Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death

Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death

Accepted Manuscript Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids a...

559KB Sizes 0 Downloads 52 Views

Accepted Manuscript Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death Yao-Kuang Huang, Sheng-Ya Chen, Min Yi Wong, Cheng-Hsun Chiu, Chishih Chu PII:

S0882-4010(18)30288-2

DOI:

https://doi.org/10.1016/j.micpath.2018.12.035

Reference:

YMPAT 3328

To appear in:

Microbial Pathogenesis

Received Date: 19 February 2018 Revised Date:

17 November 2018

Accepted Date: 18 December 2018

Please cite this article as: Huang Y-K, Chen S-Y, Wong MY, Chiu C-H, Chu C, Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death, Microbial Pathogenesis (2019), doi: https://doi.org/10.1016/j.micpath.2018.12.035. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT 1 2 3

Pathogenicity differences of Salmonella enterica serovars Typhimurium, Enteritidis, and Choleraesuis-specific virulence plasmids and clinical S. Choleraesuis strains with large plasmids to the human THP-1 cell death

4 5 6

Yao-Kuang Huanga, Sheng-Ya Chena,b, Min Yi Wonga, and Cheng-Hsun Chiuc,d, Chishih Chub*

8 9

a

10 11

b

12 13

c

14 15

d

RI PT

7

Division of Thoracic and Cardiovascular Surgery, Chiayi Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan

SC

Department of Microbiology, Immunology, and Biopharmaceuticals, National Chiayi University, Chiayi, Taiwan

Molecular Infectious Disease Research Center, Chang Gung Memorial Hospital, Taoyuan, Taiwan

M AN U

Division of Pediatric Infectious Diseases, Department of Pediatrics, Chang Gung Children’s Hospital and Chang Gung University, Taoyuan, Taiwan

16 17

Running title: Different pathogenicity of Salmonella virulence plasmid

18

*Corresponding author.

20

Dr. Chishih Chu

21

Department of Microbiology, Immunology, and Biopharmaceuticals

22

National Chiayi University

23

Address: No.300, University Rd., Chiayi City 60004, Taiwan

24

Tel: 886-5-2717898

25

Fax: 886-52717830

26

E-mail: [email protected]

EP

AC C

27

TE D

19

1

ACCEPTED MANUSCRIPT 28

Abstract Salmonella is a common foodborne and zoonotic pathogen. Only a few serovars carry a

30

virulence plasmid (pSV), which enhances the pathogenicity of the host. Here, we investigated

31

the pathogenicity roles of the pSVs among wild-type, plasmid-less, and complemented S.

32

Typhimurium, S. Enteritidis S. Choleraesuis in invasion, phagocytosis, and intracellular

33

bacterial survival in human THP-1 cells and cell death patterns by flow cytometry and

34

difference in cell death patterns between pig and human S. Choleraesuis isolates with large

35

pSCVs. Virulence plasmid (pSTV) led to slightly increasing cellular apoptosis for S.

36

Typhimurium; virulence plasmid (pSEV) enhanced apoptosis and necrosis significantly for S.

37

Enteritidis; and pSCV reduced apoptosis significantly for S. Choleraesuis. After

38

complementation, pSTV increased the intracellular survival of pSCV-less Choleraesuis and

39

the cytotoxicity against human THP-1 cells. Using the Cytochalasin D to differentiate the

40

invasion of S. Choleraaesuis and phagocytosis of THP-1 cells determined that pSCV were

41

responsible for invasion and phagocytosis at 0 h and inhibited intracellular replication in

42

THP-1 cells, and pSTV were responsible for invasion and increased intracellular survival for

43

S. Choleraesuis in THP-1 cells. The human isolates with large pSCV induced more cellular

44

apoptosis and necrosis than the pig isolates. In conclusion, human S. Choleraesuis isolates

45

carrying large pSCVs were more adapted to human THP-1 cells for more cell death than pig

46

isolates with large pSCV. The role of pSVs in invasion, phagocytosis, intracellular survival

47

and apoptosis differed among hosted serovars.

AC C

EP

TE D

M AN U

SC

RI PT

29

48

49

Keywords: Salmonella, S. Choleraesuis, S. Enteritidis, S. Typhimurium, Virulence

50

plasmid, Apoptosis, Necrosis, intracellular survival

51 2

ACCEPTED MANUSCRIPT 52

1. Introduction Salmonella is one of the most prevalent foodborne pathogens worldwide, and salmonellosis

54

in humans is frequently associated with the consumption of food products of animal origin and their

55

byproducts. Salmonella species constantly face different stress conditions in the environment

56

and in the gastrointestinal tract, such as the extreme pH conditions that prevail in the stomach

57

[1-3]. Among the nontyphoid serovars involved in salmonellosis, S. Choleraesuis and S. Dublin

58

cause bacteremia rather than diarrhea [4]. In C3H/HeN mice, bacteremic non-typhoidal S.

59

Schwarzengrund, S. Bredeney and S. Virchow are low in stimulating systemic disease, S.

60

Schwarzengrund and S. Enteritidis are profound and elongated shedding, and S.

61

Typhimurium cause chronic infection and lower intracellular growth [5], suggesting

62

enhanced invasion and intracellular growth not enough to become bacteremic strains.

63

Furthermore, SPI-2 allows the bacteria to reside and proliferate within macrophages,

64

furthering their reach and survival within the lymphatic system and bloodstream [6-8].

M AN U

SC

RI PT

53

The innate immune system provides the first line of defense against invading

66

microorganisms by inflammatory and antimicrobial responses [9,10]. Further, apoptosis plays

67

an essential role in normal development, homeostasis, and pathogenic processes [11]. Signals

68

such as phosphatidylserine are exposed on the outside of apoptotic bodies to attract

69

macrophages for phagocytosis more efficiently [12]. In Salmonella, S. Dublin induces DNA

70

fragmentation of early apoptosis in human THP-1 cell dependent on phoP expression, not

71

spvB, and caspase-3, 8, and 9 [13]. By contrast, necrosis is morphologically characterized by

72

the swelling of organelles, disruption of the plasma membrane, increase in cell volume, and

73

loss of intracellular contents [14].

AC C

EP

TE D

65

74

The serovar-associated virulence plasmids (pSVs) in Salmonella may correlate with host

75

specificity. These pSVs differ in gene composition, plasmid size and possibly origins [15,16], but

76

all of them encode an 8-kb spvRABCD operon, which is positively regulated by the SpvR and the 3

ACCEPTED MANUSCRIPT sigma factor RpoS in response to the stationary phase of bacterial growth and nutritional limitation

78

within host cells [17-19]. In zebrafish, the spv operon inhibits the functions of neutrophils and

79

macrophages of early innate immune responses and accelerates the replication and

80

dissemination of S. Typhimurium [20]. Furthermore, the spv locus inhibits transcription of

81

the tbx21 gene and suppresses expression of the cytokines IFN-γ, IL-12, and TNF-α, but it

82

enhances the expression of the gata3 gene and the cytokines IL-4, IL-10, and IL-13 [21].

83

Furthermore, SpvB prevents actin polymerization through ADP-ribosylation, facilitates apoptosis

84

[12] and inhibits early autophagy formation in zetafish [22], suppresses autophagosome

85

formation and increases inflammation in murine macrophage-like cells J774A.1 and human

86

epithelial HeLa cells during infection [23].

M AN U

SC

RI PT

77

Expression of rck gene on virulence plasmid is regulated by the temperature and quorum-

88

sensing regulator SdiA on the chromosome [24], demonstrating the interaction between genome

89

and virulence plasmid during infection. Previously, we reported that large pSCVs are evolved

90

through recombination of the R plasmid and the 50-kb pSCV in human and pig isolates

91

[25,26]. However, we did not determine the virulence among these clinical human and swine

92

isolates with larger pSCV.

EP

93

TE D

87

2. Materials and Methods

95

2.1 Bacterial strains

96

AC C

94

The bacterial strains listed in Table 1 were employed to investigate the role of pSVs in three

97

serovars and in clinical S. Choleraesuis swine strains CN30 and CN32, and human strains SCB52,

98

SCB74, SCB97, SCB130, and MS22872 with large pSCV through pathogenicity assays. These

99

strains included wild-type S. Typhimurium (OU5045), S. Enteritidis (OU7130), and S. Choleraesuis

100

(OU7085) with 90-kb pSTV, 60-kb pSEV and 50-kb pSCV, respectively; their pSV-less strains

101

OU5046, OU7067, and OU7266; and complemented strains OU5193, OU5194, OU7294, and 4

ACCEPTED MANUSCRIPT OU7295 with transposon Tn5 on the pSVs conferring kanamycin resistance (KmR). All above

103

strains were kindly provided by Dr. Jonathan T. Ou and examined by plasmid analysis and PCR

104

amplification of invC for Salmonella and spvA for pSV (Fig. 1). All of the bacterial strains used for

105

cell lysis assays grew routinely on xylose lysine deoxycholate (XLD) plates. A single black colony

106

was grown overnight in Luria-Bertani (LB) broth at 37 °C. All strains for

107

2.2 Cell and bacterial culture

RI PT

102

Murine RAW 264.7 cells grew in RPMI 1640 medium (Sigma, R6504) supplemented

109

with 10% preheated fetal bovine serum (FBS, Sigma, SAB-1900SC-500 ml), D-glucose (J.T.

110

Baker, 1916-01), sodium bicarbonate (Sigma, S5761), 10 mM HEPES (Sigma, H0887), 1

111

mM sodium pyruvate (Sigma, S8636), and a 1% antibiotic solution of penicillin,

112

streptomycin, and amphotericin (Sigma, A5955). Human monocyte THP-1 cells were

113

incubated routinely in RPMI 1640 medium (GIBCO, ThermoFisher Scientific, Taiwan)

114

supplements with 10% FBS, 10 mM HEPES buffer, 2.5 mM sodium pyruvate, 2 mM L-

115

glutamine, and 100 µg/ml penicillin/streptomycin at 37 °C and 5% CO2.

TE D

M AN U

SC

108

For the phagocytosis and intracellular survival tests, 50 µM of phorbol myristate acetate

117

(Sigma, P8139) were added to THP-1 cells to stimulate the macrophage differentiation at

118

37°C for 48 h. The bacterial strains and E. coli (ATCC 8739) were incubated at 37°C for 12 h,

119

respectively. After centrifugation for 10 min at 3,000 x g (KUBOTA 2010, Kubota, Japan),

120

the pellets were washed with phosphate-buffered saline (PBS) several times and were

121

resuspended with PBS solution. The OD550 of the bacterial solutions was measured using a

122

Bio-Rad SmartSpecTM 3000 (approximately 8.6 × 108 bacteria per mL at OD550 = 1). The

123

phagocytosis, cell death, and intracellular survival tests were performed in three independent

124

experiments with two repeats each.

125

2.3 Intracellular survival and phagocytosis

AC C

EP

116

5

ACCEPTED MANUSCRIPT In the intracellular survival test, 1 × 107 bacteria of each strain were mixed with 1 × 106

127

differentiated THP-1 cells (MOI = 10), respectively, in each well of a 24-well plate (Becton,

128

Dickinson and Company) and incubated with RPMI 1640 medium without antibiotics at

129

37°C with 5% CO2 for 30 min or 1 h. The extracellular bacteria were killed by treating with

130

gentamicin at a final concentration of 50 µg/mL for 1 h (corresponding to time 0 h). After the

131

cell lysis ith 0.5 mL of 1% sodium deoxycholate (Sigma-Aldrich), 100 µL aliquots of the

132

solution were plated on individual LB plates. The viable bacteria were counted for each plate

133

after incubation at 37°C for 24 h. Furthermore, the invasion of the differentiated THP-1 cells

134

were evaluated by treating with 5 µg/ml of cytochalasin D for 1 h at 37°C to inhibit

135

phagocytosis. Subsequent infection and intracellular bacteria number were performed as

136

described above.

137

2.4 Cytotoxicity of different serovars and clinical S. Choleraesuis strains to human

138

THP-1 cells

M AN U

SC

RI PT

126

Human monocyte THP-1 cells were cultured routinely, as described earlier.

140

Furthermore, the bacteria and cell mixtures at MOI = 10:1 as above were incubated at 37°C

141

for different periods depending on the experiment and then washed twice with PBS. After

142

centrifugation and wash with PBS twice, the cells were treated with 0.25% trypsin in 0.53

143

mM EDTA solution for 5 min and then 800 µl of RPMI 1640 medium with 10% FBS were

144

added to each well. The cells were pelleted at 800 x g for 10 min. After washing with PBS,

145

the cells were stained with propidium iodide (PI) and annexin V using an AnnexinV-FITC

146

Apoptosis Detection Kit (Cat. No. AVK250, Strong Biotech Corporation, Taiwan) for 10–15

147

min. The fluorescence of PI and annexin V was measured by flow cytometry, and the data

148

were analyzed using WinMDI software.

149

2.5 Statistical analysis

AC C

EP

TE D

139

6

ACCEPTED MANUSCRIPT Differences among multiple experimental groups were assessed with Student’s t test, a

151

one-way ANOVA, and a post-hoc Tukey’s HSD test using SPSS 18.0 software. The results

152

were considered statistically significant when p < 0.05, and all data were displayed as the

153

mean ± SD.

154

3. Results

155

3.1 Interaction of wild-type Salmonella serovars with different monocyte cell types

RI PT

150

Intracellular bacterial analysis of three serovars demonstrated that the highest bacterial

157

number was observed for broad-host S. Typhimurium and S. Enteritidis in mouse RAW 264.7

158

cells and for S. Enteritidis in human THP-1 cells and the lowest number for narrow-host S.

159

Choleraesuis in both cell lines (Table 2). However, intracellular number of S. Choleraesuis

160

was not change in RAW 264.7 cells compared to a gradual increase from 1 h to 2 h in the

161

human THP-1 cells. Furthermore, intracellular E. coli was not observed in the human THP-1

162

cells, compared to lowest number in mouse RAW 264.7, suggesting intracellular bacterial

163

number depending on cell lines. During intracellular bacterial survival analysis, we observed

164

two cell populations at the invasion and intracellular survival stages, and changes in the ratios

165

of the three aforementioned serovars were observed (Table 3). During the invasion stage, S.

166

Choleraesuis exhibited the lowest ratio and S. Typhimurium exhibited the highest ratio. At the

167

intracellular survival stage, the ratio increased gradually for S. Typhimuirum, and decreased

168

for S. Enteritidis. However, S. Cholearesuis exhibited the highest ratio at 0 h, the lowest ratio

169

at 1 h, and then an increase in ratio from 1 h to 4 h. This change in cell size may be related to

170

cell death.

171

3.2 Roles of the pSVs of three serovars in cell death

AC C

EP

TE D

M AN U

SC

156

172

All serovars induced more apoptosis than necrosis and differed in their effects on cell

173

viability, apoptosis, and necrosis (Fig. 2). Three wild-type serovars caused different viable

174

cell patterns. S. Typhimurium infection showed the highest number of viable cells at 0.5 h 7

ACCEPTED MANUSCRIPT and then decreased at 1 h and 2 h. In contrast, S. Choleraesuis infection showed reverse

176

phenomenon of increasing the viable cell number. However, S. Enteritidis infection killed all

177

cells. Apoptosis analysis showed that S. Enteritidis caused the highest rate of apoptosis, S.

178

Typhimurium showed an increase of apoptosis from 0.5 h to 2 h and S. Choleraesuis showed

179

an significant decrease of apoptosis.

RI PT

175

The patterns of cellular viability, apoptosis and necrosis differed between wild-type and

181

pSTV-less S. Typhimurium. pSTV-less OU5046 increased cellular ability and decreased

182

apoptosis positively associated with the incubation time, compared that both pSTV-

183

complemented strains OU5193 and OU5194 increased cellular viability and reduced necrosis

184

at time-dependent pattern. In the case of S. Enteritidis, the wild-type and complemented

185

strains demonstrated identical effects: no viable cells and nearly 100% apoptosis. However,

186

treatment with the pSEV-less strain yielded the highest rate of viable cells and necrosis, and

187

the least apoptotic rate. The S. Choleraesuis wild-type OU7085 and the pSCV-complemented

188

OU7294 demonstrated similar patterns of influence, increasing ratio of viable cells and

189

decreasing the ratio of apoptosis as well as necrosis. By contrast, the pSCV-less OU7266

190

decreased the rate of viable cells and increased apoptotic rate.

TE D

M AN U

SC

180

The role of the pSVs differed among serovars with slightly increase of apoptosis and

192

necrosis for pSTV of S. Tphimurium, a significantly increase in cell death for S. Enteritidis,

193

and a decrease of cell death for S. Choleraesuis.

194

3.3 Intracellular survival of wild-type, pSCV-less and pSCV-complementary S.

195

Choleraesuis strains in human THP-1 cells and infection related cell death

AC C

EP

191

196

These three strains differed in cell death and intracellular bacterial patterns (Table 4).

197

Wild-type strain displayed an increase of cell death and very few intracellular bacteria. By

198

contrast, the pSCV-less OU7266 increased in rate of cell death and higher intracellular

199

bacteria ranged from 2- to 7.6 x 103 bacteria per 103 cells. Nevertheless, Tn5-inserted pSCV 8

ACCEPTED MANUSCRIPT complementary OU7294 strain displayed similar pattern as pSCV-less strain in apoptotic

201

from 0 h to 4 h, but with higher apoptosis rate at 8 h and significantly less intracellular

202

bacteria than the pSCV-less strain and higher than the wild-type. These results confirmed

203

above pathogenicity roles of the pSCV in apoptosis and displayed that pSCV may inhibited

204

intracellular survival.

205

3.4 The role of pSVs in the invasion, phagocytosis and intracellular survival

RI PT

200

Cytochalasin D treatment can inhibit actin polymerization and, subsequently, the

207

phagocytosis of bacteria by human macrophage-like THP-1 cells. Therefore, such a treatment

208

can differentiate Salmonella invasion from the phagocytosis of macrophages. At 0 h, wild-

209

type OU7085 exhibited a limited intracellular bacterial number derived from phagocytosis,

210

whereas pSCV-less OU7226 exhibited the highest intracellular number with almost equal

211

ratios of invasion (48.4%) and phagocytosis (51.6%), suggesting pSCV may increase of

212

phagocytosis and invasion. Furthermore, intracellular bacteria changed over time (Table 5).

213

By contrast, the pSTV-complemented OU7194 demonstrated that invasion was the cause

214

(100%) of intracellular bacteria, and an increase in intracellular survival occurred in a time-

215

dependent manner. These results imply pSCV responsible for invasion and phagocytosis at o

216

h and following by decreasing intracellular survival, and pSTV responsible for invasion and

217

increase in intracellular survival in S. Choleraesuis.

218

3.5 Cytotoxicity of clinical human and swine S. Choleraesuis isolates with large pSCV to

219

human THP-1 cells

AC C

EP

TE D

M AN U

SC

206

220

Cytotoxicity against differentiated THP-1 cells differed among the human and pig

221

isolates in comparison to the laboratory OU7085 derived from humans (Table 7). At 2-h post-

222

infection, the human isolates displayed the lowest viable cells and the highest rate of

223

apoptosis, whereas the pig isolates exhibited the highest rate of necrosis. At 4-h post-infection, 9

ACCEPTED MANUSCRIPT both the human and pig isolates exhibited significantly higher apoptotic rates and lower

225

viable cells compared with OU7085. At 8-h post-infection, the human isolates displayed the

226

highest apoptotic rates and the lowest rate of viable cells and necrosis.

227

4. Discussion

RI PT

224

Different host range may be regulated by the gene acquisition via horizontal gene

229

transfer with plasmids, transposons, and phages, the loss of genes or their function

230

(pseudogenes) and interactions with the host’s immune system and commensal organisms

231

during infection [27]. S. Typhimurium, S. Enteritidis, and S. Choleraesuis differ in the host-

232

specific symptoms and invasion types. Such difference may be resulted from gene

233

inactivation more in narrow-host-range S. Choleraeuis than in broad-host-range S.

234

Typhimurium [28], more intracellular bacterial number of broad-host S. Typhimurium and S.

235

Enteritidis than S. Choleraesuis in mouse RAW 264.7 macrophage cells and human THP-1

236

cells (Table 2), gene interaction between virulence plasmid and chromosome [24] and serovar-

237

specific genes, such that S. Choleraesuis lacks the enzymes required for citrate metabolism,

238

including tricarballylate dehydrogenase and oxaloacetate decarboxylase, to evade activation

239

of the NLPR3 inflammasome and to promote pathogenicity [29]. In macrophages, Salmonella

240

evolves mechanisms to avoid host immune responses. Salmonella can quench arginine away

241

from the inducible NO synthase (iNOS) pathway via arginine transporters (ArgT) for growth

242

and survival [30]. Thus, S. Typhimurium with ArgT expression can evade being killed by

243

macrophages and cause a severe systemic infection.

AC C

EP

TE D

M AN U

SC

228

244

During intracellular growth, 20 pSTV genes in S. Typhimuriun were upregulated,

245

including repA, ccdA, rsd, virE, spvRA, tlpA, samAB, traTD, and trbH [31]. However, a few

246

of these genes are not present in pSEV and pSCV [10]. These missing genes may change of

247

the distribution of human THP-1 cell populations (Table 3). Such change may be due to

248

differences in apoptosis rather than necrosis (Figure 1) and conditions of intracellular bacteria. 10

ACCEPTED MANUSCRIPT 249

In S. Typhimurium, genes ydgT, recA, and corA are involved in the cytosolic proliferation

250

and asmA reduced the cytosolic replication [32] and a 597-nt IesR-1 sRNA encoded by the

251

pSTV in dormant S. Typhimurium persisting inside fibroblasts is upregulated [33]. Early study reported that the DNA fragmentation was related to phoP expression, not

253

related to SPI-1 and SPI-2, spvB, and caspase gene expression [11]. In this study, the

254

pathogenicity roles of the pSVs in their host serovars and pSTV in pSCV-less S. Choleraesuis

255

differed. In the original hosted serovar, pSTV and pSEV differed the apoptotic ability with

256

complete cell killing for S. Enteritidis and lesser apoptosis for S. Typhimurium, but pSCV

257

inhibited the apoptotic ability (Figure 1) and intracellular survival of S. Choleraesuis (Table

258

4). Furthermore, cytochalasin D demonstrated pSCV inhibited phagocytosis of the human

259

THP-1 cells and S. Choleraesuis invasion mainly for intracellular bacteria. By contrast, pSTV

260

enhanced both phenomena, although the increase was limited (Table 5). All above results

261

imply that pSCV and pSTV may play different pathogenicity roles for S. Choleraesuis and

262

co-evolution of the pSVs with their hosted serovars.

TE D

M AN U

SC

RI PT

252

Host adaptation and/or evolution may be associated with genome degradation, such as

264

pseudogene formation and deletion. Investigation of multiple S. Enteritidis isolates with

265

mutation in the mismatch repair gene mutS from an interleukin (IL)-12 β-1 receptor-deficient

266

individual with a 15-year recurrent blood-borne infection demonstrated genome changes

267

associated within-host evolution on the timescale [34]. Furthermore, the evolution of the 50-

268

kb pSCV to become large pSCVs (75–140 kb) is through the recombination of the R plasmid

269

[26] and the involvement of co-evolution of large pSCV associated with human isolates, not

270

pig isolates, caused the higher apoptotic rates in human THP-1 cells (Table 6). Nevertheless,

271

the pSTVs in human-specific S. Typhi and Paratyphi A did not enhance their pathogenicity

272

in infant mice [35]. These data demonstrate that coevolution of pSVs with their host serovar

AC C

EP

263

11

ACCEPTED MANUSCRIPT 273

plays an important role in host-specific infection. However, a deeper investigation is required

274

to determine whether the exchanged pSVs remain serovar-specific.

275

5. Conclusion The pSVs differed in size and genes as well as their pathogenicity role among S.

277

Typhimuirum, S. Enteritidis, and S. Choleraesuis. The pSTV and pSEV enhanced the

278

apoptosis with difference in ratio between these two, but pSCV inhibited the apoptosis.

279

Further, pSCV promoted invasion and phagocytosis as well as decreased intracellular

280

bacterial survival, and pSTV stimulated invasion and intracellular survival in S. Choleraesuis,

281

demonstrating a difference in host adaptation and coevolution of chromosome characteristics

282

and/or virulence plasmids for the virulence and adaptation to host species.

M AN U

SC

RI PT

276

283 284

Competing interests

285

The authors declare that they have no competing interests.

TE D

286 Acknowledgements

288

This work was supported by grants from the Ministry of Science and Technology (MOST 102-2320-

289

B-415-004 for CC) Executive Yuan, and Chiayi Chang Gung Memorial Hospital (grant numbers:

290

CMRPG6B0503, CMRPG6E0423, and CMRPG6G0101 for YKH), ROC (Taiwan).

AC C

291

EP

287

292

References

293

1. J.W. Foster, H.K.Hall. Inducible pH homeostasis and the acid tolerance response of

294 295 296

Salmonella typhimurium. J Bacteriol. 173(1991)5129-5135. 2. Foster JW. The acid tolerance response of Salmonella typhimurium involves transient synthesis of key acid shock proteins. J Bacteriol 175(1993)1981-1987.

12

ACCEPTED MANUSCRIPT 297

3. A. Álvarez-Ordóñez, M. Prieto, A. Bernardo, C. Hill, M. López. The Acid Tolerance

298

Response of Salmonella spp.: an adaptive strategy to survive in stressful environments

299

prevailing in foods and the host. Food Res Int. 45(2012)482-492. 4. M.J. Blaser, R.A. Feldman. Salmonella bacteremia: reports to the Centers for Disease Control, 1968-1979. J Infect Dis. 143(1981)743-746

301

RI PT

300

5. J. Suez J, Porwollik, A. Dagan, A. Marzel, Y.I. Schorr, P.T. Desai, V. Agmon, M.

303

McClelland, G. Rahav, O. Gal-Mor. Virulence gene profiling and pathogenicity

304

characterization of non-typhoidal Salmonella accounted for invasive disease in humans.

305

PLOS ONE 8 (2013) e58449.

SC

302

6. F. Garcia-del Portillo, M.B. Zwick, K.Y. Leung, B.B. Finlay. Salmonella induces the

307

formation of filamentous structures containing lysosomal membrane glycoproteins in

308

epithelial cells. Proc Natl Acad Sci U S A. 90 (1993)10544-10548.

M AN U

306

7. A. Gallois, J.R. Klein, L.A.H. Allen, B.D. Jones, W.M. Nauseef. Salmonella pathogenicity

310

island 2-encoded type III secretion system mediates exclusion of NADPH oxidase

311

assembly from the phagosomal membrane. J Immunol.166 (2001)5741-5748.

312

8.

TE D

309

D. Chakravortty, I. Hansen-Wester, M. Hensel. Salmonella pathogenicity island 2 mediates protection of intracellular Salmonella from reactive nitrogen intermediates. J

314

Exp Med. 195(2002)1155-1166.

316 317 318 319 320

9.

AC C

315

EP

313

P. Broz, M.B. Ohlson, D.M. Monack. Innate immune response to Salmonella

typhimurium, a model enteric pathogen. Gut Microbes. 3(2012)62-70. 10. D. Hurley, M.P. McCusker, S. Fanning, M. Martins. Salmonella-host interactions modulation of the host innate immune system. Front Immunol. 5(2014)481. 11. E. Valle, D.G. Guiney. Characterization of Salmonella-induced cell death in human macrophage-like THP-1 cells. Infect Immun. 73(2005)2835-2840.

13

ACCEPTED MANUSCRIPT 321

12. R.G. Deschesnes, J. Huot, K. Valerie, J. Landry. Involvement of p38 in apoptosis-

322

associated membrane blebbing and nuclear condensation. Mol Biol Cell 12(2001)1569-

323

1582.

325 326 327

13. I.K. Poon, C.D. Lucas, A.G. Rossi, K.S. Ravichandran. Apoptotic cell clearance: basic biology and therapeutic potential. Nat Rev Immunol. 14(2014)166-180.

RI PT

324

14. W. Wu, P. Liu, J. Li. Necroptosis: an emerging form of programmed cell death. Crit Rev Oncol Hematol. 82(2012):249-258.

15. C. Chu, S-F. Hong, C. Tsai, W-S. Lin, T-P. Liu, J.T. Ou. Comparative physical and

329

genetic maps of the virulence plasmids of Salmonella enterica serovars Typhimurium,

330

Enteritidis, Choleraesuis, and Dublin. Infect Immun. 67(1999)2611-2614.

332

M AN U

331

SC

328

16. C. Chu, C.H. Chiu. Evolution of the virulence plasmids of non-typhoid Salmonella and its association with antimicrobial resistance. Microbes Infect. 8(2006)1931-1936. 17. F.C. Fang, M. Krause, C. Roudier, J. Fierer, D. Guiney. Growth regulation of a

334

Salmonella plasmid gene essential for virulence. J Bacteriol. 173(1991)6783-6789.

335

18. M. Krause, F.C. Fang, D. Guiney D. Regulation of plasmid virulence gene expression in

336

Salmonella dublin involves an unusual operon structure. J Bacteriol. 174(1992)4482-

337

4489.

EP

TE D

333

19. F.C. Fang, S.J. Libby, N.A. Buchmeier, P.C. Loewen, J. Switala, J. Harwood, D.G.

339

Guiney. The alternative sigma factor katF (rpoS) regulates Salmonella virulence. Proc

340

Natl Acad Sci USA. 89(1992)11978-11982.

AC C

338

341

20. S.Y. Wu, L.D. Wang, J.L. Li, G.M. Xu, M.L. He, Y.Y. Li, R. Huang. Salmonella spv

342

locus suppresses host innate immune responses to bacterial infection. Fish Shellfish

343

Immunol. 58(2016)387-396.

14

ACCEPTED MANUSCRIPT 344

21. S.Y. Wu, L.D. Wang, G.M. Xu, S.D. Yang, Q.F. Deng, Y.Y. Li, R. Huang. spv locus

345

aggravates Salmonella infection of zebrafish adult by inducing Th1/Th2 shift to Th2

346

polarization. Fish Shellfish Immunol. 67(2017)684-691. 22. Y.Y. Li, T. Wang, S. Gao, G.M. Xu, H. Niu, R. Huang, S.Y. Wu. Salmonella plasmid

348

virulence gene spvB enhances bacterial virulence by inhibiting autophagy in a zebrafish

349

infection model. Fish Shellfish Immunol. 49(2016)252-259.

RI PT

347

23. Y. Chu, S. Gao, T. Wang, J. Yan, G. Xu, Y. Li, H. Niu, R. Huang, S. Wu. A novel

351

contribution of spvB to pathogenesis of Salmonella Typhimurium by inhibiting

352

autophagy in host cells. Oncotarget 7(2016)8295-8309.

SC

350

24. N. Abed, O. Grépinet, S. Canepa, G.A. Hurtado-Escobar, N. Guichard, A. Wiedemann, P.

354

Velge, I. Virlogeux-Payant. Direct regulation of the pefI-srgC operon encoding the Rck

355

invasin by the quorum-sensing regulator SdiA in Salmonella Typhimurium. Mol

356

Microbiol. 942(2014)254-271.

M AN U

353

25. C. Chu, C.H. Chiu, W.Y. Wu, C.H. Chu, T.P. Liu, J.T. Ou. Large drug resistance

358

virulence plasmids of clinical isolates of Salmonella enterica serovar Choleraesuis.

359

Antimicrob Agents Chemother. 45(2001)2299-2303.

TE D

357

26. J.I. Tzeng, C.H. Chu, S.W. Chen, C.M. Yeh, C.H. Chiu, C.S. Chiou, J.H. Lin, C. Chu.

361

Reduction of Salmonella enterica serovar Choleraesuis carrying large virulence

362

plasmids after the foot and mouth disease outbreak in swine in southern Taiwan, and

363

their independent evolution in human and pig. J Microbiol Immunol Infect.

364

45(2012)418-425.

AC C

EP

360

365

27. S.L. Foley, T.J. Johnson, S.C. Ricke, R. Nayak, J. Danzeisen. Salmonella pathogenicity

366

and host adaptation in chicken-associated serovars Microb Mol Bio Rev. 77(2013)582–

367

560

15

ACCEPTED MANUSCRIPT 368

28. C.H. Chiu, P. Tang, C. Chu, S. Hu, Q. Bao, L. Yu, Y.Y. Chou, H.S. Wang, Y.S. Lee. The

369

genome sequence of Salmonella enterica serovar Choleraesuis, a highly invasive and

370

resistant zoonotic pathogen. Nucleic Acids Res. 33(2005)1690-1698. 29. M.A. Wynosky-Dolfi, A.G. Snyder, N.H. Philip, P.J. Doonan, M.C. Poffenberger, D.

372

Avizonis, E.E. Zwack, A.M. Riblett, B. Hu, T. Strowig, R.A. Flavell, R.G. Jones, B.D.

373

Freedman, I.E. Brodsky. Oxidative metabolism enables Salmonella evasion of the

374

NLRP3 inflammasome. J Exp Med. 211(2014)653-668.

RI PT

371

30. P. Das, A. Lahiri, A. Lahiri, M. Sen, N. Iyer, N. Kapoor, K.N. Balaji, D. Chakravortty.

376

Cationic amino acid transporters and Salmonella Typhimurium ArgT collectively

377

regulate arginine availability towards intracellular Salmonella growth. PLoS One.

378

5(2010)e15466.

M AN U

SC

375

31. S. Eriksson, S. Lucchini, A. Thompson, M. Rhen, J.C. Hinton. Unravelling the biology

380

of macrophage infection by gene expression profiling of intracellular Salmonella

381

enterica. Mol Microbiol. 47(2003)103-118.

TE D

379

32. M. Wrande, H. Andrews-Polymenis, D.J. Twedt, O. Steele-Mortimer, S. Porwollik, M.

383

McClelland, L.A. Knodler. Genetic determinants of Salmonella enterica Serovar

384

Typhimurium poliferation in the ctosol of epithelial cells. Infect Immun. 84(2016)3517-

385

3526.

AC C

EP

382

386

33. J. Gonzalo-Asensio, A.D. Ortega, G. Rico-Perez, M.G. Pucciarelli, F. Garcıa-del Portillo.

387

A novel antisense RNA from the Salmonella virulence plasmid pSLT expressed by

388

non-growingbacteria inside eukaryotic cells. PLoS One 8(2013) e77939.

389

34. E.J. Klemm, E. Gkrania-Klotsas, J. Hadfield, J.L. Forbester, S.R. Harris, C. Hale, J.N.

390

Heath, T. Wileman, S. Clare, L. Kane, D. Goulding, T.D. Otto, S. Kay, R. Doffinger,

391

F.J. Cooke, A. Carmichael, A.M.L. Lever, J. Parkhill, C.A. MacLennan, D.

392

Kumararatne, G. Dougan, R.A. Kingsley. Emergence of host-adapted Salmonella 16

ACCEPTED MANUSCRIPT 393

Enteritidis through rapid evolution in an immunocompromised host. Nat Microbiol.

394

1(2016):15023. 35. M.J. Santander, R. Curtiss III. Salmonella enterica Serovars Typhi and Paratyphi A are

396

avirulent in newborn and infant mice even when expressing virulence plasmid genes of

397

Salmonella Typhimurium. J Infect Dev Ctries. 4(2010)723.

AC C

EP

TE D

M AN U

SC

398

RI PT

395

17

ACCEPTED MANUSCRIPT 399

Legends

400

Figure 1. Plasmid and PCR analysis of wildtype, virulence plasmid (pSV)less, and pSV

401

complementatry strains of S.Typhimurium, S. Enteritidis, and S. Choleraesuis. A)

402

Plasmid analysis. B) PCR identification of

403

identification and 571-bp spvC fragment for pSV identification. M: 100 bp marker, N:

404

Negative, 1: OU5045, 2: OU5046, 3: OU5193, 4: OU5194, 5: OU7130, 6: OU7067, 7:

405

OU7415, 8: OU7085, 9: OU7266, 10: OU7194, 11: and OU7294.

RI PT

244-bp invA fragment for Salmonella

SC

406

Figure 2. Difference in death type and rate of human THP-1 cells infected by wildtype,

408

virulence plasmid (pSV)less, and pSV complementatry strains of S.Typhimurium, S.

409

Enteritidis, and S. Choleraesuis.

AC C

EP

TE D

M AN U

407

18

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

1

ACCEPTED MANUSCRIPT Table 1. Characteristics of S. Typhimurium, S. Enteritidis and S. Choleraesuis strains Serovars

Strains

The pSV Characteristics

OU5045

Wild type with a 90-kb pSTV

OU5046

pSTV-less strain derived from wild type

S. Typhimurium

RI PT

OU5193

Tn5-tagged pSTV complemented strains OU5194

OU7067

pSEV-less strain derived from wild type

OU7415

Tn5-tagged pSEV complemented strain

OU7085

Wild type with a 50-kb pSCV

OU7266 OU7194

SC

Wild type with a 60-kb pSEV

M AN U

S. Enteritidis

OU7130

pSCV-less strain derived from wild type

Tn5-tagged pSTV complemented strains OU7294

S. Choleraesuis

TE D

CN30

CN32

EP

SCB52 SCB97

Clinical isolates with a large pSCV

AC C

SCB130 SCB74

MS22872

2

ACCEPTED MANUSCRIPT

Table 2. Intracellular numbers of different serovars in mouse and human macrophage cells Mouse RAW 264.7 cells

Human THP-1 cells

Strains 2h

4h

1h

2h

4h

4 x101

9 x101

4 x101

7.2 x101

0

0

0

>3 x 103

OU5045 OU7085 2.3 x102

1.0 x 103

3.33 x 102 2.37 x 103 2.05 x 104

4.8 x 102 4.3 x 102

>3 x 103

7.67x101

3.47x103

3.17 x 103

1.52 x 104 7.30 x 104 4.10 x 105

AC C

EP

TE D

M AN U

OU7130

RI PT

1h

SC

E. coli

0h

3

ACCEPTED MANUSCRIPT Table 3. Ratio of larger cell population over small cell population of human THP-1 cells Invasion

Intracellular survival

Serovars 0

1

2

4

OU5045

0.536

1.022

1.584

2.458

3.346

OU7085

0.441

1.664

0.960

1.205

1.469

OU7130

0.503

1.621

1.021

0.837

0.710

AC C

EP

TE D

M AN U

SC

RI PT

1h

4

ACCEPTED MANUSCRIPT Table 4. Intracellular number and cytotoxicity of S. Choleraesuis isolates in human THP-1 cells after infection with MOI=10 Item

0h

2h

4h

8h

Control

Cell death (%)

0

3

4

5

Cell death (%)

0

24

31

32

Intracellular No/103 cells)

0.03

0.17

1.9

2.8

Cell death (%)

0

27

34

45

Intracellular No/103 cells

2 x103

4 x103

7.6 x 103

3.6 x103

pSCV-complemented

Cell death (%)

0

25

37

58

OU7294

Intracellular No/103 cells

2.5 x102

5.6 x 101

Wild type OU7085

SC

pSCV-less OU7266

RI PT

Strain

AC C

EP

TE D

M AN U

2.8 x 101 3.4 x 102

5

ACCEPTED MANUSCRIPT

Table 5. Invasion of different S. Choleraesuis strains and phagocytosis of human THP-1 cells Bacteria Cytochalasin D 0h 2h 4h 0

0

0

+

0

0

0

-

0.18/100

0.15/100

1.8/100

+

0.02/11.1

0/0

0.06/3.3

(-) – (+)

0.16/88.9

0.15/100

1.74/96.7

1.50/96.2

-

3.1 x 103/100

7.4 x 103/100

7.2 x 103/100

9.4 x 103/100

+

1.5 x 103/48.4

1.8 x103/24.3 2.7 x 103/37.5

2.7 x 103/28.7

1.6 x 103/51.6 5.6 x 103/75.7 4.5 x 103/62.5

6.6 x 103/71.3

-

1.7 x 101/100

6.9 x 102/100

4.7 x 102/100

+

2.3 x 101/100 6.9 x 101/40.6 1.4 x 102/20.2

2.1 x 102/44.7

1.2 x 101/59.4 5.5 x 102/79.8

2.6 x 102/55.3

(-) – (+)

-6/0

AC C

(-) – (+)

0

2.6/100

SC

1.7 x 102/100

EP

pSTV-complemented OU7194

M AN U

TE D

pSCV-less OU7266

0

RI PT

None

Wild type OU7085

8h

6

0.1/3.8

ACCEPTED MANUSCRIPT Table 6. Cell deaths of human macrophage-like THP-1 cells infected with OU7085 and clinic swine isolates and human isolates with large pSCV in different periods

8h

Apoptosis

Necrosis

OU7085

89.0±0.31a

9.8±0.27b

1.3±0.20b

Pig isolates

87.2±0.98a

9.6±0.56b

3.2±0.67a

Human isolates

77.0±0.90b

22.0±0.83a

OU7085

90.5±0.46a

8.2±0.26c

Pig isolates

85.9±1.05b

10.1±0.51b

Human isolates

81.3±0.63c

17.6±0.56a

1.1±0.17b

OU7085

90.8±0.26a

7.5±0.45c

1.7±0.46a

Pig isolates

88.3±1.23ab

9.9±1.02b

1.9±0.32a

Human isolates

87.3±0.50b

12.4±0.48a

0.3±0.03b

RI PT

4h

Viability

1.0±0.87b

1.3±0.24b 4.0±0.87a

SC

2h

Source

M AN U

Time

AC C

EP

TE D

Pig isolates: CN30 and CN32; human isolates: SCB52, SCB74, SCB97, SCB130, and MS22872. All values are the mean ± SE. a-c indicate significant differences between sources.

7

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

AC C

EP

TE D

M AN U

SC

RI PT

ACCEPTED MANUSCRIPT

ACCEPTED MANUSCRIPT Highlight

The pSVs differed in size and genes as well as their pathogenicity role among S.

RI PT

Typhimuirum, S. Enteritidis, and S. Choleraesuis. The pSTV and pSEV enhanced the apoptosis with difference in ratio between these two, but pSCV inhibited the apoptosis. Further, pSCV promoted invasion and phagocytosis as well as decreased intracellular bacterial survival, and pSTV

SC

stimulated invasion and intracellular survival in S. Choleraesuis, demonstrating a difference in host

AC C

EP

TE D

virulence and adaptation to host species.

M AN U

adaptation and coevolution of chromosome characteristics and/or virulence plasmids for the