AAC Accepted Manuscript Posted Online 26 January 2015 Antimicrob. Agents Chemother. doi:10.1128/AAC.04414-14 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Neutralization of Staphylococcal enterotoxin B by aptamer

2

antagonist

3

Kaiyu Wanga,b, Longjie Ganb, Li Jiangb, Xianhui Zhangb, Xiangyue Yanga,b, Min Chena,b,

4

Xiaopeng Lana,b

5

a

6

Fuzhou 350025, PR China, and Department of Laboratory Medicine, Dongfang Hospital,

7

Xiamen University, Fuzhou 350025, P.R. China

8

b

9

Fuzhou 350025, P.R. China

10 11

*

Institute of Laboratory Medicine, Fuzhou General Hospital of Nanjing Military Command,

Department of Laboratory Medicine, Fuzong Clinical College, Fujian Medical University,

Email: [email protected]; Tel: +86 591 22859490

12

Abstract

13

Staphylococcal enterotoxin B (SEB) is a major virulence factor for

14

Staphylococcal toxic shock syndrome (TSS). SEB activates a large subset of the T

15

lymphocytic

16

SEB-initiated toxicity may be an effective strategy for treating TSS. Using a

17

systematic evolution of ligands by exponential enrichment (SELEX) process, we

18

identified an aptamer that can antagonize SEB with nanomolar binding affinity (Kd =

19

64 nM). The aptamer antagonist effectively inhibits SEB-mediated proliferation and

20

cytokine secretion in human peripheral blood mononuclear cells. Moreover, a

21

PEGlyated aptamer antagonist significantly reduced mortality in a “double-hit” mouse

22

model of SEB-induced TSS, established via sensitization with D-galactosamine

23

followed by SEB challenge. Therefore, our novel aptamer antagonist may offer

24

potential therapeutic efficacy against SEB-mediated TSS.

25

Keywords: superantigens; aptamer; Staphylococcal enterotoxin B; toxic shock

population,

releasing

pro-inflammatory

cytokines.

Blocking

26

1. Introduction

27

Staphylococcal enterotoxins (SEs) are prototype superantigens excreted

28

primarily by Staphylococcus aureus and Streptococcus pyogenes. SEs are potent

29

activators of the immune system that can stimulate up to 40% of the naive T cell

30

population and cause diverse human diseases, including food poisoning, autoimmune

31

diseases, and toxic shock syndrome (TSS) (1). SEs can activate both

32

monocytes/macrophages and T lymphocytes by directly binding to MHC II molecules

33

and to the variable region of the β-chain of T-cell receptors (TCR), without being

34

taken up and presented by antigen presenting cells (2).

35

These toxins lead to the proliferation of monocytes/macrophages and T

36

lymphocytes and stimulate secretion of excess pro-inflammatory cytokines, including

37

interferon gamma (IFNγ), interleukin 2 (IL-2), and tumor necrosis factor alpha

38

(TNFα). Among these SEs, Staphylococcal enterotoxin B (SEB) is considered to be an

39

important cause of Staphylococcal TSS, which is characterized by high fever,

40

erythematous rash, hypotension, multi-organ system failure, and even death (3).

41

Remarkably, SEB is highly heat resistant, easily transformed into an aerosol, and

42

readily produced on a large scale using genetic engineering techniques. Humans are

43

extremely susceptible to this superantigen, so SEB is often referred to as an

44

"incapacitating agent" by the military who may use this toxin as an anti-personnel

45

aerosol or as a food supply contaminant (4).

46

At present, SEB is thought to be an effective treatment strategy for TSS by

47

blocking early processes prior to T cell activation and initiation of the toxic cascade.

48

Studies suggest multiple approaches for interfering with SEB binding to MHC-II

49

and/or TCR. These include vaccine immunization to induce anti-SEB antibodies (5, 6),

50

passive immunoprophylaxis with natural or artificial antibodies (7, 8), and the use of

51

synthetic peptides (9) or receptor mimics (10, 11) as antagonists to prevent the

52

formation of the MHC-II/SEB/TCR complex. Research is ongoing for new

53

compounds with greater half-lives, improved SEB binding affinity, and less

54

immunogenicity.

55

Aptamers are artificial short, single-stranded oligonucleotides (ssDNA or ssRNA)

56

that can form elaborate 3-D structures and specifically bind to desired targets with

57

high affinity (12, 13). Because of their intrinsic properties, aptamers are non-toxic and

58

non-immunogenic. In addition, aptamers composed of modified nucleotides have a

59

long in vivo half-life of hours to days.

60

During the past two decades, numerous aptamers have been developed to inhibit

61

pathogenic antigen activity and many have substantial clinical potential for treating

62

various diseases (14). Several researchers have reported on the selection of specific

63

DNA aptamers against SEB (15-17). Methods using these aptamers as capture

64

molecules have also been established to detect SEB in various samples (15, 18).

65

However, as far as we know, no aptamer has been selected to inhibit the biological

66

activity of SEB.

67

Therefore, we selected DNA aptamers directed against native SEB in vitro and

68

identified aptamer-antagonists capable of neutralizing SEBs by studying their

69

therapeutic effects on SEB-mediated toxic shock (TSS) in vitro using human

70

peripheral blood mononuclear cell (PBMC) cultures as well as a lethal murine model.

71

72

2. Material and Methods

73

2.1 SELEX library and primers

74

The ssDNA library, which contained random 60mer and fixed primer regions and

75

all primers used are described previously (ESI Table S1†) (19). All oligonucleotides

76

were synthesized and purified with HPLC (Invitrogen, Guangzhou, China). For

77

animal experiments, the aptamer candidate was conjugated at the 5’ end with 40-KDa

78

polyethylene glycol (PEG). The PEGlyated aptamer was prepared by treating 40-KDa

79

PEG N-hydroxysuccinimide ester with 5'-amino and 3'-inverted dT modified aptamer

80

candidates. All PEG-aptamer conjugates were purified by anion exchange and then by

81

reverse-phase HPLC (Seebio Biotech, Shanghai, China).

82

2.2 Aptamer screening for specific SEB binding

83

ssDNA aptamers specific to SEB were screened by a SELEX process as

84

described previously with minor modifications (20). Briefly, highly purified SEB

85

(Toxin Technology, Sarasota, USA) was coupled to M-270 carboxylic acid dynabeads

86

(DYNAL, Oslo, Norway) according to the manufacturer's instructions. Prior to

87

selection, the ssDNA library dissolved in 500 μl selection buffer (50 mM Tris. HCl

88

pH7.4, 5 mM KCl, 100 mM NaCl, 1 mM MgCl2) was denatured by heating at 95 oC

89

for 5 min and immediately placed on ice for 10 min. The denatured ssDNA library

90

and a 5-fold molar excess of yeast tRNA (Invitrogen) were incubated with SEB (100

91

ng) immobilized on magnetic beads in a 500 μl reaction, and were incubated at 37 oC

92

for 60 min while shaking.

93

Nonbinding oligonucleotides were removed by washing the DNA-bead complex

94

three times with selection buffer (0.2% BSA). SEB binding species were eluted by

95

heating at 100 oC for 5 min in 200 μl of sterile ddH2O. ssDNA was amplified by PCR

96

with FITC-labeled sense primers and biotin-labeled antisense primers (20-30 cycles of

97

0.5 min at 94 oC, 0.5 min at 52 oC, 0.5 min at 72 oC, followed by 5 min at 72 oC). All

98

PCR products were purified using a BioSpin gel extraction kit (Bioer Technology,

99

Hangzhou, China). From all purified PCR products from each SELEX round, large

100

aliquots were alkaline-denatured and used to prepare the aptamer strand using M-280

101

streptavidin dynabeads (DYNAL AS, Oslo, Norway) according to the manufacturer's

102

instructions, and small aliquots were stored at -80 oC prior to use.

103

To create aptamers with high affinity and specificity, we progressively increased

104

the selective pressure by increasing the number of washes and KCl, and by decreasing

105

the ratio of ssDNA to SEB and the incubation time (ESI Table S2†). The

106

counter-selection (depletion of aptamers that bind to non-target molecules) was

107

introduced against BSA-or Staphylococcal protein A (SPA)-coupled M-270 beads in

108

the 7th, the 9th, and the 11th round, respectively. BSA was the blocking protein for SEB

109

coupling, and SPA is a major protein of Staphylococcus aureus(21). To reduce the

110

matrix binders, reaction tubes were blocked with 1% BSA-PBS, and preselection

111

steps with uncoupled streptavidin beads were performed.

112

2.3 Cloning, sequencing and bioinformatic analysis

113

After rounds of SELEX selection, the selected ssDNA pool was PCR-amplified

114

using unlabeled primers under the same conditions described for screening. PCR

115

products were purified, and then cloned into pGEM-T vector (Promega, Madison,

116

USA) according to the manufacturer’s instructions. The resulting products were

117

transformed into Escherichia coli DH5α. Individual cultured colonies were chosen

118

randomly and their inserts were sequenced by Invitrogen Company. The aptamer

119

sequences were analyzed by ClustalX software and the secondary structure was

120

predicted by free-energy minimization algorithm according to Zuker (22) using the

121

internet-tool mfold (http://mfold.bioinfo.rpi.edu/cgi-bin/dna-form1.cgi).

122

2.4 Aptamer binding assay for SEB

123

A fluorescently labeled ssDNA binding assay was performed to monitor the

124

enrichment of each SELEX round and to evaluate aptamer binding affinity. In brief,

125

fluorescently labeled ssDNA was thermally denatured in 200 μl of selection buffer

126

and then incubated in the dark with SEB immobilized on magnetic beads at 37 oC for

127

1 h while shaking. After incubation, non-bound ssDNA was collected, and SEB-bound

128

ssDNA was eluted by heating at 100 oC for 5 min in 200 μl of selection buffer. The

129

fluorescent intensity of applied, non-bound, and eluted ssDNA was measured

130

respectively using a TBS-380 Mini-Fluorometer (Turner Biosystems, Sunnyvale, CA).

131

All binding assays were repeated three times. To monitor the enrichment of each

132

SELEX round, the percent of ssDNA (200 nM) binding with SEB (20 μM) was

133

calculated.

134

To determine the binding affinity of a selected aptamer, the binding assay was

135

performed as described above but with increasing amounts of ssDNA aptamer (0 to

136

400 nM) and a constant amount of SEB (20 μM) for each assay. To calculate the

137

dissociation constants (Kd) of the aptamers, the aptamer quantity bound to targets was

138

plotted and data points were fitted to the equation Y=BmaxX/(Kd+X) via non-linear

139

regression analysis, using Graphpad-Prism-V5.01 (Graphpad, San Diego, CA).

140

2.5 Human PBMC proliferation assays

141

To study the effect of aptamers on SEB-mediated T cell proliferation, an MTT

142

cell proliferation assay was performed with human PBMCs as described previously

143

(23). Human PBMCs were isolated from heparinized venous blood by Ficoll-Hypaque

144

density gradient centrifugation from healthy human donors. After lysing erythrocytes,

145

PBMCs were cultured in RPMI 1640 medium (Gibco, Grand Island, USA)

146

supplemented with 2 mM L-glutamine, 100 U/ ml penicillin, 100 mg/ml streptomycin,

147

10 mM HEPES and 10% (v/v) fetal bovine serum (Gibco) at 37 oC. PBMCs (105

148

cells/well) were plated with SEB (200 ng/ml) alone or combined with varying

149

concentrations of aptamer candidates in triplicate in a 96-well plate.

150

PBMCs cultivated in RPMI-1640 medium alone served as negative control.

151

Plates were incubated in a 5% CO2 humidified incubator at 37 oC for 48 h. After

152

stimulation, cell proliferation was measured by adding 20 μl of MTT solution (Sigma,

153

St. Louis, USA), prepared in PBS at 5 mg/ml (pH 7.2), to each well. After 2 h of

154

incubation, 100 μl of sodium dodecyl sulfate-dimethylformamide (SDS-DMF)

155

solution was added to each well and the assay was allowed to develop overnight. The

156

plates were read at 570 nm. Data are expressed as values of the stimulation index (SI).

157

The SI value was calculated as the ratio of the absorbent value at 570 nm of pools

158

incubated with SEB compared to pools incubated with medium only.

159

2.6 Cytokine quantitation

160

To measure cytokines, a standard curve was first prepared using known

161

concentrations of recombination cytokines. After stimulation with SEB with/without

162

the addition of the candidate aptamer, human TNFα, IL-1β, IL-6, IL-2, and IFNγ in

163

culture supernatants from PBMCs were measured with ELISA according to the kit

164

instructions (R&D Systems, Minneapolis, USA). Cytokines in each sample were

165

measured from the standard curve and the detection limit of all assays was 20 pg/ml.

166

2.7 Aptamer binding assay for the MHC II molecules

167

The binding ability of aptamer A11 to the MHC II molecules was measured with

168

a competition assay as described previously (16). Human Raji cells (2 × 105 cells)

169

were incubated in 100 μl of selection buffer (1% BSA) with varying final

170

concentration of SEB (0, 25, 50, 75, or 100 μg/ml) and with or without 5-fold molar

171

of A11 for 40 min on ice. Cells were washed in selection buffer and incubated with

172

FITC-conjugated SEB (FITC-SEB, 50 μg/ml), prepared using a FluoReporter® FITC

173

Protein Labeling Kit (Amresco, Ohio, USA). After extensive washing, Raji cells were

174

resuspended and analyzed by flow cytometry. The surface fluorescence of Raji cells

175

was measured as mean fluorescence of three independent experiments.

176

2.8 Murine experiments

177

To investigate the therapeutic efficacy of aptamers in vivo on SEB-induced TSS,

178

a “double-hit” murine model was established (24). Pathogen-free female BALB/c

179

mice of 10–12 weeks-of-age were purchased from the Experimental Animal Centre of

180

Fuzhou General Hospital of Nanjing Military Command (Fuzhou, China). The mice

181

were housed under controlled conditions and fed commercial mouse chow and water.

182

Groups of mice (N=19) were sensitized by intraperitoneal injection of

183

D-galactosamine (GalN, 20 mg/animal, Sigma, St. Louis, USA), and then challenged

184

by intraperitoneal injection of SEB (20 μg/animal) at 4 h. After treatment, mice were

185

fasted but allowed ad libitum access to water.

186

For therapeutic investigations, PEGlyated aptamer (14 nmol) was injected

187

intravenously 1 h before or 2 h after SEB injection. All agents were dissolved in

188

sterile nonpyrogenic saline solution (pH=7.2) and were injected intravenously via the

189

tail vein at 10 μl/g. Mouse survival was calculated by Kaplan-Meier survival analyses.

190

The IACUC of Fuzhou General Hospital of Nanjing Military Command approved all

191

experimental procedures.

192

2.9 Statistical analysis

193

Results are presented as means ± S.D. and analyzed for significant differences by

194

the Student's t-test. Kaplan-Meier methods were used to compute survival analyses. A

195

value of P < 0.05 was considered statistically significant. All data were processed by

196

SPSS 12.0 for Windows.

197

198

3. Results

199

3.1 In vitro selection and sequence analysis of SEB binding aptamers

200

3.1.1 Relative binding rate of ssDNA and SEB

201

Eleven rounds of in vitro selection were used to isolate aptamers that could

202

favorably bind to SEB. Figure 1 depicts the binding ratio of the ssDNA pool to SEB

203

which increased gradually with increasing number of selection rounds, and then

204

remained nearly constant in the last three rounds. This suggested that SEB binding

205

aptamers with good binding affinity were enriched, and the binding ability of

206

selection pools plateaued after about ten rounds of selection.

207

3.1.2 Cloning, sequencing, and sequence analysis of oligonucleotide aptamers

208

The enriched ssDNA pool of the 11th round was cloned into E. coli DH5α using

209

the pGEM-T vector system. Forty individual clones were chosen randomly and

210

sequenced. Multiple sequence alignments revealed that the primary clone structures

211

were modestly enriched. In total, 6 nonredundant sequences were identified, and 13

212

sequences were identical to aptamer A1 and 8 variants of aptamer A1, which were

213

highly homologous with aptamer A1, with only one or two base differences. Of all

214

sequences, 5 A2, 3 A3, 2 A4, 1 A5, and 1 A6 were identified. Although each aptamer

215

had the characteristic stem-loop or hairpin, their secondary structures predicted by

216

software were significantly different. Although all aptamers can bind with SEB (ESI

217

Figure S1 †), and some had dissociation constants in the low nanomolar range, none

218

could inhibit SEB function except aptamer A2, which had a weak inhibitory effect

219

(Figure 2A).

220

3.2 Screening functional aptamers by PBMC proliferation assay

221

To identify functional aptamers that can inhibit SEB superantigen activity, 40

222

individual clones from the 10th round were sequenced further as mentioned above. In

223

total, 29 nonredundant sequences were identified, and A1-A6 was from the 11th round

224

and A7-A29 was from the 10th round. Because superantigens can polyclonally activate

225

T cells, these 29 oligonucleotides were all chemically synthesized, and were tested for

226

inhibition of SEB-mediated proliferation of human PBMCs. Data show that

227

SEB-mediated proliferation of PBMCs decreased most significantly by A11 in a

228

dose-response manner, with maximal inhibition (93%) achieved at 20 times the

229

molarity of SEB compared to controls incubated with toxin alone (P < 0.05). At the

230

same concentration, A2 achieved 34% inhibition (P < 0.05). However, the other 27

231

did not significant inhibit binding (Fig. 2). A2 and A11 both bound with high affinity

232

to SEB, with dissociation constants of 26 nM and 64 nM respectively (Fig. 3A).

233

Although no significant between-aptamer sequence similarity was found, secondary

234

structures of A11 and A2 were predicted to be similar, and both had the characteristic

235

stem-loop (Fig. 3B). Thus, the candidate aptamer A11 could potentially inhibit

236

superantigen activity of SEB in vitro.

237

3.3 Inhibition of SEB-mediated cytokine release from human PBMCs

238

To confirm the inhibitory role of A11, the effect of A11 on the production of

239

pro-inflammatory cytokines was examined with ELISA in human PBMCs incubated

240

with SEB. Fig. 4 shows that A11 dose dependently reduced cytokine production from

241

PBMCs incubated with SEB, compared to controls incubated with toxin alone

242

(P

Neutralization of staphylococcal enterotoxin B by an aptamer antagonist.

Staphylococcal enterotoxin B (SEB) is a major virulence factor for staphylococcal toxic shock syndrome (TSS). SEB activates a large subset of the T ly...
1MB Sizes 1 Downloads 8 Views