1 uninfected mosquito bites confer protection against...

33
1 Uninfected Mosquito Bites Confer Protection Against Infection 1 with Malaria Parasites. 2 3 RUNNING TITLE : Mosquito Bites Confer Protection Against Plasmodium 4 5 Michael J. Donovan 1 , Andrew S. Messmore 2 , Deborah A. Scrafford 1 , David 6 L. Sacks 2 , Shaden Kamhawi 2 , and Mary Ann McDowell 1* 7 8 1 Center for Global Health and Infectious Diseases, Department of Biological Sciences, 9 University of Notre Dame, Notre Dame, IN 46556 10 11 2 Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, 12 National Institutes of Health, Bethesda, Maryland 20892 13 14 15 16 17 18 19 20 * Corresponding author. Mailing address: 215 Galvin Life Sciences, Department 21 of Biological Science, University of Notre Dame, Notre Dame, IN 46656. Phone: 22 (574) 631-9771. Fax: (574) 631-7413. E-mail: [email protected] 23 ACCEPTED Copyright © 2007, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved. Infect. Immun. doi:10.1128/IAI.01928-06 IAI Accepts, published online ahead of print on 5 March 2007 on September 12, 2018 by guest http://iai.asm.org/ Downloaded from

Upload: phungkhuong

Post on 13-Sep-2018

212 views

Category:

Documents


0 download

TRANSCRIPT

1

Uninfected Mosquito Bites Confer Protection Against Infection 1

with Malaria Parasites. 2

3

RUNNING TITLE: Mosquito Bites Confer Protection Against Plasmodium 4

5

Michael J. Donovan1, Andrew S. Messmore2, Deborah A. Scrafford1, David 6

L. Sacks2, Shaden Kamhawi2, and Mary Ann McDowell1* 7

8

1Center for Global Health and Infectious Diseases, Department of Biological Sciences, 9

University of Notre Dame, Notre Dame, IN 46556 10

11

2Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, 12

National Institutes of Health, Bethesda, Maryland 20892 13

14

15

16

17

18

19

20

* Corresponding author. Mailing address: 215 Galvin Life Sciences, Department 21

of Biological Science, University of Notre Dame, Notre Dame, IN 46656. Phone: 22

(574) 631-9771. Fax: (574) 631-7413. E-mail: [email protected] 23

ACCEPTED

Copyright © 2007, American Society for Microbiology and/or the Listed Authors/Institutions. All Rights Reserved.Infect. Immun. doi:10.1128/IAI.01928-06 IAI Accepts, published online ahead of print on 5 March 2007

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

2

Abstract 24

Despite decades of research and multiple initiatives, malaria continues to be one 25

of the world’s most debilitating infectious diseases. New insights for malaria 26

control and vaccine development will be essential to thwart the staggering 27

worldwide impact of this disease (7); ultimately successful vaccine strategies will 28

undoubtedly be multi-factorial, incorporating multiple antigens and targeting 29

diverse aspects of the parasite’s biology (23). Using a murine model of malaria 30

infection we show here that exposure to uninfected mosquito bites prior to 31

Plasmodium yoelii infection influences the local and systemic immune response 32

and limits parasite development within the host. In hosts pre-exposed to 33

uninfected mosquito bites reduced parasite burdens were detected early in the 34

liver and remained lower during the blood stage of the life-cycle as compared to 35

hosts that only received mosquito bites at the time of infection. Repeated 36

exposure to uninfected mosquito bites skewed the immune response towards a 37

T-helper 1 (Th1) phenotype as indicated by increased levels of interleukin-12 (IL-38

12), interferon-gamma (IFN-g) and inducible nitric oxide synthase (iNOS). These 39

data suggest that the addition of mosquito salivary components to anti-malaria 40

vaccines may be a viable strategy for creating a Th1 biased environment known 41

to be effective against malaria infection. Furthermore, this strategy may be 42

important for the development of vaccines to combat other mosquito-transmitted 43

pathogens. 44

45

46

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

3

Introduction 47

Malaria continues to be a major public health threat and has an enormous 48

economic impact, resulting in nearly 3 million deaths annually and ranking eighth 49

as a contributor to loss of global disability adjusted life years (7). Emerging drug 50

resistance in the Plasmodium parasites that cause malaria and insecticide 51

resistance in mosquito vectors that transmit these pathogens emphasizes the 52

urgent need for developing an effective malaria vaccine to control the devastating 53

burden of malarial disease. The complex biology of malaria parasites coupled 54

with antigenic polymorphism, poor antigen immunogenecity, and parasite-55

induced immuno-suppression distinguishes the quest for a malaria vaccine as 56

extraordinarily daunting. 57

58

Malaria is transmitted to humans via the bite of its insect vector, a female 59

anopheline mosquito. During blood feeding mosquitoes inject infective 60

Plasmodium sporozoites into the avascular skin tissue of its host where they 61

eventually migrate into the circulation (53); simultaneously, a plethora of 62

pharmacologically active compounds in mosquito saliva are introduced into the 63

host. These compounds have substantial anti-haemostatic, anti-inflammatory, 64

and immunosuppressive activities that aid the mosquito in the blood feeding 65

process (40). Furthermore, many of the salivary components are immunogenic 66

and elicit strong immune responses, evidenced by the swelling and itching that 67

accompanies a mosquito bite (38). This substantial effect of immune activation 68

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

4

by saliva creates an inflammatory context for further responses to co-injected 69

pathogens. 70

71

A role for arthropod saliva in modifying the outcome of infection is not novel to 72

mosquitoes and malaria parasites; increased pathogen infectivity has been 73

described for ticks, sand flies and mosquitoes [for review see (50)]. While 74

studies primarily have focused on the enhancement of transmission and disease 75

when pathogens are introduced in the presence of vector saliva, some studies 76

have explored the effect of repeated exposure of vector saliva on the outcome of 77

infection. Although the mechanism has yet to be completely elucidated, repeated 78

infestation with pathogen-free Ixodes scapularis ticks induces resistance to 79

Borrelia burgdorferi transmission (56). The most striking host-parasite-vector 80

system that has been studied is Leishmania infection by the bites of 81

phlebotomine sand flies. Interestingly, multiple exposures to uninfected sand fly 82

bites prior to infection confers resistance to L. major, due to increases in the 83

cytokines responsible for cell-mediated immunity (24). Mosquito bites also have 84

been shown to influence immunity and potentiate viral disease in mouse models 85

(18, 26, 45, 46), possibly through modulation of host systemic cytokine 86

responses (46, 58). 87

88

Mosquito bites induce immediate, delayed and systemic hypersensitivity 89

reactions in hosts (38); consequently, we hypothesized that the local tissue and 90

systemic environment when ‘immunized’ by mosquito salivary components can 91

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

5

enhance malaria immunity. We tested this hypothesis using the prototypic 92

murine model of malaria infection: sporozoite infection in mice via P. yoelii 93

infected Anopheles stephensi mosquitoes. 94

95

Materials and Methods 96

Mice Balb/c mice aged 6-8 weeks were bred at the Friemann Life Sciences 97

Center at the University of Notre Dame under approved Institutional Animal Care 98

and Use Committee (IACUC) protocols. All mice were female and age-matched 99

for all experiments. Interferon gamma deficient (IFN-g KO) and Balb/c wild type 100

(WT) counterparts were purchased (Jackson Labs, Bar Harbor, ME) and used at 101

6-8 weeks of age. 102

103

Pre-sensitization Mice were anesthetized, and their ears were exposed for 20 104

min. to a screened vial containing 15-20 fully-matured female adults every two 105

weeks for six weeks (pre-sensitized). A control group of age-matched mice were 106

only anesthetized at each time point (naïve). Mice were challenged 2 weeks 107

following the last pre-exposure. Twenty-four (cytokine responses) or 40 108

(parasite quantification) hours after the final exposure, ears, liver, and spleen 109

samples were harvested and stored in RNAlater (Qiagen, Valencia, CA). 110

111

Mosquito Infections P. yoelii (17XNL) parasites were maintained by alternating 112

passage of parasites through A. stephensi and Balb/c mice. Murine parasitemia 113

was observed through thin-layered blood smears. Smears were fixed in 100% 114

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

6

methanol and stained with Geimsa. Once gametocytes were present, infected 115

animals were anesthetized and A. stephensi were allowed to feed. Four to eight 116

days after infection, A. stephensi were anesthetized and midguts were dissected 117

out, and stained with mercurochrome (Fisher Scientific, Chicago, IL), and the 118

number of oocysts per midgut were counted. Average infection rates were 119

between 75 and 100 percent. After the parasite matured to the salivary gland-120

sporozoite stage (14 days post-infection), appropriate groups were exposed to P. 121

yoelii-infected A. stephensi. Mice were exposed to 10 infected mosquitoes for 15 122

minutes on the right ear, the same vial of mosquitoes was transferred to the left 123

ear for an additional 15 minutes. Feeding success was assessed by visual 124

observation, looking for blood in the midgut; no obvious differences between 125

feeding on naïve versus pre-sensitized mice were detected. Sporozoites were 126

isolated as previously described (17). 127

128

Blood Stage Quantification Mice were pre-sensitized as described above. 129

Two weeks following 3 exposures to uninfected mosquitoes, mice were exposed 130

to bites of P. yoelii-infected A. stephensi. 24 hours post-exposure, blood 131

samples were taken, thin-layer blood smears were made, and were stained with 132

Geimsa. Subsequently, samples were taken and quantified each day until 133

mouse euthanasia on day 7 post-infection. For determination of parasitemia, 134

1000 cells were counted from each sample. 135

136

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

7

RNA Isolation and Generation of cDNA RNA was isolated using an RNeasy 137

Mini kit (Qiagen, Valencia, CA) for ears and Trizol Reagent (Invitrogen, Carlsbad, 138

CA) for spleens and livers. The entire harvested organ was homogenized in the 139

respective lysis buffers. Contaminating DNA was removed from 1µg of RNA via 140

DNase I treatment (Invitrogen, Carlsbad, CA), using 1 Unit of DNase I, and a final 141

concentration of DNAse buffer containing 20mM Tris-HCl (ph 8.4), 2mM MgCl, 142

and 50mM KCl. DNA-free RNA was used to generate cDNA with oligo dT (for 143

cytokine analysis) (Invitrogen, Carlsbad, CA) or Random Primers (for infection 144

studies) (Invitrogen, Carlsbad, CA) at a final concentration of 10ng and 15ng, 145

respectively, in addition to 500nM dNTPs (Invitrogen, Carlsbad, CA), 200 units of 146

Superscript III Reverse Transcriptase (Invitrogen, Carlsbad, CA), 40 units RNase 147

Out (Invitrogen, Carlsbad, CA), and 5mM DTT. 148

149

Quantitative PCR and Analysis cDNA was used for quantitative real time PCR 150

analysis using the 2x SYBR Green Kit (Applied Biosystems, Foster City, CA). 151

Reactions were run on the ABI 7700 Sequence Detector machine. The SYBR 152

Green kit was used at a 1x proportion, along with 300 nM of forward and reverse 153

primers for each reaction. Primers used (IDT Coralville, IA) were HPRT 5’-GTT 154

GGA AGG CCA GAC TTT GTT-3’ and 5’-GAT TCA ACT TGC GCT CAT CTT 155

AGG C-3’; IFN-g 5’-AGA GCC AGA TTA TCT CTT TCT ACC TCA-3’ and 5’-CCT 156

TTT TCG CCT TGC TGT TG-3’; IL-4 5’-ACG AGG TCA CAG GAG AAG GA-3’ 157

and 5’-AGC CCT ACA GAC GAG CTC ACT C-3; IL-12p40 5’ AACCAT CTC CTG 158

GTT TGC CA-3’and 5’-CGG GAG TCC AGT CCA CCT C-3’;and iNOS primers 159

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

8

as previously described (39). Cytokine primers were designed to overlap introns 160

to help assure no DNA amplification occurred. In order to be assured of proper 161

amplification, a melting curve analysis was performed on each product. We 162

employed the comparative threshold cycle method to determine relative 163

differences in parasite burdens. After generation of Ct values (the cycle number 164

at which the reaction crosses the threshold), relative copy number was 165

determined according to the following equation: number of copies = 2-∆∆ct, 166

where ∆∆ct = ∆ct(sample) – ∆ct(calibrator), ∆ct = ∆ct(sample) – ∆ct(HPRT), ct = 167

cycle at which a statistically significant increase in the emission intensity over the 168

background, and ∆ct(calibrator) = the mean ∆ct for the naïve control. Parasite 169

liver loads were determined using primers for P. yoelii 18s rRNA 40 hours after 170

infection, as previously described (9). The amount of RNA of the different 171

samples was normalized based on the measurement of the mRNA levels from 172

the mouse housekeeping gene HPRT as described above. Cytokine graphs are 173

expressed as the mean value of induction levels that have been normalized to 174

the mean values from naïve tissues. Infection levels were expressed in 40-∆∆ct 175

as previously described (51). Significance levels were determined using a 176

Student’s T Test with a confidence level of 95%. 177

178

ELISA analysis Serum was collected from each mouse at the time of organ 179

harvest and IFN-g was quantified by ELISA (Pierce Biotechnology, Rockford, IL) 180

according to the manufacturer’s instruction. 181

182

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

9

Results and Discussion 183

In some areas a single individual can receive nearly 200 mosquito bites/day (52) 184

or over 10,000 bites/year (22). Malaria infection rates in these vectors range 185

from below 0.1% to 10% (22, 28), indicating that even in the highest areas of 186

transmission a single individual is exposed to drastically more mosquito saliva 187

than malaria parasites. To assess the impact of previous exposure to uninfected 188

mosquito bites on Plasmodium development, we compared P. yoelii burdens in 189

mice pre-exposed to uninfected A. stephensi bites (pre-sensitized) to those of 190

unexposed (naïve) mice. We utilized a system where infected and uninfected 191

mosquito bites were limited to the ears so that local and systemic responses 192

could be separated easily. Forty hours post-infection, after parasite 193

differentiation and amplification but prior to parasite release into the circulation, 194

pre-sensitized mice exhibited significantly reduced liver burdens of P. yoelii 195

compared to naïve mice (Fig. 1a), corresponding to a 9-fold reduction in parasite 196

numbers following the natural infection (Fig. 1b). These reduced parasite 197

burdens also were evident when blood parasitemias were assessed (Fig. 1c). 198

While the pre-patent period (i.e. the time it takes to visually detect blood 199

parasites) was not effected by pre-sensitization, the parasitemia levels were 200

reproducibly lower in mice pre-exposed to uninfected mosquito bites. 201

202

The hepatic stage is the most vulnerable of the Plasmodium life-cycle for 203

intervention and substantial research on liver stage immunity, primarily using 204

murine models, exists. The predominant effector mechanism mediating this pre-205

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

10

erythrocytic immunity is the production of IFN-g that inhibits parasite 206

development within hepatocytes via nitric oxide (16). To investigate the effect 207

that repeated exposure of mosquito saliva has on cytokine profiles, both local 208

tissue and systemic IFN-g and interleukin-4 (IL-4) mRNA levels were assessed. 209

Local IFN-g expression was relatively low in naïve animals and mice that were 210

only exposed a single time (naïve bit), while pre-sensitized animals readily 211

produced IFN-g in response to A. stephensi bites (Fig. 2a). Furthermore, pre-212

sensitized mice express reduced levels of IL-4 as compared to naive animals 213

(Fig. 2b). The pre-sensitized mice are Th1 biased, illustrated by cytokine ratio 214

(IFN-g:IL-4) with much more IFN-g being expressed than IL-4 (Fig. 2c). 215

Therefore, A. stephensi saliva significantly changes the local cytokine 216

environment at the tissue site where parasites are introduced; Th1 bias following 217

mosquito pre-sensitization also occurs systemically in the liver (Fig. 2d-f), spleen 218

(Fig. 2g-i), and serum (data not shown), indicating a systemic cytokine shift to a 219

Th1 profile. These results suggest that up-regulation of IFN-g is part of the 220

protective phenotype against P. yoelii infection associated with mosquito saliva 221

pre-sensitization. 222

223

To further evaluate the role of IFN-g, WT and IFN-g KO BALB/c mice were pre-224

sensitized to uninfected mosquito bites, or left naïve, prior to natural infection 225

with P. yoelii. Pre-sensitization-associated protection against P. yoelii infection 226

was abrogated in the absence of IFN-g (Fig. 3a), indicating that IFN-g is essential 227

for the protective response. Because nitric oxide is required for immunity to P. 228

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

11

yoelii liver infection (5), we measured inducible nitric oxide synthase (iNOS) 229

mRNA levels in the livers of naïve and pre-sensitized WT and IFN-g KO mice 230

(Fig. 3b). The 10-fold up-regulation of iNOS levels in response to A. stephensi 231

pre-exposure was not observed in IFN-g KO mice, suggesting that the IFN-g 232

induced following pre-sensitization to mosquito bites leads to NO-induced killing 233

of malarial parasites. Interestingly iNOS mRNA was also increased by pre-234

sensitization in the local ear environment as early as 5 hours post infection (Fig. 235

4). 236

237

As interleukin-12 (IL-12) is necessary for immunity against P. yoelii (17) and has 238

been proposed as a potential adjuvant for anti-malaria vaccines (44, 47), we 239

evaluated IL-12p40 mRNA levels in the livers (Fig. 5a) and spleens (Fig. 5b) of 240

pre-sensitized animals. We detected significantly higher levels of IL-12p40 241

mRNA in the organs of pre-sensitized animals as compared to naïve mice. 242

243

We show that the immunity induced following A. stephensi pre-sensitization 244

involves local and systemic up-regulation of IFN-g and iNOS (Figs. 2-4, data not 245

shown). These results raise the question of what tissue is primarily affected by 246

this protective mechanism and the timing of this response. The majority of 247

infectious malaria sporozoites released during mosquito blood feeding do not 248

immediately enter the circulation, instead sporozoites are deposited into the skin 249

where they eventually move into dermal vessels (53). Although the timing of 250

vascular entry remains a matter of debate, it is clear that some sporozoites 251

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

12

remain in the skin and may take substantially longer to enter the blood stream 252

(53). This delayed timing coupled with the suggestion that sporozoites are 253

directly susceptible to NO-mediated killing (29) invokes a model of reduction in P. 254

yoelii burdens in the liver due to lower numbers of parasites entering the 255

circulation. To determine the chronology and location of parasite killing in pre-256

sensitized mice, we evaluated the relative parasite burdens in both the ear and 257

the liver at various time points (5-30 hours) following natural infection in naïve 258

and pre-sensitized mice (Fig. 6a & b). By 10 hours post infection, parasite 259

quantification in the ear returned to background levels in both groups of mice, 260

indicating that the same numbers of sporozoites leave the bite site (Fig. 6a) and 261

enter the liver (Fig. 6b) in naïve and pre-sensitized mice. The difference in liver 262

parasite burdens is not evident until 20 hours post-infection. As increases in 263

parasite burden during the first 5 – 40 hours of infection is primarily due to 264

parasite multiplication our data suggests that the protective mechanism due to 265

pre-sensitization is operating in the liver. To conclusively address whether liver 266

protection could be occurring, we exposed naïve and pre-sensitized mice to 267

uninfected mosquito bites and immediately challenged intravenously in the tail 268

vein, rather than exposing mice to infected mosquitoes. Forty hours post-i.v. 269

challenge, P. yoelii liver burdens remained significantly lower in pre-sensitized 270

mice, demonstrating that the protection is due to, in part, the response in the liver 271

(Fig. 6c). 272

273

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

13

The substantial effort towards the production of an efficacious malaria vaccine 274

has yet to yield a suitable product. Following the initial report that vaccination 275

with radiation-attenuated sporozoites can protect against malaria challenge (36), 276

extensive research has elucidated the immunological parameters that confer 277

such protection. The protective mechanisms identified for malaria sporozoite and 278

liver stages are remarkably similar in rodent models and human volunteers (30). 279

It is possible that residual salivary components remaining from the isolation of 280

sporozoites can partially explain the protection observed in the studies involving 281

radiation-attenuated sporozoites. One original study investigating irradiated 282

sporozoites as a vaccine used repeated vaccination with large amounts of 283

mosquito salivary gland homogenate (70 glands) as a control and demonstrated 284

that this procedure conferred partial protection to P. berghei infection in mice (1, 285

2). More recently it has been demonstrated that P. gallinaceum parasitemias are 286

increased in the presence of Aedes fluviatilis saliva in a chicken malaria model; a 287

response that was reversed with prior exposure to mosquito saliva (43). 288

289

It is well established that naïve travelers and children are at increased risk for 290

severe malarial disease as compared to adult endemic populations and that 291

these adults do not exhibit sterile immunity; rather, this degree of natural 292

immunity results in asymptomatic infections with lower parasite burdens in the 293

circulation (42). Historically this phenomenon has been attributed to the gradual 294

onset of immunity due to repeated parasite infections (48). We propose that the 295

extensive exposure to mosquito saliva that accrues over time in malaria endemic 296

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

14

regions results in a Th1 climate that influences pathogen establishment. 297

Coupled with specific immunity to malaria parasites this inhospitable environment 298

may contribute to lower malarial burdens in endemic adults. Here we utilize a 299

model whereby we infect with 10 mosquitoes with infection rates ranging from 300

75-90% with oocyst counts of 24-39 per mosquito to ensure adequate parasite 301

numbers for detection. The majority of mosquitoes in the field harbor only one 302

oocyst (12) with infection rates never greater than 10% (22, 28). We did not 303

observe sterile immunity in our model system; however, it is possible that the 304

cytokine shift that we detected may be adequate to control the small numbers of 305

parasites an individual encounters in a natural setting. 306

307

The IFN-g up-regulation due to pre-sensitization that we detected diminishes 308

over time as is evident by the lower levels of IFN-g mRNA that are present in 309

ears and liver in pre-sensitized mice that did not receive one final bite prior to 310

analysis. This observation is consistent with the typical rapid loss of anti-malarial 311

immunity when an individual leaves an endemic region (48). We suggest that 312

this loss of immunity can partially be explained by an absence, or drastically 313

lower level, of mosquito boosting. In our study mosquito exposure ceased after 314

the infection was initiated. It is intriguing to postulate that increased resistance 315

may be observed if hosts continued to be exposed to mosquito bites throughout 316

the infection as would be the case in an endemic region. IFN-g production is 317

associated with primary immune responses to blood stage Plasmodium 318

infections in mice (13, 34) and humans (31) and appears to be crucial for the 319

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

15

development of protective immunity (15, 49). Although in this study we did not 320

explore blood stage immunity, the increased levels of IFN-g in the serum induced 321

by repeated mosquito exposures suggest that blood stage immunity may be 322

influenced and continued mosquito boosting may increase this response. 323

324

The systemic shift in cytokine balance that we detected is not unique as bite-325

induced systemic responses have been documented with Rhipicephalus 326

sanguineus (tick) (19), Culex pipiens and Aedes aegypti (58). The majority of 327

studies assessing arthropod modulation of host immune responses have focused 328

on one-time exposures to bites or salivary gland components [for review see (6, 329

8, 50)]. Combined these studies engender a model where initial exposure to 330

arthropod saliva induces a Th2 immune response, potentiating infectivity of a 331

variety of vector-borne pathogens. For mosquito transmitted pathogens, 332

infectivity of P. berghei (54), Cache Valley virus (18), La Crosse virus (37), and 333

vesicular stomatitis virus (VSV) (26, 27) is enhanced in the presence of saliva. 334

For VSV infection, increased viral loads are associated with a SGH dependent 335

decrease of type I interferons in vitro (26). Feeding of both Culex pipiens and 336

Aedes aegypti mosquitoes on mice induces increased levels of systemic IL-4 and 337

IL-10 with a concomitant decrease in IFN-g production (58). Similarly, 338

inoculation of Ae. Aegypti SGH with Sindbis virus results in higher levels of Th2 339

cytokines and reduced expression of interferons (46). 340

341

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

16

While we detected increased Th2 responses in naïve mice versus pre-sensitized 342

hosts, we did not observe an increase in IL-4 upon one exposure to A. stephensi. 343

Our results are in agreement with previous studies investigating skin and 344

lymphnode cytokine production in response to A. stephensi bites (14). In 345

contrast to our observations, studies exploring splenic cytokine production in 346

response to exposure to Ae. Aegypti or C. pipiens mosquitoes detected an 347

increase in Th2 cytokine production in C3H/HeJ mice; however, in a congenic 348

host strain (C3H/RV) Th1 cytokines predominated (58). In vitro studies suggest 349

that mosquito species may differ in their ability to modulate host immune 350

responses (55); the discrepancies between our results and previous work 351

investigating in vivo immune modulation by mosquito exposure may be attributed 352

to variation in the experimental approaches or may reflect actual differences 353

between mosquito species. 354

355

Data exploring the effect of multiple, repeated exposures to arthropod saliva are 356

scant. While single exposures to sand fly bites (24) or SGH (4, 32, 35) are 357

associated with increased levels of Th2 cytokines, repeated exposure leads to a 358

switch to Th1 immunity (4, 24). This switch to primarily IFN-g production at the 359

bite site induces resistance to L. major infection. In contrast, repeated 360

exposures to Ae. aegypti bites results in elevated production of antigen-specific 361

IL-4 production in cultured spleen cells, although this response is not detected in 362

response to ConA stimulation (10). Multiple infestations of ticks are generally 363

thought to lead to heightened levels of Th2 cytokines. As the literature conflicts 364

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

17

as to whether tick infestation blocks B. borgdorferi transmission (40, 55) and 365

what type of cytokine response is favorable or detrimental for spirochete 366

transmission (24, 56), the role that the IL-4 induced by repeated tick infestation 367

plays in resistance to Lyme disease is unclear. In contrast to sand flies and 368

mosquitoes, ticks take several days to complete feeding. The modulation of host 369

immunity towards Th2 cytokine expression may provide an evolutionary 370

advantage to ticks to avoid host sensitization to tick feeding; rapid feeding 371

arthropods may not require such immunomodulation. Interestingly, animal 372

species that have acquired immunity to tick feeding express cutaneous basophil 373

hypersensitivity reactions (CBH) at attachment sites (3), a reaction mediated by 374

Th1 responses (20). Furthermore, even though the immune response of mice to 375

repeated tick infestations is predominated by IL-4, IFN-g (21) and IgG2a (11) 376

levels increase with multiple exposures. In conjunction with our observations, a 377

model is beginning to emerge indicating that repeated exposure to rapid feeding 378

arthropods induces Th1 profiles that lead to increased resistance to pathogen 379

transmission. 380

381

It previously has been demonstrated that treatment of BALB/c mice with 382

recombinant IL-12 prior to sporozoite challenge protects against P. yoelii 383

infection (47), suggesting that short-term prophylaxis with rIL-12 could be used to 384

combat malaria. The increased level of IL-12p40 mRNA detected in pre-385

sensitized animals suggests that it is possible that ‘vaccination’ with uninfected 386

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

18

mosquitoes may be an efficient method to induce IL-12 and avoid the toxicity 387

associated with treatment with recombinant proteins. 388

389

It is thought that individuals living in mosquito intense areas naturally become 390

desensitized to mosquito bites. The particular mechanism of this tolerance 391

remains to be defined, but is known to be associated with a loss of the wheal and 392

flare reactions of type I hypersensitivity as well as delayed reactions (38). 393

Although cytokine responses have not been evaluated in desensitized 394

individuals, the hypersensitivity reactions in response to mosquito saliva are Th2 395

mediated (i.e. IgE-mediated for Type I and eosinophil mediated for Type IV). 396

Therefore it is possible that desensitization may cause a reduction in Th2 397

cytokines, thus promoting an even greater Th1 environment than we detected in 398

our model system. 399

400

New vaccine targets and novel strategies will be essential for the ultimate 401

success of malaria vaccine development and data suggests that any measure 402

that limits parasite densities in the liver will reduce the morbidity and mortality 403

associated with malaria infection (33). Our findings imply that mosquito salivary 404

constituents could be effective components in such a vaccine. In this context, 405

saliva can be thought of as a non-specific potentiator; as long as vaccinated 406

individuals encounter malaria together with mosquito saliva the potentiator will be 407

effective at inducing a Th1 biased environment that is known to be effective 408

against malaria infection. 409

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

19

Acknowledgements 410

411

We thank Freimann Life Science Center for excellent animal care, and Dr. John 412

Adams for helpful discussions concerning murine malaria. We also are grateful 413

to Dr. Tom McCutchan for advice on the Plasmodium 18S RNA assays and to 414

Ursula Krzych for her insightful suggestions. This work was supported by a grant 415

from the Defense Advanced Research Projects Agency of the Department of 416

Defense (#W911NF-04-1-0380).417

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

20

References 418

1. Alger, N. E., and J. Harant. 1976. Plasmodium berghei: sporozoite challenge, 419

protection, and hypersensitivity in mice. Exp Parasitol 40:273-280. 420

2. Alger, N. E., J. A. Harant, L. C. Willis, and G. M. Jorgensen. 1972. Sporozoite 421

and normal salivary gland induced immunity in malaria. Nature 238:341. 422

3. Allen, J. R. 1989. Immunology of interactions between ticks and laboratory 423

animals. Experimental & applied acarology 7:5-13. 424

4. Belkaid, Y., S. Kamhawi, G. Modi, J. Valenzuela, N. Noben-Trauth, E. 425

Rowton, J. Ribeiro, and D. L. Sacks. 1998. Development of a natural model of 426

cutaneous leishmaniasis: powerful effects of vector saliva and saliva preexposure 427

on the long-term outcome of Leishmania major infection in the mouse ear dermis. 428

J Exp Med 188:1941-1953. 429

5. Belnoue, E., F. T. Costa, T. Frankenberg, A. M. Vigario, T. Voza, N. Leroy, 430

M. M. Rodrigues, I. Landau, G. Snounou, and L. Renia. 2004. Protective T 431

cell immunity against malaria liver stage after vaccination with live sporozoites 432

under chloroquine treatment. J Immunol 172:2487-2495. 433

6. Billingsley, P. F., J. Baird, J. A. Mitchell, and C. Drakeley. 2006. Immune 434

interactions between mosquitoes and their hosts. Parasite Immunol 28:143-153. 435

7. Bjorkman, A., and A. Bhattarai. 2005. Public health impact of drug resistant 436

Plasmodium falciparum malaria. Acta Trop 94:163-169. 437

8. Brossard, M., and S. K. Wikel. 2004. Tick immunobiology. Parasitology 129 438

Suppl:S161-176. 439

9. Bruna-Romero, O., J. C. Hafalla, G. Gonzalez-Aseguinolaza, G. Sano, M. 440

Tsuji, and F. Zavala. 2001. Detection of malaria liver-stages in mice infected 441

through the bite of a single Anopheles mosquito using a highly sensitive real-time 442

PCR. Int J Parasitol 31:1499-1502. 443

10. Chen, Y. L., F. E. Simons, and Z. Peng. 1998. A mouse model of mosquito 444

allergy for study of antigen-specific IgE and IgG subclass responses, lymphocyte 445

proliferation, and IL-4 and IFN-gamma production. Int Arch Allergy Immunol 446

116:269-277. 447

11. Christe, M., B. Rutti, and M. Brossard. 2000. Cytokines (IL-4 and IFN-448

gamma) and antibodies (IgE and IgG2a) produced in mice infected with Borrelia 449

burgdorferi sensu stricto via nymphs of Ixodes ricinus ticks or syringe 450

inoculations. Parasitology research 86:491-496. 451

12. Collins, F. H., F. Zavala, P. M. Graves, A. H. Cochrane, R. W. Gwadz, J. 452

Akoh, and R. S. Nussenzweig. 1984. First field trial of an immunoradiometric 453

assay for the detection of malaria sporozoites in mosquitoes. Am J Trop Med Hyg 454

33:538-543. 455

13. De Souza, J. B., K. H. Williamson, T. Otani, and J. H. Playfair. 1997. Early 456

gamma interferon responses in lethal and nonlethal murine blood-stage malaria. 457

Infect Immun 65:1593-1598. 458

14. Depinay, N., F. Hacini, W. Beghdadi, R. Peronet, and S. Mecheri. 2006. Mast 459

cell-dependent down-regulation of antigen-specific immune responses by 460

mosquito bites. J Immunol 176:4141-4146. 461

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

21

15. Dodoo, D., F. M. Omer, J. Todd, B. D. Akanmori, K. A. Koram, and E. M. 462

Riley. 2002. Absolute levels and ratios of proinflammatory and anti-inflammatory 463

cytokine production in vitro predict clinical immunity to Plasmodium falciparum 464

malaria. J Infect Dis 185:971-979. 465

16. Doolan, D. L., and S. L. Hoffman. 2000. The complexity of protective immunity 466

against liver-stage malaria. J Immunol 165:1453-1462. 467

17. Doolan, D. L., and S. L. Hoffman. 1999. IL-12 and NK cells are required for 468

antigen-specific adaptive immunity against malaria initiated by CD8+ T cells in 469

the Plasmodium yoelii model. J Immunol 163:884-892. 470

18. Edwards, J. F., S. Higgs, and B. J. Beaty. 1998. Mosquito feeding-induced 471

enhancement of Cache Valley Virus (Bunyaviridae) infection in mice. J Med 472

Entomol 35:261-265. 473

19. Ferreira, B. R., and J. S. Silva. 1999. Successive tick infestations selectively 474

promote a T-helper 2 cytokine profile in mice. Immunology 96:434-439. 475

20. Fong, T. A., and T. R. Mosmann. 1989. The role of IFN-gamma in delayed-type 476

hypersensitivity mediated by Th1 clones. J Immunol 143:2887-2893. 477

21. Ganapamo, F., B. Rutti, and M. Brossard. 1995. In vitro production of 478

interleukin-4 and interferon-gamma by lymph node cells from BALB/c mice 479

infested with nymphal Ixodes ricinus ticks. Immunology 85:120-124. 480

22. Gil, L. H., F. P. Alves, H. Zieler, J. M. Salcedo, R. R. Durlacher, R. P. Cunha, 481

M. S. Tada, L. M. Camargo, E. P. Camargo, and L. H. Pereira-da-Silva. 482 2003. Seasonal malaria transmission and variation of anopheline density in two 483

distinct endemic areas in Brazilian Amazonia. J Med Entomol 40:636-641. 484

23. Good, M. F., D. Stanisic, H. Xu, S. Elliott, and M. Wykes. 2004. The 485

immunological challenge to developing a vaccine to the blood stages of malaria 486

parasites. Immunol Rev 201:254-267. 487

24. Kamhawi, S., Y. Belkaid, G. Modi, E. Rowton, and D. Sacks. 2000. Protection 488

against cutaneous leishmaniasis resulting from bites of uninfected sand flies. 489

Science 290:1351-1354. 490

25. Keane-Myers, A., C. R. Maliszewski, F. D. Finkelman, and S. P. Nickell. 491

1996. Recombinant IL-4 treatment augments resistance to Borrelia burgdorferi 492

infections in both normal susceptible and antibody-deficient susceptible mice. J 493

Immunol 156:2488-2494. 494

26. Limesand, K. H., S. Higgs, L. D. Pearson, and B. J. Beaty. 2003. Effect of 495

mosquito salivary gland treatment on vesicular stomatitis New Jersey virus 496

replication and interferon alpha/beta expression in vitro. J Med Entomol 40:199-497

205. 498

27. Limesand, K. H., S. Higgs, L. D. Pearson, and B. J. Beaty. 2000. Potentiation 499

of vesicular stomatitis New Jersey virus infection in mice by mosquito saliva. 500

Parasite Immunol 22:461-467. 501

28. Lines, J. D., T. J. Wilkes, and E. O. Lyimo. 1991. Human malaria 502

infectiousness measured by age-specific sporozoite rates in Anopheles gambiae in 503

Tanzania. Parasitology 102 Pt 2:167-177. 504

29. Luckhart, S., Y. Vodovotz, L. Cui, and R. Rosenberg. 1998. The mosquito 505

Anopheles stephensi limits malaria parasite development with inducible synthesis 506

of nitric oxide. Proc Natl Acad Sci U S A 95:5700-5705. 507

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

22

30. Luke, T. C., and S. L. Hoffman. 2003. Rationale and plans for developing a 508

non-replicating, metabolically active, radiation-attenuated Plasmodium 509

falciparum sporozoite vaccine. J Exp Biol 206:3803-3808. 510

31. Luty, A. J., B. Lell, R. Schmidt-Ott, L. G. Lehman, D. Luckner, B. Greve, P. 511

Matousek, K. Herbich, D. Schmid, F. Migot-Nabias, P. Deloron, R. S. 512 Nussenzweig, and P. G. Kremsner. 1999. Interferon-gamma responses are 513

associated with resistance to reinfection with Plasmodium falciparum in young 514

African children. J Infect Dis 179:980-988. 515

32. Mbow, M. L., J. A. Bleyenberg, L. R. Hall, and R. G. Titus. 1998. 516

Phlebotomus papatasi sand fly salivary gland lysate down-regulates a Th1, but up-517

regulates a Th2, response in mice infected with Leishmania major. J Immunol 518

161:5571-5577. 519

33. McElroy, P. D., J. C. Beier, C. N. Oster, C. Beadle, J. A. Sherwood, A. J. 520

Oloo, and S. L. Hoffman. 1994. Predicting outcome in malaria: correlation 521

between rate of exposure to infected mosquitoes and level of Plasmodium 522

falciparum parasitemia. Am J Trop Med Hyg 51:523-532. 523

34. Meding, S. J., S. C. Cheng, B. Simon-Haarhaus, and J. Langhorne. 1990. 524

Role of gamma interferon during infection with Plasmodium chabaudi chabaudi. 525

Infect Immun 58:3671-3678. 526

35. Norsworthy, N. B., J. Sun, D. Elnaiem, G. Lanzaro, and L. Soong. 2004. Sand 527

fly saliva enhances Leishmania amazonensis infection by modulating interleukin-528

10 production. Infect Immun 72:1240-1247. 529

36. Nussenzweig, R. S., J. Vanderberg, H. Most, and C. Orton. 1967. Protective 530

immunity produced by the injection of x-irradiated sporozoites of plasmodium 531

berghei. Nature 216:160-162. 532

37. Osorio, J. E., M. S. Godsey, G. R. Defoliart, and T. M. Yuill. 1996. La Crosse 533

viremias in white-tailed deer and chipmunks exposed by injection or mosquito 534

bite. Am J Trop Med Hyg 54:338-342. 535

38. Peng, Z., and F. E. Simons. 2004. Mosquito allergy: immune mechanisms and 536

recombinant salivary allergens. Int Arch Allergy Immunol 133:198-209. 537

39. Poluektova, L., S. Gorantla, J. Faraci, K. Birusingh, H. Dou, and H. E. 538

Gendelman. 2004. Neuroregulatory events follow adaptive immune-mediated 539

elimination of HIV-1-infected macrophages: studies in a murine model of viral 540

encephalitis. J Immunol 172:7610-7617. 541

40. Ribeiro, J. M., and I. M. Francischetti. 2003. Role of arthropod saliva in blood 542

feeding: sialome and post-sialome perspectives. Annu Rev Entomol 48:73-88. 543

41. Richter, D., A. Spielman, and F. R. Matuschka. 1998. Effect of prior exposure 544

to noninfected ticks on susceptibility of mice to Lyme disease spirochetes. 545

Applied and environmental microbiology 64:4596-4599. 546

42. Riley, E. M., L. Hviid, and T. G. Theander. 1994. Parasitic Infections and the 547

Immune System. New York. 548

43. Rocha, A. C., E. M. Braga, M. S. Araujo, B. S. Franklin, and P. F. Pimenta. 549

2004. Effect of the Aedes fluviatilis saliva on the development of Plasmodium 550

gallinaceum infection in Gallus (gallus) domesticus. Mem Inst Oswaldo Cruz 551

99:709-715. 552

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

23

44. Sakai, T., H. Hisaeda, Y. Nakano, M. Zhang, M. Takashima, K. Ishii, Y. 553

Maekawa, S. Matsumoto, Y. Nitta, J. Miyazaki, S. Yamamoto, and K. 554 Himeno. 2003. Gene gun-based co-immunization of merozoite surface protein-1 555

cDNA with IL-12 expression plasmid confers protection against lethal 556

Plasmodium yoelii in A/J mice. Vaccine 21:1432-1444. 557

45. Schneider, B. S., L. Soong, Y. A. Girard, G. Campbell, P. Mason, and S. 558

Higgs. 2006. Potentiation of West Nile encephalitis by mosquito feeding. Viral 559

immunology 19:74-82. 560

46. Schneider, B. S., L. Soong, N. S. Zeidner, and S. Higgs. 2004. Aedes aegypti 561

salivary gland extracts modulate anti-viral and TH1/TH2 cytokine responses to 562

sindbis virus infection. Viral immunology 17:565-573. 563

47. Sedegah, M., F. Finkelman, and S. L. Hoffman. 1994. Interleukin 12 induction 564

of interferon gamma-dependent protection against malaria. Proc Natl Acad Sci U 565

S A 91:10700-10702. 566

48. Struik, S. S., and E. M. Riley. 2004. Does malaria suffer from lack of memory? 567

Immunol Rev 201:268-290. 568

49. Su, Z., and M. M. Stevenson. 2000. Central role of endogenous gamma 569

interferon in protective immunity against blood-stage Plasmodium chabaudi AS 570

infection. Infect Immun 68:4399-4406. 571

50. Titus, R. G., J. V. Bishop, and J. S. Mejia. 2006. The immunomodulatory 572

factors of arthropod saliva and the potential for these factors to serve as vaccine 573

targets to prevent pathogen transmission. Parasite Immunol 28:131-141. 574

51. Totemeyer, S., N. Foster, P. Kaiser, D. J. Maskell, and C. E. Bryant. 2003. 575

Toll-like receptor expression in C3H/HeN and C3H/HeJ mice during Salmonella 576

enterica serovar Typhimurium infection. Infect Immun 71:6653-6657. 577

52. Trung, H. D., W. Van Bortel, T. Sochantha, K. Keokenchanh, N. T. Quang, 578

L. D. Cong, and M. Coosemans. 2004. Malaria transmission and major malaria 579

vectors in different geographical areas of Southeast Asia. Trop Med Int Health 580

9:230-237. 581

53. Vanderberg, J. P., and U. Frevert. 2004. Intravital microscopy demonstrating 582

antibody-mediated immobilisation of Plasmodium berghei sporozoites injected 583

into skin by mosquitoes. Int J Parasitol 34:991-996. 584

54. Vaughan, J. A., L. F. Scheller, R. A. Wirtz, and A. F. Azad. 1999. Infectivity 585

of Plasmodium berghei sporozoites delivered by intravenous inoculation versus 586

mosquito bite: implications for sporozoite vaccine trials. Infect Immun 67:4285-587

4289. 588

55. Wanasen, N., R. H. Nussenzveig, D. E. Champagne, L. Soong, and S. Higgs. 589

2004. Differential modulation of murine host immune response by salivary gland 590

extracts from the mosquitoes Aedes aegypti and Culex quinquefasciatus. Medical 591

and veterinary entomology 18:191-199. 592

56. Wikel, S. K., R. N. Ramachandra, D. K. Bergman, T. R. Burkot, and J. 593

Piesman. 1997. Infestation with pathogen-free nymphs of the tick Ixodes 594

scapularis induces host resistance to transmission of Borrelia burgdorferi by ticks. 595

Infect Immun 65:335-338. 596

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

24

57. Zeidner, N., M. Dreitz, D. Belasco, and D. Fish. 1996. Suppression of acute 597

Ixodes scapularis-induced Borrelia burgdorferi infection using tumor necrosis 598

factor-alpha, interleukin-2, and interferon-gamma. J Infect Dis 173:187-195. 599

58. Zeidner, N. S., S. Higgs, C. M. Happ, B. J. Beaty, and B. R. Miller. 1999. 600

Mosquito feeding modulates Th1 and Th2 cytokines in flavivirus susceptible 601

mice: an effect mimicked by injection of sialokinins, but not demonstrated in 602

flavivirus resistant mice. Parasite Immunol 21:35-44. 603

604

605

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

25

Figure Legends 606

Figure 1. Lower Parasite Burden in mice pre-sensitized to A. stephensi 607

saliva. Mice in naïve and pre-sensitized groups were exposed to the bites of P. 608

yoelii-infected A. stephensi. (A) Parasite Burden in the liver was detected by 609

quantitative real time RT-PCR and infection levels were normalized to mouse 610

HPRT and expressed as 40-∆∆ct. (B) Naïve (open squares) and Pre-sensitized 611

(open triangles) RNA isolated from the liver also was analyzed using a standard 612

curve generated using RNA harvested from known numbers of salivary gland 613

sporozoites (closed squares). (C) Blood stage infection was monitored each day 614

for 7 days via blood smear from both naïve (solid bars) and pre-sensitized (lined 615

bars) mice. This data is representative of three independent experiments (n=4-5 616

per experiment). 617

* p<0.05 and **p<0.10 by student’s T Test. 618

619

Figure 2. Pre-sensitization skews response towards Th1 Phenotype. 620

Cytokine levels were quantified in naïve (Naïve), pre-sensitized (Pres) and in 621

naïve (Naïve Bit) and pre-sensitized (Pres Bit) mice that received one final 622

exposure to A. stephensi bites 24 hours prior to analysis. Local (ear) (A-C) and 623

systemic (liver (D-F) and spleen (G-I)) tissue IFN-g (A,D,G) and IL-4 (B,E,H) 624

mRNA levels were quantified by quantitative real time PCR. (C,F,I) IFN-g and IL-625

4 expression levels were used to create a cytokine ratio (IFN-g:IL-4). Data is 626

representative of four independent experiments (n=5 per experiment). Error bars 627

representative of S.E.M. * p<0.05 and ** p<0.001 by Student’s T Test. 628

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

26

629

Figure 3. Lower hepatic parasitemia is IFN-g-dependent. Balb/c WT (closed) 630

and IFN-g KO (open) animals in both Naïve (squares) and Pres (triangles) 631

groups were exposed to the bites of P. yoelii-infected A. stephensi. (A) parasites 632

and (B) iNOS mRNA were quantified by quantitative real time RT-PCR 40 hours 633

post infection. Infection levels were normalized to mouse HPRT and expressed 634

as relative parasite burden (40-∆∆ct). Data is representative of two independent 635

experiments. (n=5 per experiment * p< 0.05). Error bars are representative of 636

S.E.M.. 637

638

Figure 4. Local induction of iNOS occurs 5 hours after exposure to 639

infected A. stephensi. Balb/c animals in both Naïve and Pres groups were 640

exposed to the bites of P. yoelii-infected A. stephensi and iNOS mRNA was 641

quantified by quantitative real time RT-PCR 5 hours post infection. Error bars 642

are representative of S.E.M. *p< 0.05 by Student’s T Test. Data is representative 643

of two independent experiments. (n=5 per experiment). 644

645

Figure 5. IL-12p40 is induced by Pre-sensitization. Balb/c animals in both 646

Naïve and Pres groups were exposed to the bites of P. yoelii-infected A. 647

stephensi and IL-12p40 mRNA was quantified in liver (A) and spleen (B) 40 648

hours post infection by quantitative real time RT-PCR. Error bars are 649

representative of S.E.M. *p< 0.05 by Student’s T Test. Data is representative of 650

two independent experiments. (n=4 per experiment). 651

ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

27

652

Figure 6. Protective phenomenon associated with pre-sensitization to 653

mosquito saliva evident at 20 hours post-infection and is localized to the 654

liver. Pre-sensitized (lined bars) and naïve (solid bars) animals were subjected 655

to the bites of P. yoelii 17xNL-infected A. stephensi. Animals were sacrificed at 656

5, 10, 20, and 30 hours post-infection and 18s rRNA levels were quantified in 657

ears (A) and livers (B). Pre-sensitized and naïve animals were infected with 1000 658

P. yoelii 17xNL sporozoites intravenously through the tail vein immediately 659

following a 4th pre-sensitization to uninfected bites, and euthanized 40 hours 660

post-infection, subsequently parasite 18s rRNA levels were quantified by 661

quantitative real time RT-PCR (C). Infection levels were normalized to mouse 662

HPRT and expressed as relative parasite burden (40-∆∆ct). Data is 663

representative of two independent experiments (n=4). Error bars represent the 664

S.E.M. *p<0.05 by Student’s T Test. 665 ACCEPTED

on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from

ACCEPTED on Septem

ber 12, 2018 by guesthttp://iai.asm

.org/D

ownloaded from