IAI Accepts, published online ahead of print on 13 October 2014 Infect. Immun. doi:10.1128/IAI.02334-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Mucosal immunization with a live attenuated vaccine SPY1 induces humoral and

2

Th2-Th17-Treg cellular immunity and protects against pneumococcal infection

3

Xiuyu Xu1, 2, Hong Wang1, Yusi Liu1, Yiping Wang1, Lingbing Zeng1, Kaifeng Wu1,

4

Jianmin Wang1, Feng Ma1, Wenchun Xu1, Yibing Yin1, and Xuemei Zhang1*

5

1 Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine

6

(Ministry of Education), Chongqing Medical University, Chongqing, People’s

7

Republic of China

8

2 Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing

9

Medical University, Chongqing, People’s Republic of China

10

Abstract

11

Mucosal immunization with attenuated vaccine can protect against pneumococcal

12

invasion infection, but the function was unknown. Our study found that mucosal

13

delivery with the live attenuated vaccine SPY1 strain can confer T cells and B cells

14

dependent protection against pneumococcal colonization and invasive infection, yet

15

it’s still unclear which cell subsets contribute to the protection and their roles in

16

pneumococcal colonization and invasion remain elusive. Adoptive transfer of

17

anti-SPY1 antibody conferred protection to naïve μMT mice and immune T cells were

18

indispensable to protection examined in nude mice. A critical role of IL-17A in

19

colonization was demonstrated in mice lacking IL-17A and vaccine-specific Th2 * Corresponding authors at: Mailing address: Department of Laboratory Medicine, Key Laboratory of Diagnostic Medicine (Ministry of Education), Chongqing Medical University, 1 Yixueyuan Road, Yuzhong District, Chongqing 400016, People’s Republic of China. Tel.: +86 23 68485216; fax: +86 23 68485005. E-mail addresses: [email protected] (X. Zhang). 1

20

immune subset was necessary for systemic protection. Of note, we found that SPY1

21

could stimulate immunoregulatory response and SPY1-elicited regulatory T cells

22

participated in protection against colonization and lethal infection. The data presented

23

here aid our understanding of how live attenuated strains are able to function as

24

effective vaccines, and may be contribute to a more comprehensive evaluation of live

25

vaccines and other mucosal vaccines.

26

Key words:

27

Streptococcus pneumoniae; live attenuated vaccine; Th2; Th17; regulatory T cell

28

Running title: SPY1 induces humoral and Th2-Th17-Treg immunity

29

Introduction

30

Vaccination is an indispensable strategy to prevent infection caused by Streptococcus

31

pneumonia (S. pneumoniae), which is estimated to cause mortality rate of more than

32

50 deaths in every 1000 births in children under 5 years of age in some countries (1).

33

The commercially available 23-valent polysaccharide vaccine contains the most

34

common types causing pneumococcal infection and is effective in adults but fails to

35

protect children of less than 2 years of age, who are most vulnerable to pneumococcal

36

infection. The recent extensive introduction of conjugated capsular polysaccharide

37

vaccine (PCV) has drastically decreased the child morbidity and mortality caused by

38

strains of S. pneumoniae expressing capsular serotypes included in the vaccine,

39

however, the serotypes coverage of PCV is limited and growing evidences showed

40

that PCV could induce selective pressure and gradual replacement with nonvaccine 2

41

types (serotype replacement) (2, 3). The conjugated vaccine is also very expensive

42

and is complex in design, making its application in the low income countries having

43

the highest burden of S. pneumoniae infections more difficult (4).

44

As a consequence of these shortcomings with the commercially available S.

45

pneumoniae vaccines, other approaches have been explored, including protein

46

antigens vaccines, killed whole cell S. pneumoniae, or attenuated live S. pneumoniae

47

vaccines (5, 6). The wide range of antigenic molecules present in live attenuated

48

vaccines promising the immune responses they induce are likely to be multiple and

49

powerful and may also more closely mimic those obtained in natural infection

50

compared to immune responses to a sub-component or killed bacterial vaccine (7).

51

Some of live bacterial vaccines have been clinically used, including the BCG (8) and

52

vaccines for preventing typhoid (9). Hence live vaccines could be potentially useful

53

against S. pneumoniae, and there are some reports about live attenuated vaccine

54

candidates. Roche et al. proved a live unencapsulated attenuated S. pneumoniae

55

strains elicited effective systemic and mucosal protection (10), and Richards et al.

56

demonstrated that prior nasopharyngeal colonization with a pneumolysin deficient

57

pneumococcal mutant resulted in serotype independent protection against invasive

58

pneumoccoal infection (11). More recently, serotype cross protection induced by a

59

bacterial lysis deficient pneumococcal mutant was reported in systemic and mucosal

60

infection mice model (12).

61

Streptococcus pneumoniae strain SPY1 is a capsule-negative mutant obtained

62

accidently when we were studying the molecular function of SPD 1672 gene which 3

63

was deleted by allelic replacement with erm cassette from D39 strain using the

64

method we previously described (13). The SPY1 vaccine strain formed small,

65

rough-like colonies, in sharp contrast with the big and smooth colonies of wild type

66

D39 strain. In addition to variation in SPD 1672 gene, several mutations outside the

67

SPD 1672 gene including mutation in the promoter of the cps locus were identified in

68

SPY1 by whole genome sequencing, which contributed to the significant attenuation

69

of SPY1 (14). In the previous study, we have found that intranasal immunization with

70

SPY1 can reduce colonization and protect against otherwise-lethal challenge of

71

homologous pneumococcal strains in a serotype-independent manner (14, 15).

72

However, the immunity mechanisms mediating the protection elicited by SPY1 were

73

not clear. Many researches have shown that the protection induced by living vaccine

74

not only depends on the effect of B cells, but also on the effect of CD4 + T cells (16,

75

17). And the vaccine-specific antibody has been recognized to participate in the

76

protection. In the previous study, we have found that SPY1 can induce the foundation

77

of IFN-γ, IL-17A, IL-10 and IL-4 in BALB/c mice. But, the subtypes of CD4 + T

78

cells (Th1, Th2, of Th17 and Treg cells) have been shown playing different role in the

79

process of resistance to S. pneumoniae infection. IL-17A has been reported to play

80

different roles in local and lethal pneumococcal infections (16, 17). Also, there was no

81

article reporting the function of Treg cells in pneumococcal vaccine induced

82

protection.

83

In this study, mechanisms mediating the protection elicited by SPY1 were

84

evaluated in immune deficient mice model. Vaccine-specific protection is mediated by 4

85

humoral and T cell immunity together. Th17-mediated phagocytes infiltration and Th2

86

immune subset are responsible for protection against colonization and lethal

87

pneumococcal challenge, respectively. We showed, for the first time, that

88

SPY1-specific T regulatory cells were protective in the mucosal and systemic

89

protection against pneumococcal infection. Together, these findings demonstrate the

90

immune mechanisms involved in protection elicited by mucosal vaccination of SPY1,

91

may contribute to a more comprehensive evaluation of living vaccines and other

92

mucosal vaccines.

93 94

Materials and methods

95

Mice. C57BL/6 mice, BALB/c mice and nude BALB/c mice (4-6 weeks, female)

96

were purchased from Chongqing Medical University, Chongqing, China. B cell

97

deficient BALB/c mice (μMT mice) were obtained from Chinese Academy of

98

Sciences, China. IFN-γ deficient C57BL/6 mice (B6.129S7-Ifngtm1Ts/J) and IL-4

99

deficient C57BL/6 mice (B6.129P2-Il4tm1Cgn/J) were purchased from The Jackson

100

Laboratories. IL-17A deficient C57BL/6 mice were obtained from Nankai University,

101

China. Mice were kept under specific-pathogen-free conditions at the animal facilities

102

of Chongqing Medical University during the time of the experiments. All the animal

103

experiments were done in accordance with the Institutional Animal Care and Use

104

Committee’s guidelines of Chongqing Medical University.

105

Bacterial strains and immunogens. S. pneumoniae strain NCTC 7466 (D39,

106

serotype 2) was obtained from the National Collection of Type Cultures (NCTC, 5

107

London, UK). S. pneumoniae strain TIGR4 (serotype 4) was obtained from the

108

American Type Culture Collection (ATCC, Manassas, USA). S. pneumoniae clinical

109

isolates CMCC 31693 (serotype 19F), CMCC 31614 (serotype 14), CMCC 31207

110

(serotype 6B), CMCC 31436 (serotype 3) and CMCC 31203 (serotype 3) were

111

obtained from the National Center for Medical Culture Collections (CMCC, Beijing,

112

China). SPY1 is a novel live attenuated S. pneumoniae which could be used as

113

vaccine (14, 15). All S.pneumoniae strains were grown on Columbia sheep blood agar

114

plates or in C plus Y medium at 37°C in 5% CO2.

115

Intranasal immunization and challenge. SPY1 was grown at 37°C in 5% CO2 in C

116

plus Y medium to approximately 2×108 CFU/ml. The cells were collected by

117

centrifugation, washed twice, and then resuspended in sterile PBS. The final vaccine

118

mixture for routine immunization contained 1×108 CFU of SPY1 and 1μg of adjuvant

119

cholera toxin (CT, Sigma-Aldrich) per 20 μl dose. SPY1 was demonstrated to be

120

rapidly cleared from the nasopharynx within 24 hours post intranasal vaccination in

121

BALB/c mice, and no SPY1 colonies were found in mice lungs 12 hours post

122

immunization (14). Similar colonization results were observed in wild C57BL/6 mice,

123

as well as in the mutant mice. C57BL/6 mice were anesthetized with ethyl ether and

124

received vaccine or adjuvant alone intranasally for four times at one week interval.

125

The positive control group comprised mice that received intraperitoneal injection of

126

PPV23 (Chengdu Institute of Biological Products, China) for three times (5 μg per

127

serotype) at one-week interval. For colonization model, two weeks after the last

128

vaccination, mice were anesthetized and challenged with 1×108 CFU of pneumococcal 6

129

strain CMCC 31693 (serotype 19F) and strain TIGR4, respectively. Mice were

130

sacrificed and CFUs in nasal washes and lung homogenates were determined as

131

previously described 72 hours post challenge (18). For lethal intranasal challenge

132

model, C57BL/6 mice were anesthetized and then inoculated intranasally with either

133

pneumococcal strain NCTC 7466 (D39, serotype 2, 5×107 CFU), strain CMCC 31436

134

(serotype 3, 1.5×108 CFU), strain CMCC 31203 (serotype 3, 4×108 CFU), strain

135

CMCC 31207 (serotype 6B, 5×108 CFU) or strain CMCC 31614 (serotype 14, 4×108

136

CFU) in 20 μl of PBS. Survival was monitored for 21 days.

137

Adoptive transfer. Passive/adoptive transfer was performed as described previously

138

(19). Briefly, serum (200 μl) or approximately 1×108 whole spleen cells from

139

immunized or control mice were intraperitoneally transferred to μMT mice or nude

140

mice, respectively. Then, about 24 hours later, the mice were intranasally infected

141

with 5×107 CFU of strain NCTC 7466 (D39, serotype 2) or 1×108 CFU of strain

142

CMCC 31693 (serotype 19F), respectively. Survival rates as well as CFUs in the

143

upper respiratory tract were observed as described above.

144

Killing assays. Phagocytic killings for S. pneumoniae TIGR4 by vaccine-specific

145

antibodies and splenocyte supernatants using differentiated HL-60 cells (promyelotic

146

leukemia cells, CCL240; American Type Culture Collection, Rockville, MD) (20)

147

were performed as described previously (20, 21). Pneumococcal strain TIGR4 (1×103

148

CFU/ 20 μl) were primed with 20 μl of SPY1-specific serum at 37℃ and 5% CO2 for

149

15 min. Following this incubation period, sterile baby rabbit serum as complement

150

resource (10 μl) was added and washed differentiated HL-60 cells (4×105 cells/ 40 μl) 7

151

was added to each well. The assay plate was incubated at 37°C for 45 min with

152

horizontal shaking (220 rpm). After this incubation period, the incubations were

153

diluted and the dilutions were plated onto blood agar plates. To evaluate killing

154

enhanced by vaccine-specific cellular immunity, the neutrophils were added into 24

155

well plates, cultured for 4 hours in 50% conditioned media (from vaccinated or naive

156

splenocytes exposed to killed SPY1 for 3 days) plus 50% complete media (DMEM +

157

10% FBS). The conditioned media was then aspirated, and the pneumococcal strain

158

TIGR4 was added to the wells in fresh DMEM plus 10% fetal bovine serum. Bacteria

159

were added to the wells at a ratio of 10:1 cells to bacteria. The cells were incubated at

160

37℃ for 1 h and after this incubation period, mixtures were diluted and the dilutions

161

were plated onto blood agar plates. Killing was defined as the percent reduction CFUs

162

in wells containing co-cultures of phagocytes cells and bacteria compared to wells just

163

containing bacteria.

164

Cytokines measurement. One week after the final immunization, after washing and

165

removal of red blood cells by hemolysis, splenocytes from immunized or control mice

166

were plated into 24-well tissue culture plates (2×106 cells/well) in 1 ml of DMEM

167

culture medium with 10% fetal calf-serum (Hyclone). Following 72-hour stimulation

168

with concanavalin A (5 mg/ml, Sigma) or 70% ethanol-killed SPY1 (equivalent to 107

169

CFU/ml), levels of IFN-γ, IL-4, IL-10 and IL-17A in the culture supernatants were

170

detected

171

recommendations. Samples were diluted when required.

172

P17 peptide challenge. P17 peptide (KRIWFIPRSSWYERA, purity>95% by HPLC)

by

ELISA

kits

(Biolegend)

8

according

to

the

manufacturer’s

173

was synthesized by GL Biochem (Shanghai, China). The procedure of intraperitoneal

174

injection of P17 to down-regulate Treg cells was performed as described previously

175

(22, 23).

176

Flow cytometry. Mouse lung cell suspensions, at 2×108 cells/ml were incubated with

177

purified anti-Fc receptor blocking antibody (anti-CD16/CD32; eBioscience) before

178

addition of the specific antibodies. A combination of FITC anti-mouse Ly-6G

179

antibody (clone RB6-8C5; BD Pharmingen) and PE anti-mouse CD-11b antibody

180

(clone M1/70; BD Pharmingen) was used for neutrophils and macrophages detection.

181

For Treg cells detection, cells were stained with anti-mouse CD4-FITC (clone RM4-5;

182

eBioscience), anti-mouse CD25-APC (clone PC61.5; eBioscience) followed by

183

anti-mouse FoxP3-APC (clone FJK-16s; eBioscience) according to the manufacturer

184

instructions. Then samples were analyzed using a Becton Dickinson FACScalibur

185

flow cytometer.

186

Statistical analysis. Data were compared using Student’s t-test or the Mann-Whitney

187

U-test. Survival rates were analyzed with the Log Rank test. Differences with P

Mucosal immunization with the live attenuated vaccine SPY1 induces humoral and Th2-Th17-regulatory T cell cellular immunity and protects against pneumococcal infection.

Mucosal immunization with attenuated vaccine can protect against pneumococcal invasion infection, but the mechanism was unknown. Our study found that ...
1MB Sizes 0 Downloads 6 Views

Recommend Documents