Susceptibility to ticks and Lyme disease spirochetes is not affected in ...

2 downloads 62 Views 2MB Size Report
Feb 16, 2016 - Hosts. 31 experimentally co-infected with H. polygyrus and .... The remaining carcass was placed in screw top jars over water to allow ...
IAI Accepted Manuscript Posted Online 16 February 2016 Infect. Immun. doi:10.1128/IAI.01309-15 Copyright © 2016 Maaz et al. This is an open-access article distributed under the terms of the Creative Commons Attribution 4.0 International license.

1 2

Susceptibility to ticks and Lyme disease spirochetes is not affected in mice co-infected with nematodes

3 4 5 6

Denny Maazac, Sebastian Rauscha, Dania Richterb, Jürgen Krückenc, Anja A. Kühld, Janina Demelerc, Julia Blümkec, Franz-Rainer Matuschkae, Georg v. Samson-Himmelstjernac, Susanne Hartmanna#

7

Institute of Immunology, Centre of Infection Medicine, Freie Universität Berlin, Germanya

8

Environmental Systems Analysis, Institute of Geoecology, Technical University of Braunschweig,

9

Germanyb

10

Institute of Parasitology and Tropical Veterinary Medicine, Freie Universität Berlin, Germanyc

11

Department of Medicine I for Gastroenterology, Infectious Disease and Rheumatology, Research Center

12

ImmunoSciences, Charité Berlin, Germanyd

13

Outpatient Clinic, University of Potsdam, Germanye

14 15

Running head: Nematode/tick co-infection

16 17

# Address correspondence to Susanne Hartmann, [email protected]

18 19 20 21 22

Phone: +49 30-838-51824 Fax: +49 30-838-451824 D.M. and S.R contributed equally to this work.

1

23 24 25

Abstract

26

Small rodents serve as reservoir hosts for tick-borne pathogens, such as spirochetes causing Lyme disease.

27

Whether natural co-infections with other macroparasites alter the success of tick feeding, anti-tick immunity

28

and the host’s reservoir competence for tick-borne pathogens remains to be determined. In a parasitological

29

survey of wild mice in Berlin approximately 40% of Ixodes ricinus infested animals harboured the nematode

30

Heligmosomoides spp. simultaneously. We, therefore, aimed to analyse the immunological impact of the

31

nematode/tick co-infection as well as its effect on the tick-borne pathogen Borrelia afzelii. Hosts

32

experimentally co-infected with H. polygyrus and larval/nymphal I. ricinus ticks developed substantially

33

stronger systemic Th2 responses, based on GATA-3 and IL-13 expression, than did single-infected mice.

34

During repeated larval infestations, however, anti-tick Th2 reactivity and an observed partial immunity to

35

tick feeding was unaffected by concurrent nematode infections. Importantly, the strong systemic Th2

36

immune response in co-infected mice did not affect the susceptibility to tick-borne B. afzelii. An observed

37

trend for decreased local and systemic Th1 reactivity against B. afzelii in co-infected mice did not result in

38

higher spirochete burden, neither facilitated bacterial dissemination nor induced signs of immunopathology.

39

Hence, this study indicates that strong systemic Th2 responses in nematode/tick co-infected house mice do

40

not affect the success of tick feeding and the control of the causative agent of Lyme disease.

41

2

42 43 44

Introduction

45

Ticks are the most important arthropod vectors of pathogens in temperate regions of the northern

46

hemisphere. In particular, ticks of the genus Ixodes transmit several pathogens affecting the health of

47

humans and life stock, such as Borrelia spp. causing Lyme disease (LD) or relapsing fever and Rickettsia

48

spp. of the spotted fever group causing diseases such as aneruptive fever and tick-borne lymphadenopathy

49

[1]. Small rodents serve as reservoir hosts for many tick-borne pathogens and the larval stage of Ixodes

50

ricinus, the main vector species in Western and Central Europe, frequently feeds on mice and voles [2]. The

51

larval tick stage efficiently acquires the pathogen when feeding on infected rodents [3]. After moulting,

52

nymphal ticks constitute the major source for infections in people [4]. Two genospecies of LD spirochetes

53

account for the majority of infections in Eurasia, Borrelia garinii and Borrelia afzelii, of which the latter

54

mainly and efficiently perpetuates in rodents [5-8]. Mice and voles in urban and rural areas are frequently

55

infected by intestinal helminths, such as the trichostrongyloid Heligmosomoides polygyrus [9-11], raising

56

the question whether such natural co-infections may affect the feeding success of ticks and the transmission

57

of tick-borne pathogens.

58

Ticks initiate type 2 T cell (Th2) responses during their blood meal [12]. Although feeding of I. ricinus ticks

59

results in protective immunity in guinea pigs and rabbits, animals non-native to this tick’s distribution,

60

immune responses are insufficient in protecting most autochthonous wild mouse species and laboratory

61

mouse strains against repeated infestations with ticks [12-16]. On the other hand, feeding Ixodes ticks

62

suppress Th1 responses against tick-borne LD spirochetes in mice [17], and suppression of Th2 cytokines or

63

reconstitution of TNF-α, IL-2 and IFN-γ after tick attachment reduces infection rates in disease-susceptible

64

laboratory mouse strains [18, 19]. Infections with the enteric nematode H. polygyrus elicit strong local and

65

systemic Th2 and regulatory responses [20, 21] and, thereby, affect the Th1-dependent control of

66

concomitant bacterial and protozoan infections [22-28]. It may be that the concurrent presence of enteric

67

nematodes synergistically enhances Th2 and associated innate effector cell responses towards repeated tick 3

68

infestations and as a result affects tick feeding success and transmission of tick-borne pathogens. The

69

expected strong general Th2 skewing of immune responses in mice simultaneously harbouring the two

70

macroparasites, H. polygyrus and I. ricinus, may restrain immune control of tick-borne pathogens and

71

subsequently increase the reservoir competence of rodents.

72

To investigate how frequently mice simultaneously harbour enteric nematodes and ticks in nature, we

73

captured wild rodents in Berlin and determined the frequency of infection with Heligmosomoides spp. and

74

infestation with I. ricinus. Using a defined experimental laboratory system we then assessed parameters of

75

anti-tick immunity and the reservoir competence for LD spirochetes using inbred house mice. To examine

76

whether the expected strong Th2 immune responses associated with intestinal nematode infections interfere

77

with, first, the experimental host’s development of anti-tick immunity measured my means of tick-specific

78

cytokine responses and success of repeated tick feeding, and, second, the control of tick-borne pathogens,

79

we established an experimental model of co-infection in inbred laboratory mice, compared susceptibility to

80

tick feeding and to tick-borne infection with LD spirochetes and evaluated local and systemic immune

81

responses.

82

Here we show that (1) Heligmosomoides spp. and I. ricinus frequently parasitize wild Apodemus spp. mice

83

in Berlin; (2) local anti-I. ricinus immune responses in laboratory house mice are not altered by a concurrent

84

nematode infection, despite the induction of extraordinarily high systemic Th2 responses by nematode/tick

85

co-infections; (3) the nematode infection does not alter the feeding success of pathogen-free tick larvae and

86

spirochete-infected nymphs as well as the development of partial immunity towards repeated tick

87

infestations, and (4) the nematode co-infection does not affect tick-borne transmission, replication and

88

dissemination of B. afzelii spirochetes in house mice.

89

4

90 91

Material and Methods

92

Wild rodent trapping

93

Wild rodents were trapped at two urban (Moabit, Steglitz) and two peri-urban study sites (Gatow, Tegel) in

94

Berlin, Germany in November 2010 and from April to November 2011 (Landesamt für Gesundheit und

95

Soziales, Berlin, Germany, LAGeSo Reg. Nr. G 0210/10). Sites, trapping method and euthanasia were

96

described in Krücken & Blümke et al., 2015 (submitted). After animals were transferred to the laboratory,

97

the fur was screened for ticks under a stereo microscope. Subsequently, a complete necroscopy was

98

performed and the gastro-intestinal tract was thoroughly screened for Heligmosomoides spp., which were

99

stored in 80% ethanol. The remaining carcass was placed in screw top jars over water to allow detachment

100

of remaining ticks. The water and the rodent body were examined during the following week. Ticks were

101

stored in 70% ethanol. Parasite species were determined microscopically according to morphologic literature

102

of nematodes [29] and ticks [30-32].

103 104

Laboratory mice, parasites and Borrelia

105

Six to eight week old specific pathogen-free (SPF) female C57Bl/6JRj, BALB/cJRj and C3H/HeNRj mice

106

(Janvier, France) were used for the experiments. The trichostrongyloid nematode H. polygyrus was

107

maintained by serial passage in C57Bl/6 mice. Specific pathogen-free larval I. ricinus ticks were obtained

108

from egg batches of two adult females after feeding on laboratory beagle dogs (Landesamt für Gesundheit

109

und Soziales, Berlin, Germany, LAGeSo Reg. Nr. H0078/10). Absence of tick-borne pathogens in tick

110

larvae used for experimentation was surveyed by published PCRs [33, 34] for a 153bp fragment of the hbb

111

gene of Borrelia burgdorferi sensu lato species, a 203bp fragment of the gltA gene of spotted-fever

112

Rickettsia spp., a 602-639bp fragment of the 18S rRNA gene of piroplasmida [33], a 257bp fragment of the

113

16S rRNA gene of Anaplasmataceae [34] in isolated DNA [33] from subsets of 20 larvae randomly chosen

114

from each tick batch. Borrelia afzelii-infected I. ricinus nymphs were derived from larvae that had engorged 5

115

on experimentally infected mice (Landesamt für Umwelt, Gesundheit und Verbraucherschutz, Potsdam,

116

Germany, licence number 23-2347-A-24-1-2010).

117 118

Animal experimentation

119

Mice were infected with 200 H. polygyrus L3 larvae via oral gavage. In an initial experiment, mice of three

120

strains (C57Bl/6, BALB/c and C3H) were infested with I. ricinus larvae on day 15 post H. polygyrus

121

infection and dissected 20 days later. To survey the feeding success of larval ticks and anti-larval tick

122

immune responses, C57Bl/6 mice were infected with H. polygyrus and subsequently repeatedly infested

123

with 50 specific pathogen-free I. ricinus larvae on day 4, 18 and 34 p.i. in order to cover the early phase of

124

local Th2 reactivity (approx. day 6 p.i.), maximal Th2 reactivity (around day 21 p.i.) and chronic phase of

125

infection with declining magnitude of the nematode-induced Th2-reaction (approximately day 35 p.i.) by bi-

126

weekly tick infestations. Mice exposed either to nematodes or ticks and naïve mice served as controls. To

127

investigate the effect of H. polygyrus on the control of a tick-borne pathogen, nematode-infected and -free

128

groups were infested with 4-6 B. afzelii-infected I. ricinus nymphs on day 16 post H. polygyrus infection in

129

order to challenge mice with B. afzelii at the peak of nematode-induced Th2-reactivity. Age-matched naïve

130

mice served as controls. During the period of tick infestation, all mice including naïve controls wore a collar

131

to prevent removal of ticks placed in the head region. Mice were housed individually at 80% humidity in

132

filter-topped cages in ventilated cabinets. To optimize recovery of detached ticks, mice were kept on metal

133

grids over water and food was restricted to 3g per day during tick feeding. Mice received water ad libitum.

134

In several experiments, mice were anesthetized with isoflurane once a week for retroorbital blood collection

135

with haematocrit capillary tubes. To assess the feeding success of ticks, I. ricinus larvae or nymphs

136

detaching from hosts were collected from the water and counted twice a day, pooled for every mouse in a

137

screen-capped glass vial and transferred to a desiccator with supersaturated MgSO4 solution. Feeding

138

duration was calculated as mean recovery time of engorged ticks. Seven days after all ticks had detached,

139

the engorgement weight of pooled ticks was determined using a micro scale and the mean weight calculated. 6

140

Moulting rates of fed larvae from the 1st and 2nd infestation or of fed nymphs were determined 3 months

141

after infestation. All animal experiments were approved by and conducted in accordance with guidelines of

142

the appropriate committee (Landesamt für Gesundheit und Soziales, Berlin, Germany) under licence number

143

LAGeSo Reg. Nr. G0282/12.

144 145

Preparation of nematode, tick and spirochete antigens

146

H. polygyrus antigen (Ag) was prepared as described previously [28]. Antigen from feeding larval ticks was

147

acquired from specific pathogen-free I. ricinus larvae 48 h after attachment to C57Bl/6 mice, during

148

intensive saliva production. Ticks were removed from the host, washed in sterile PBS, homogenised and

149

sonicated (3×10 s, 60W) in PBS on ice and centrifuged at 20,000×g for 10 min at 4°C. The supernatant was

150

sterile-filtered through 0.22µm filters (Millipore, USA). For B. afzelii antigen, spirochetes cultured in BSK-

151

H medium (Sigma-Aldrich, Germany) supplemented with 6% rabbit serum were washed three times by

152

centrifugation at 10,000×g at 4°C for 30 min in PBS containing 5mM MgCl2, sonicated on ice (8×15 s) and

153

centrifuged at 10,000×g at 4°C for 30 min. Protein contents of the supernatants were determined by BCA

154

protein assay kit (Pierce, USA). Antigens were stored at -80°C until use.

155 156

Cell culture

157

Splenocytes and cells from gut-draining and head-skin-draining lymph nodes were isolated from euthanized

158

mice by passing the organs through a 70µm cell strainer (BD, USA). Cells were cultured as triplicates of

159

3.5-5×105 cells in 200µl RPMI 1640 (PanBiotech, Germany) containing 10% fetal calf serum, 20mM L-

160

glutamine, 100U/ml penicillin and 100µg/ml streptomycin. Cultures were stimulated with 40µg/ml worm or

161

tick Ag, 30µg/ml B. afzelii Ag (all for 72 h) or with 1µg/ml αCD3 and αCD28 (BD, USA) for 48 h at 37°C

162

and 5% CO2. Supernatants were stored at -20°C for cytokine detection.

163 7

164

Immunohistology

165

Formalin fixed tissue samples were dehydrated and embedded in paraffin. Tibiotarsal joints were decalcified

166

prior to paraffin embedding. 1-2µm paraffin sections were de-waxed and hydrated and histochemically

167

stained for overview with hematoxylin and eosin (Merck), for mast cells using toluidine blue and for

168

eosinophil granulocytes employing a modified sirius red staining protocol [35]. Sections were stained with

169

toluidine blue (Sigma) for 10 min, washed with distilled water and dehydrated by dipping in 1% acetic acid

170

and 100% ethanol. Sections were cleared in xylol and coversliped with corbit balsam (Hecht). For staining

171

of eosinophils, sections were stained for 1 min with hematoxylin (Merck), washed with tap water and

172

dehydrated with 100% ethanol before staining with direct red 80 (Sigma) for 2 h. Sections were washed with

173

tap water, dehydrated with 100% ethanol and cleared in xylol before coverslipping with corbit balsam.

174

Images were acquired using a fluorescence microscope (AxioImager Z1) equipped with a CCD camera

175

(AxioCam MRm) and processed with Axiovision software (Carl Zeiss AG, Germany).

176 177

ELISA

178

Culture supernatants were screened for IFN-γ, IL-13 (Ready-Set-Go! ELISA, eBioscience, USA) and IL-10

179

(BD, USA) according to the manufacturer’s instructions.

180 181

Flow cytometry

182

Blood was sampled in PBS with 0.2% BSA and 5mM EDTA and cells were stained with αCD4-FITC

183

(RM4-5, eBioscience, USA) and fixable viability dye-eFluor780 (eBioscience, USA) for dead cell

184

exclusion. Red blood cells were lysed in FACS Lysing Solution (BD, USA) before leucocytes were stained

185

intracellularly with αFoxp3-eFluor450 (FJK-16s, eBioscience, USA), αGATA-3-eFluor660 (TWAJ,

186

eBioscience, USA) and αT-bet-PE (eBio4B10, eBioscience, USA) using a fixation/permeabilization buffer

187

kit (eBioscience, USA). Cells from spleen and lymph nodes were labelled with αCD4-PerCP-eFluor710 8

188

(GK1.5, eBioscience, USA) and fixable viability dye-eFluor780 (eBioscience, USA) in PBS with 0.2%

189

BSA. Transcription factors were stained according to white blood cells. For intracellular detection of

190

cytokines, cells were incubated with 1µg/ml PMA and ionomycin for 30 min and 5µg/ml brefeldin A

191

(Sigma, Germany) for another 2.5 h. Subsequently, they were stained with αIFNγ-eFluor450 (XMG1.2,

192

eBioscience, USA), αIL-13-AlexaFluor647 (eBio13A, eBioscience, USA), αIL-10-PE (JES5-16E3,

193

eBioscience, USA) and αIL-17-AlexaFluor488 (TC11-18H10, BD, USA). Cells were acquired using LSRII

194

and Canto II flow cytometers (BD, USA) and analysed using FloJo 8.8.7 software (TreeStar, USA).

195 196

Spirochete quantification

197

For quantification of spirochetes in mouse organs in relation to tissue weight, a touchdown-qPCR assay was

198

developed for absolute quantification with λ phage genome copies as an external standard. For DNA

199

isolation, 80 ±1mg of head skin and 70 ±1mg of heart were sampled with sterile scalpel blades, the skinned

200

left tibiotarsal joint (range 32.6-39.1mg) was obtained using clean scissors. Prior to DNA isolation with the

201

FastDNA Spin Kit with Lysing Matrix A (MPBiomedicals, France), 104 copies of 48502bp λ genome

202

(Thermo Fisher Scientific, USA) per mg head skin or heart and 105 copies per tibiotarsal joint were added as

203

an external standard for tissue weight. To calculate copy numbers, the DNA concentration (0.3µg/µl

204

measured spectrophotometrically by the company) and the molecular weight of the genome were used.

205

Samples were homogenised with the FastPrep 24 (MP Biomedicals, France) for 4×30 s with 6.0m/s and

206

cooled on ice between homogenisation steps. Variations of the DNA amount in the samples used in qPCR

207

have a major impact on the accuracy of the absolute quantification, since qPCR efficiency is decreased with

208

higher DNA concentrations. As common spectrophotometrical DNA quantification is strongly impacted by

209

contaminations with salts and proteins, a more precise fluoroscopical DNA quantification assay was

210

developed using the LCGreen Plus 10× dye (BioChem, USA), exclusively binding on dsDNA and the

211

LightCycler480 II System (Roche Molecular Diagnostics, USA) for fluorescence measurement. Accurate

212

DNA quantification in a range of 1 to 100ng was obtained. On 96-well plates, DNA samples, standards (20, 9

213

40, 60, 80 and 100ng λ phage DNA) and water controls were prepared in duplicates in 10µl assay volume.

214

After vigorous vortexing of sealed plates, samples were centrifuged and fluorescence was measured by the

215

LightCycler480 II after 2 min incubation at 25°C during short heating from 25 to 26°C with 100 acquisitions

216

per °C. For extrapolation of dsDNA-quantity in samples, a calibration curve was calculated from

217

fluorescence values of the standards using a 4-parameter logistic regression model. Two real-time PCR

218

assays targeting a 153bp fragment of the hbb gene of LD spirochetes and a 158 bp fragment of λ phage

219

genome were conducted with the LightCycler480 II using LightCycler 480 SYBR Green I Master (Roche

220

Molecular Diagnostics, USA), 0.5µM of each primer and exactly 100ng sample DNA in a 20µl volume.

221

Primers used for hbb fragment are published elsewhere [36], forward and reverse primer used for λ phage

222

genome were 5'-TTTTGATTTTGCTGTTTCAA-3' and 5'-ACCTTTTCCATGAATTGGTA-3'. All PCR

223

reactions were performed as duplicates and included a negative control and standards, each with 100ng

224

target-free mouse DNA of the respective organ. Standards consisted of a dilution series of 104-101 and 3

225

copies of an hbb plasmid standard or λ phage DNA. The thermocycling conditions consisted of an initial

226

denaturation step at 95°C for 5 min followed by 50 cycles of 95°C for 5 s, annealing for 30 s (initial

227

temperature 62°C, then decreasing by 0.25°C/cycle until 57°C) and 72°C for 15 s. Touchdown amplification

228

was performed to increase binding specificity of primers during the first cycles. After amplification, a high

229

resolution melting step consisting of 95°C for 5 s, 40°C for 1 min and progressive heating from 65 to 95°C

230

with 20 acquisitions per °C was performed to identify target fragments by melting temperature in

231

combination with their fragment size in subsequent agarose gel electrophoresis. Borrelia hbb copies

232

extrapolated from the standard curve were multiplied with the ratio of extrapolated λ phage DNA copies in

233

the sample and number of copies previously added per mg skin/heart or per tibiotarsal joint, respectively.

234

The detection limit was less than 3 copies and the quantification limit 5-10 copies per 100ng DNA for both

235

touchdown-qPCR assays.

236 237

Statistics 10

238

Confidence intervals (CI) of prevalences were calculated as Wilson score intervals using R Statistics and the

239

“PlotCIs” package. Negative binomial regression of tick numbers was performed using R Statistics Software

240

and “MASS” package to estimate the influence of the variable Heligmosomoides spp. count and categorical

241

variables mouse species (A. flavicollis and A. sylvaticus) and trapping location (Gatow, Moabit, Steglitz and

242

Tegel) on the number of I. ricinus ticks on wild mice.

243

GraphPad Prism 5.01 (San Diego, USA) was used for plots and statistical analysis of experimental infection

244

data. Statistical differences were tested using unpaired t-test, one-way ANOVA with Bonferroni-corrected

245

post-test (Fig. 3 and 5) and two-way-ANOVA (Fig. 2) under the assumption of normal distribution of FACS

246

fluorescence data, ELISA optic density data and the majority of the experimental parasite infection data. The

247

post-tests following two-way-ANOVA for individual pairs using the Bonferroni correction were performed

248

with the “Post test calculator” at http://graphpad.com/quickcalcs. The number of engorged tick nymphs and

249

moulting rate in Fig. 7A, D and cell counts in Fig. S1 B were not normally distributed and differences were

250

tested using the Mann-Whitney-U-Test. Levels of significance below p=0.05 were considered to be

251

significant.

252

11

253 254

Results

255

Nematode/tick co-infection is frequent in urban and peri-urban wild mice

256

A total of 107 potential murine hosts of Heligmosomoides spp., representing 82 Yellow-necked mice

257

(Apodemus flavicollis) and 25 long-tailed wood mice (A. sylvaticus), were live-trapped at 2 urban (Moabit,

258

Steglitz) and 2 peri-urban (Gatow, Tegel) sites in Berlin and examined for their parasite burden. No house

259

mice (Mus musculus) were caught as traps were placed exclusively outside buildings. Both species

260

frequently hosted Heligmosomoides spp. (47.7%, 95% CI 38.5-57.0, N=107) with intensities of 1-246

261

specimens (Table 1). In addition, 55.1% (95% CI 45.7-64.2) were infested by up to 59 subadult I. ricinus

262

ticks (92.1% larvae, 7.9% nymphs). A high percentage (41.3%, 95% CI 29.1-53.4) of tick-infested mice

263

(N=59) were simultaneously infected by Heligmosomoides spp. A negative binomial regression model

264

including all 107 mice showed an important influence of the trapping location and/or the rodent species on

265

the number of I. ricinus ticks on the rodents but could not distinguish between both variables because of

266

total collinearity (Fig. 1). A. sylvaticus mice were exclusively trapped in Moabit whereas A. flavicollis only

267

occurred at the three other locations. A. flavicollis from Gatow were infested with significantly more ticks

268

than those from Steglitz (p