as chem...
0 downloads 0 Views 1002KB Size
Subscriber access provided by CMU Libraries - http://library.cmich.edu

Letter

De novo biosynthesis of #-valienamine in engineered Streptomyces hygroscopicus 5008 Li Cui, Ying Zhu, Xiaoqing Guan, Zixin Deng, Linquan Bai, and Yan Feng ACS Synth. Biol., Just Accepted Manuscript • DOI: 10.1021/acssynbio.5b00138 • Publication Date (Web): 05 Oct 2015 Downloaded from http://pubs.acs.org on October 6, 2015

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

ACS Synthetic Biology is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

1

De novo biosynthesis of β-valienamine in engineered

2

Streptomyces hygroscopicus 5008

3 4

Li Cui, Ying Zhu, Xiaoqing Guan, Zixin Deng, Linquan Bai,* and Yan Feng*

5

State Key Laboratory of Microbial Metabolism, School of Life Science &

6

Biotechnology, and Joint International Research Laboratory of Metabolic &

7

Developmental Sciences, Shanghai Jiao Tong University, Shanghai 200240,

8

China

9 10

ABSTRACT: The C7N aminocyclitol β-valienamine is a lead compound for the

11

development of new biologically active β-glycosidase inhibitors as chemical

12

chaperone therapeutic agents for lysosomal storage diseases. Its chemical

13

synthesis is challenging due to the presence of multi-chiral centers in the

14

structure. Herein, we took advantage of a heterogeneous aminotransferase

15

with stereospecificity and designed a novel pathway for producing

16

β-valienamine in Streptomyces hygroscopicus 5008, a validamycin producer.

17

The aminotransferase BtrR from Bacillus circulans was able to convert

18

valienone to β-valienamine with an optical purity of up to > 99.9% enantiomeric

19

excess value in vitro. When the aminotransferase gene was introduced into a

20

mutant of S. hygroscopicus 5008 accumulating valienone, 20 mg/L of

21

β-valienamine was produced after 96 hours cultivation in shaking flasks. This

22

work provides a powerful alternative for preparing the chiral intermediates for

23

pharmaceutical development.

24

KEYWORDS: β-valienamine, valienone, aminotransferase, validamycin,

25

biosynthesis

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

26

ACS Paragon Plus Environment

Page 2 of 23

Page 3 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

27

C7N aminocyclitols remain increasing interest in the biochemistry of

28

glycosidase inhibitors for their significant bioactivities.1, 2 For instance,

29

β-valienamine, (1S, 2S, 3R, 6R)-6-amino-4-(hydroxymethyl) cyclohex-4-ene-1,

30

2, 3-triol is an optical building block for synthesizing the inhibitors of

31

β-glycosidases.2-4 Its two derivatives, N-octyl-β-valienamine and

32

N-octyl-4-epi-β-valienamine, have been proven to be promising chemical

33

chaperone therapeutic agents for lysosomal storage diseases including

34

Gaucher, GM1-gangliosidosis, and Morquio B disease caused by the disorder

35

of β-glycosidase.5-8 The chemical synthetic routes of this compound have been

36

investigated since 1980s.9-15 However the presence of multiple chiral centers

37

in β-valienamine is quite challenging due to an insufficient stereospecificity of

38

the chemical catalysts. Moreover, the multi-steps, harsh reactions (lower than

39

-70°C), and chemical pollutions also cause synthetic and economic hurdles.

40

Biosynthesis has emerged as a powerful alternative,16,17 especially for chiral

41

amines in asymmetric synthesis.18 Recently, instead of using traditional

42

rhodium, an aminotransferase was found to be remarkably able to convert

43

prositagliptin ketone into sitagliptin with enantiomeric excess (e.e.) value >

44

99.9%.19 However, there is no similar report using aminotransferase for the

45

biosynthesis of β-valienamine. It is also valuable to construct a biosynthetic

46

pathway for producing β-valienamine in vivo.

47

In our previous work, 2-epi-5-epi-valiolone, 5-epi-valiolone and valienone

48

were found to be the intermediates in the biosynthetic pathway of validamycin

49

A in Streptomyces hygroscopicus 5008 (Figure 1, 1-3).20,21 It is noteworthy that

50

valienone has a similar structure with β-valienamine except that the former has

51

a carbonyl group, while the latter has an amino group on C6. Such similarity

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

52

suggests that valienone would be an ideal precursor for certain

53

aminotransferase to produce β-valienamine directly by a transamination

54

reaction (Figure 1). Therefore, we focused on screening aminotransferases

55

that can catalyze the conversion of valienone into β-valienamine in a desired

56

stereospecific manner, and we designed an artificial biosynthetic pathway in S.

57

hygroscopicus 5008 for microbial synthesis of β-valienamine.

58

The key challenge in designing this biosynthetic pathway is the selection of

59

an aminotransferase for the bioconversion of valienone into β-valienamine. To

60

identify suitable enzyme candidates, we used an integrative platform (RxnIP)22

61

to collect enzyme information, especially for enzymes recognizing cyclitol

62

substrates, which is similar to valienone in structure. The sugar

63

aminotransferases (SATs) convert keto-sugars into amino-sugars in the

64

biosynthesis of macrolide antibiotics and belong to the subgroup

65

DegT_DnrJ_EryC1 in the Pfam database.23 We chose SATs that typically use

66

scyllo-inosose and NDP-4-keto-sugar as substrates for further analysis. As

67

shown in Figure 2, four main evolutionary branches existed in a maximum

68

likelihood phylogenetic tree, and one aminotransferase was therefore selected

69

from each branch. These aminotransferases were BtrR24 from Bacillus

70

circulans catalyzing the transamination of 2-deoxy-scyllo-inosose and

71

2-deoxy-3-amino-scyllo-inosose in butirosin A biosynthesis, DesI25 from

72

Streptomyces venezuelae in the biosynthesis of

73

TDP-4-amino-4,6-dideoxyglucose, Per26 from Caulobacter crescentus for

74

GDP-4-amino-6-deoxy-mannose biosynthesis, and ArnB27 from Escherichia

75

coli for UDP-4-amino-arabinose biosynthesis. The four candidate genes were

76

amplified by PCR or chemically synthesized, cloned into plasmid pET28a, and

ACS Paragon Plus Environment

Page 4 of 23

Page 5 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

77

overexpressed in E. coli BL21 (DE3) to yield soluble proteins. The recombinant

78

proteins with the N-terminal His-tag were purified by Ni-NAT columns and

79

confirmed to be a homogeneous band by SDS-PAGE (Figure S1).

80

To assess the feasibility of these aminotransferases to convert the

81

non-natural substrate valienone into β-valienamine, we checked both the

82

catalytic activity and enantio-preference of these enzymes in vitro. The

83

aminotransferase activity assay was performed at 37°C in sodium phosphate

84

buffer (pH 7.5) with valienone and cofactor pyridoxal 5′-phosphate (PLP). To

85

identify the appropriate amino donors, L-glutamine and L-glutamate were

86

tested. As shown in Figure S2a, L-glutamine showed a higher activity for all of

87

the selected enzymes and was therefore used for further enzyme kinetic

88

assays. Quantitative analysis and the stereo-purity of β-valienamine were

89

detected through pre-column derivatization with ortho-phthaldialdehyde (OPA)

90

using high-performance liquid chromatography (HPLC) with fluorescence

91

detection. The derivatives of standards valienamine (S-configuration) and

92

β-valienamine (R-configuration) were separated through HPLC by an Eclipse

93

XDB-C18 column with retention times of 5.1 and 5.7 min, respectively (Figure

94

3a).

95

The mixture of the reaction was analyzed with the same condition. The

96

results indicated that BtrR, Per, and ArnB showed detectable activity after 3

97

hours of incubation (Figure 3c-e). More interestingly, BtrR and Per

98

demonstrated a stringent stereospecificity with >99.9% e.e. value, while ArnB

99

had low stereospecificity with 61% e.e. value for its β-valienamine product.

100

Moreover, BtrR showed the highest activity among these three candidates and

101

converted 5 mM valienone into β-valienamine with 76% conversion after 12

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

102 103

hours at the enzyme concentration of 5 mg/mL for sample preparation. The molecular weight and the absolute configuration of the purified amine

104

product were further verified by high-resolution mass spectrometry (HR-MS)

105

with positive mode, 1H NMR, NOE, NOESY, 13C NMR spectra. The product

106

gave an ion peak at m/z 176.0936 [M+H]+ (Table S1 and Figure S3), and its

107

OPA derivative gave an ion peak at m/z 374.1041 [M+Na]+ (Table S2 and

108

Figure S6a), matching the molecular formula of β-valienamine and its OPA

109

derivative well. 1H NMR analysis of the substrate (valienone) and the product

110

(β-valienamine) showed that the chemical shifts of all of the protons moved to

111

a higher magnetic field, especially the adjacent protons (δ of H-5 from 6.20 to

112

5.50 ppm) and (δ of H-1 from 4.16 ppm to 3.27 ppm ), and a new signal

113

appeared at δ3.35 (s, 1H,H-6), which is consistent with the chemical shift of a

114

methylene that is attached to an amino group. The signal of H-1 was changed

115

from a doublet to a triplet with a coupling constant of 9.8 Hz, indicating that

116

both C-2 and C-6 have the same relative configuration (Figure S4a, b). Further

117

NOE and NOESY analysis showed that there was a correlation between H-2

118

(δ3.50 ppm) and H-6, confirming the configuration of the aminated C-6 as the

119

R-configuration (Figure S4c, d). Meanwhile, the transamination also can be

120

observed through the chemical shifts in 13C NMR spectra (Figure S4e, f).

121

Further comparison of the catalytic efficiency of BtrR and Per towards

122

valienone (Figure 4) was performed by Michaelis-Menten kinetic analysis with

123

L-glutamate dehydrogenase (L-GDH) as a coupling enzyme (Figure S2b).28

124

The Km value of BtrR is only one-sixth that of Per toward valienone, indicating

125

its stronger substrate-binding capacity. Considering the native substrates of all

126

candidates, the BtrR prefers small scyllo-inosose substrates

ACS Paragon Plus Environment

Page 6 of 23

Page 7 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

127

(2-deoxy-scyllo-inosose and 2-deoxy-3-amino-scyllo-inosose), whereas others

128

prefer large NDP-4-keto-sugars. Possibly, this is the reason why BtrR shows a

129

higher binding affinity for valienone. Additionally, the kcat value of BtrR was 3.6

130

folds of that of Per, resulting in an overall 13-fold higher catalytic efficiency (kcat

131

/Km) than Per. Combining e.e. values and kinetic parameters summarized in

132

Figure 4, BtrR was proven to be the best catalytic candidate to transfer an

133

amino group from L-glutamine to valienone for the production of chiral

134

β-valienamine in vitro.

135

The biosynthetic pathway of validamycin A has been thoroughly

136

studied,20,21,29,30 and its producer, S. hygroscopicus 5008, gives a stable yield

137

of validamycin A. To achieve the acquisition of β-valienamine in the microbe

138

via a fermentation process, we designed a novel pathway by introducing the

139

btrR gene into an engineered S. hygroscopicus 5008 to generate a new

140

pathway for the biosynthesis of β-valienamine. We have identified that the C7

141

cyclitol kinase ValC in the validamycin A biosynthetic pathway is responsible

142

for the phosphorylation of valienone.29 Obviously, the presence of ValC is

143

supposed to consume valienone and form a competitive reaction with BtrR.

144

Therefore, the gene valC was knocked out using REDIRECT Technology.29

145

The mutant ∆valC and wild-type 5008 were cultured in the fermentation media

146

for 72 hours, and the production of valienone and validamycin A were analyzed

147

by HPLC. The results showed that the production of validamycin A was

148

abolished (Figure S5a) and valienone was accumulated in the ∆valC mutant

149

(Figure 5b, S5b). Subsequently, the btrR gene was cloned on the integrative

150

vector pPM927 under the control of the strong PvalA promoter30 and

151

integrated into the ∆valC mutant. Thus, a ValA-ValD-ValK-BtrR pathway was

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

152

constructed for the biosynthesis of β-valienamine in the mutant ∆valC::btrR.

153

Mutant ∆valC::btrR and ∆valC were cultured with 50 mL fermentation medium

154

for 5 days in shaking flasks, and the productivity of β-valienamine was

155

monitored by HPLC with OPA-derivatization for every 12 hours. As shown in

156

Figure 5a, the characteristic peak of β-valienamine with a retention time of 5.7

157

min was observed in the fermentation broth of the mutant ∆valC::btrR but

158

absent in mutant ∆valC, which was confirmed by further HR-MS analysis (data

159

not shown). The accumulation of β-valienamine was increased with prolonged

160

incubation time and reached the highest of 20 mg/L at 96 hours (Figure 5b).

161

The fermented β-valienamine product was also verified by HR-MS with

162

pre-column derivatization using OPA. The spectrum matched well with that of

163

the standard β-valienamine and showed the characteristic ion peak at m/z

164

374.1038 [M+Na]+ (Table S2 and Figure S6b). The above data showed that the

165

designed ValA-ValD-ValK-BtrR pathway for β-valienamine biosynthesis is

166

functional, and the engineered strain is able to produce the unnatural product

167

β-valienamine.

168

In summary, we successfully designed and constructed an artificial

169

pathway for generating highly stereospecific β-valienamine both in vitro and in

170

vivo. Compared with the current chemical methods for β-valienamine

171

synthesis, our biological strategy represents a promising alternative by

172

integrating a heterogeneous aminotransferase gene and engineering host

173

metabolic flux due to its sustainable and efficient process. To further increase

174

β-valienamine production, we are investigating the engineering of BtrR for

175

improved catalytic activity on valienone, by in silico design based on the

176

protein structure analysis and systematically optimizing the pathway flux of the

ACS Paragon Plus Environment

Page 8 of 23

Page 9 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

177

host microbe. Our research not only builds a new and efficient route to

178

synthesize β-valienamine, but also facilitates further synthesis of its active

179

derivatives with glycosidase inhibitory activity for treating related diseases.

180

MATERIALS AND METHODS

181

Chemicals. Valienone was synthesized by WuXi AppTec Company (Wu Xi,

182

China). β-Valienamine was prepared and identified after bioconversion with

183

BtrR in vitro in our lab. Valienamine was kindly provided by professor Yuguo

184

Zheng (Zhejiang University of Technology). Other chemicals were purchased

185

from Sigma-Aldrich, Inc. (St. Louis, MO).

186

Molecular phylogenetic analysis by Maximum Likelihood. For

187

phylogenetic analysis, full-length amino acid sequences were aligned by

188

Clustal X (Des Higgins et.al., Conway Institute UCD Dublin) with the following

189

parameters: protein weight matrix = Gonnet; gap open = 10; gap extension =

190

0.2. Maximum likelihood analysis was employed by MEGA6 software with the

191

JTT matrix-based model.

192

Expression and purification of SATs. Candidate SAT genes were amplified

193

and ligated into the pET28a plasmid. Expression plasmids were transformed

194

into E.coli BL21 (DE3). Transformants were grown in LB medium with 100

195

µg/mL kanamycin at 37°C until the cell density at 600 nm reached 0.3-0.5

196

followed by induction with 0.8 mM isopropyl-β-D-thiogalactopyranoside for 10

197

hours to produce N-terminal His6-tagged recombinant proteins. The cells were

198

harvested by centrifugation, resuspended in 20 mM potassium phosphate

199

buffer (pH 7.5), and disrupted by sonication. The fusion proteins were purified

200

with Ni-NTA resin column with 20 mM potassium phosphate buffer (pH 7.5)

201

containing different concentrations of imidazole. The purified proteins were

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

202

detected by SDS- PAGE.

203

Enzymatic activity and the kinetic parameter assays. To measure the

204

activity of sugar aminotransferases (SATs), 1 mg/mL purified SAT was

205

incubated with 5 mM valienone as amino acceptor and 5 mM L-glutamine as

206

an amino donor in 20 mM potassium phosphate (pH 7.5) containing 0.3 mM

207

pyridoxal 5′-phosphate as a cofactor at 37°C for 3 hours followed by

208

pre-column ortho-phthaldialdehyde derivatization and fluorescence detection.

209

The kinetic parameters were determined at 37°C using NADH-dependent

210

L-glutamate dehydrogenase (L-GDH) as a coupling enzyme with a reaction

211

system involving 0.5 U L-GDH, 5 mM ammonium chloride and 0.5 mM NADH

212

in addition to the normal reaction system. The reduction in the absorbance of

213

NADH (ε340=6220 M-1 cm-1) was monitored continuously at 340 nm by a

214

multi-scan spectrum microplate spectrophotometer (Spectra Max, Molecular

215

Devices). One unit of enzyme activity was defined as 1 µmol of NADH

216

consumed per min.

217

Stereopurity determination of β-valienamine by HPLC with pre-column

218

derivatization using OPA. Stereometric purity of β-valienamine was

219

determined by online fluorescence derivatization of ortho-phthaldialdehyde on

220

Agilent 1200 Infinity LC System. β-Valienamine and its (S)-enantiomer

221

standards were derivatized by 0.15 M OPA in the 0.40 M (pH 9.0) borate buffer.

222

The separation of the derivative mixture was performed using an Eclipse

223

XDB-C18 (5 µm, 4.6×150 mm) column eluted with 22% acetonitrile at 30°C for

224

18 min under the detection with the fluorescence detector at 445 nm emission

225

and 340 nm excitation wavelengths with medium sensitivity. β-valienamine and

226

valienamine showed the retention times at 5.7 and 5.1 min, respectively.

ACS Paragon Plus Environment

Page 10 of 23

Page 11 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

227

β-Valienamine purification from the reaction mixture and the

228

fermentation broth of ∆valC::btrR. A mixture of the reaction was filtered

229

through a Microcon YM-10 (Millipore) to remove protein (>10,000 MW) and

230

then purified by Dowex 1×2 (OH- form) (0.5×15 cm) eluted with water. Isolation

231

of β-valienamine from fermentation broth of ∆valC::btrR through two-column

232

chromatography system was performed including Dowex 50×8 (H+ form)

233

eluted with 0.5 M ammonia water and Dowex 1×2 (OH- form) eluted with water.

234

High resolution-mass spectroscopy (HR-MS) and nuclear magnetic

235

resonance spectroscopy (NMR). High-resolution mass spectral analysis was

236

performed using an Agilent 1290-MS 6230 TOF-MS system with positive

237

model. For NMR identification, the purified samples were dried and

238

redissolved in D2O solution, and one-dimensional 1H, 13C and two-dimensional

239

1

240

MHz spectrometer.

241

Construction of ∆valC::btrR mutant. To construct the ∆valC::btrR mutant, a

242

0.9 kb XhoI/NdeI PvalA promoter fragment and a 1.3 kb NdeI/XbaI fragment

243

containing the btrR gene were cloned into pBluescript II SK(+) plasmid

244

digested by XhoI and XbaI. Subsequently, the fragment containing PvalA and

245

btrR was cleaved with EcoRI from the recombinant plasmid and cloned into the

246

integrative vector pPM927. The resulting plasmid was introduced into the

247

∆valC mutant by intergeneric conjugation mediated by E. coli ET12567

248

(pUZ8002) and confirmed by PCR amplification.

249

Fermentation of S. hygroscopicus 5008 and its mutants. S. hygroscopicus

250

5008 and its mutants were cultivated for 72-120 hours at 37°C at 220 rpm in

251

the fermentation medium (gram per liter: corn powder 100, soybean flour 25,

H-1H COSY, NOESY NMR spectra were obtained using Bruker Avance III 400

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

252

yeast extract 5, NaCl 1, and KH2PO4 1.5). Samples were collected every 12

253

hours. The supernatant of the fermentation broth was extracted with

254

chloroform and centrifuged at 12,000 rpm for 10 min before HPLC analysis.

255

Validamycin A analysis by HPLC. The fermentation broth of S.

256

hygroscopicus 5008 and ∆valC mutant were separated through Eclipse

257

XDB-C18 (5 µm, 4.6×150 mm) column, eluted with CH3CN/H2O (3: 97) as

258

mobile phase at 30°C for 18 min and monitored at 210 nm by diode array

259

detector.

260

Valienone accumulation analysis by HPLC with DNPH pre-column

261

derivatization. The accumulation of valienone in the fermentation broth of the

262

∆valC mutant was evaluated by HPLC with a pre-column derivatization using

263

2,4-dinitrophenylhydrazine. The reaction was performed with 7.57 mM DNPH

264

in 5% H3PO4 solution. Analysis was performed using an Eclipse XDB-C18 (5

265

µm, 4.6×150 mm) column, eluted with 50% acetonitrile at 30°C for 18 min, and

266

detected at 380 nm by diode array detector.

267 268

ASSOCIATED CONTENT

269

Supporting Information

270

Additional figures as described in the text. This material is available free of

271

charge via the Internet at http://pubs.acs.org

272 273

AUTHOR INFORMATION

274

Corresponding Authors

275

* Yan Feng Tel: +86-21-34207189

276

* Linquan Bai Tel: +86-21-34206722 E-mail: [email protected]

E-mail: [email protected]

ACS Paragon Plus Environment

Page 12 of 23

Page 13 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

277

Notes

278

The authors declare no competing financial interest.

279 280

ACKNOWLEDGMENTS

281

This work was supported by National 973 Program (2012CB721003), National

282

Science Foundation of China (31200061), and Research Fund for the Doctoral

283

Program of Higher Education (13Z102090066). We are grateful to Prof. Yuguo

284

Zheng for the generous gift of valienamine standard, Prof. Shuangjun Lin for

285

the help on NMR analysis, and Prof. Meifeng Tao and Dr. Guangyu Yang for

286

the help on plasmid construction.

287 288

REFERENCES

289

(1) Mahmud, T. (2003) The C7N aminocyclitol family of natural products.

290

Natural Product Reports 20, 137-166.

291

(2) Ogawa, S., Kanto, M., and Suzuki, Y. (2007) Development and medical

292

application of unsaturated carbaglycosylamine glycosidase Inhibitors.

293

Mini-Reviews in Medicinal Chemistry 7, 679-691.

294

(3) Suzuki, Y., Ogawa, S., and Sakakibara, Y. (2009) Chaperone therapy for

295

neuronopathic lysosomal diseases: competitive inhibitors as chemical

296

chaperones for enhancement of mutant enzyme activities. Perspectives in

297

Medicinal Chemistry 3, 7-19.

298

(4) Higaki, K., Ninomiya, H., Suzuki, Y., and Nanba, E. (2013) Candidate

299

molecules for chemical chaperone therapy of GM1-gangliosidosis. Future

300

Medicinal Chemistry 5, 1551-1558.

301

(5) Suzuki, Y. (2013) Chaperone therapy update: Fabry disease,

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

302

GM1-gangliosidosis and Gaucher disease. Brain & Development 35, 515-523.

303

(6) Luan, Z., Li, L., Ninomiya, H., Ohno, K., Ogawa, S., Kubo, T., Iida, M., and

304

Suzuki, Y. (2010) The pharmacological chaperone effect of

305

N-octyl-β-valienamine on human mutant acid β-glucosidases. Blood Cells,

306

Molecules & Diseases 44, 48-54.

307

(7) Suzuki, Y., Ichinomiya, S., Kurosawa, M., Matsuda, J., Ogawa, S., Iida, M.,

308

Kubo, T., Tabe, M., Itoh, M., Higaki, K., Nanba, E., and Ohno, K. (2012)

309

Therapeutic chaperone effect of N-octyl 4-epi-beta-valienamine on murine

310

G(M1)-gangliosidosis. Molecular Genetics and Metabolism 106, 92-98.

311

(8) Matsuda, J., Suzuki, O., Oshima, A., Yamamoto, Y., Noguchi, A., Takimoto,

312

K., Itoh, M., Matsuzaki, Y., Yasuda, Y., Ogawa, S., Sakata, Y., Nanba, E.,

313

Higaki, K., Ogawa, Y., Tominaga, L., Ohno, K., Iwasaki, H., Watanabe, H.,

314

Brady, R. O., and Suzuki, Y. (2003) Chemical chaperone therapy for brain

315

pathology in G(M1)-gangliosidosis. Proceedings of the National Academy of

316

Sciences of the United States of America 100, 15912-15917.

317

(9) Ogawa, S., Toyokuni, T. and Suami, T. (1980) Synthesis of

318

penta-N,O-acetyl-DL-valienamine and its related branched-chain unsaturated

319

aminocyclitols and cyclitols. Chemistry Letters, 713-716.

320

(10) Ogawa, S., Chida, N., and Suami, T. (1983) Synthetic studies on the

321

validamycins. 5.1,2 Synthesis of DL-hydroxyvalidamine and DL-valienamine.

322

Journal of Organic Chemistry 48, 1203-1207.

323

(11) Ogawa, S., Ashiura, M., Uchida, C., Watanabe, S., Yamazaki, C.,

324

Yamagishi, K., and Inokuchi, J. (1996) Synthesis of potent

325

β-D-glucocerebrosidase inhibitors: N-alkyl-β-valienamines. Bioorganic &

326

Medicinal Chemistry Letters 6, 929-932.

ACS Paragon Plus Environment

Page 14 of 23

Page 15 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

327

(12) Ogawa, S., Kobayashi, Y., Kabayama, K., Jimbo, M., and Inokuchi, J.

328

(1998) Chemical modification of beta-glucocerebrosidase inhibitor

329

N-octyl-β-valienamine: synthesis and biological evaluation of N-alkanoyl and

330

N-alkyl derivatives. Bioorganic & Medicinal Chemistry 6, 1955-1962.

331

(13) Cumpstey, I., Gehrke, S., Erfan, S., and Cribiu, R. (2008) Studies on the

332

synthesis of valienamine and 1-epi-valienamine starting from D-glucose or

333

L-sorbose. Carbohydrate Research 343, 1675-1692.

334

(14) Kuno, S., Takahashi, A., and Ogawa, S. (2013) Concise syntheses of

335

potent chaperone drug candidates, N-octyl-4-epi-β-valinenamine (NOEV) and

336

its 6-deoxy derivative, from (+)-proto-quercitol. Carbohydrate Research 368,

337

8-15.

338

(15) Li, Q. R., Kim, S. I., Park, S. J., Yang, H. R., Baek, A. R., Kim, I. S. and

339

Jung, Y.H. (2013) Total synthesis of (+)-valienamine and (-)-1-epi-valienamine

340

via a highly diastereoselective allylic amination of cyclic polybenzyl ether using

341

chlorosulfonyl isocyanate. Tetrahedron 69, 10384-10390.

342

(16) Lee, S. Y. (2012) Metabolic engineering and synthetic biology in strain

343

development. ACS Synthetic Biology 1, 491-492.

344

(17) Niu, W., and Guo, J. (2015) Stereospecific microbial conversion of lactic

345

acid into 1,2-propanediol. ACS Synthetic Biology 4, 378-382.

346

(18) Brenna, E. (2013) Synthetic methods for biologically active molecules:

347

Exploring the potential of bioreductions. WILEY-VCH.

348

(19) Savile, C. K., Janey, J. M., Mundorff, E. C., Moore, J. C., Tam, S., Jarvis,

349

W. R., Colbeck, J. C., Krebber, A., Fleitz, F. J., Brands, J., Devine, P. N.,

350

Huisman, G. W., and Hughes, G. J. (2010) Biocatalytic asymmetric synthesis

351

of chiral amines from ketones applied to sitagliptin manufacture. Science 329,

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

352

305-309.

353

(20) Yu, Y., Bai, L., Minagawa, K., Jian, X., Li, L., Li, J., Chen, S., Cao, E.,

354

Mahmud, T., Floss, H. G., Zhou, X., and Deng, Z. (2005) Gene cluster

355

responsible for validamycin biosynthesis in Streptomyces hygroscopicus

356

subsp. jinggangensis 5008. Applied and Environmental Microbiology 71,

357

5066-5076.

358

(21) Bai, L., Li, L., Xu, H., Minagawa, K., Yu, Y., Zhang, Y., Zhou, X., Floss, H.

359

G., Mahmud, T., and Deng, Z. (2006) Functional analysis of the validamycin

360

biosynthetic gene cluster and engineered production of validoxylamine A.

361

Chemistry & Biology 13, 387-397.

362

(22) Hu, Q. N., Deng, Z., Hu, H., Cao, D. S., and Liang, Y. Z. (2011) RxnFinder:

363

biochemical reaction search engines using molecular structures, molecular

364

fragments and reaction similarity. Bioinformatics (Oxford, England) 27,

365

2465-2467.

366

(23) Hwang, B. Y., Cho, B. K., Yun, H., Koteshwar, K., and Kim, B. G. (2005)

367

Revisit of aminotransferase in the genomic era and its application to

368

biocatalysis. Journal of Molecular Catalysis B: Enzymatic 37, 47-55.

369

(24) Tamegai, H., Nango, E., Kuwahara, M., Yamamoto, H., Ota, Y., Kuriki, H.,

370

Eguchi, T., and Kakinuma, K. (2002) Identification of L-glutamine:

371

2-deoxy-scyllo-inosose aminotransferase required for the biosynthesis of

372

butirosin in Bacillus circulans. The Journal of Antibiotics 55, 707-714.

373

(25) Burgie, E. S., and Holden, H. M. (2007) Molecular architecture of DesI: a

374

key enzyme in the biosynthesis of desosamine. Biochemistry 46, 8999-9006.

375

(26) Cook, P. D., Carney, A. E., and Holden, H. M. (2008) Accommodation of

376

GDP-linked sugars in the active site of GDP-perosamine synthase.

ACS Paragon Plus Environment

Page 16 of 23

Page 17 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

377

Biochemistry 47, 10685-10693.

378

(27) Breazeale, S.D., Ribeiro, A.A., and Raetz, C.R. (2003) Origin of lipid A

379

species modified with 4-amino-4-deoxy-L-arabinose in polymyxin-resistant

380

mutants of Escherichia coli. An aminotransferase (ArnB) that generates

381

UDP-4-deoxyl-L-arabinose.Journal of Biological Chemistry 278, 24731-24739.

382

(28) Akabayashi, A., and Kato, T. (1989) One-step and two-step fluorometric

383

assay methods for general aminotransferases using glutamate dehydrogenase.

384

Analytical Biochemistry 182, 129-135.

385

(29) Minagawa, K., Zhang, Y., Ito, T., Bai, L., Deng, Z., and Mahmud, T. (2007)

386

ValC, a new type of C7-Cyclitol kinase involved in the biosynthesis of the

387

antifungal agent validamycin A. ChemBioChem 8, 632-641.

388

(30) Wu, H., Qu, S., Lu, C., Zheng, H., Zhou, X., Bai, L., and Deng, Z. (2012)

389

Genomic and transcriptomic insights into the thermo-regulated biosynthesis of

390

validamycin in Streptomyces hygroscopicus 5008. BMC Genomics 13,

391

337-350.

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

For Table of Contents Use Only De novo biosynthesis of β-valienamine in engineered Streptomyces hygroscopicus 5008 Li Cui, Ying Zhu, Xiaoqing Guan, Zixin Deng, Linquan Bai,* and Yan Feng* 196x160mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 18 of 23

Page 19 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

Figure 1. Biosynthetic pathway of validamycin A (dashed frame) and de novo aminotransferase-catalyzed route for β-valienamine biosynthesis (solid frame) in S. hygroscopicus 5008. 117x52mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 2. Phylogenetic analysis of SATs and the reactions catalyzed by the selected (red) candidates. The multiple alignments were performed by Clustal X, and the phylogenetic tree was generated using MEGA6. BtrR_Bci (CAD41947), SpcS2_Ssp (ABW87804), StsC_Sgl (AIR96275), GtmB_Mec (CAE06512), StrS_Sgr (CAA68523), WecE_Eco (AAC76796), ArnB_Eco (AAM92146), ArnB_Sty (NP_461239), Per_Eco (AAG57096), Per_Ccr (AAK22996), LmbS_Sli (CAA55764), DesI_Sve (AAC68684), PseC_Hpy (AAD07433), PglE_Cje (AAD09304). 574x210mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 20 of 23

Page 21 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

Figure 3. HPLC analysis of the reaction mixture via pre-column derivatization using OPA. (a) Valienamine and β-valienamine standards; (b) Reaction mixture without enzyme as the blank control; (c, d, e) Reactions catalyzed by BtrR, Per, and ArnB, respectively. 200x139mm (300 x 300 DPI)

ACS Paragon Plus Environment

ACS Synthetic Biology

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Figure 4. Enzymatic conversion and kinetic properties of selected SATs. (Column) Enzymatic conversion of valienone to β-valienamine by selected SATs. Data were collected from three measurements with 1 mg/mL enzymes incubated with 5 mM valienone, 5 mM L-glutamine, and 0.3 mM PLP at 37°C in sodium phosphate buffer (pH 7.5); (Table) Kinetic parameters of different SATs; 0.5 U L-GDH was added into the reaction system as a coupling enzyme with 5 mM ammonium chloride and 0.5 mM NADH. 210x77mm (300 x 300 DPI)

ACS Paragon Plus Environment

Page 22 of 23

Page 23 of 23

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

ACS Synthetic Biology

Figure 5. Production of β-valienamine in the ∆valC mutant integrated with the heterogeneous aminotransferase gene btrR. (a) HPLC analysis of 96-hour fermentation broths with pre-column derivatization using OPA; (b) Time course of β-valienamine and valienone titer in ∆valC and ∆valC::btrR mutant. 297x420mm (300 x 300 DPI)

ACS Paragon Plus Environment