IAI Accepted Manuscript Posted Online 13 July 2015 Infect. Immun. doi:10.1128/IAI.00262-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Immunization with the MAEBL M2 domain protects against

2

lethal Plasmodium yoelii infection

3 4

Running title: MAEBL M2 domain as a malaria vaccine candidate.

5 6

Juliana A Leite1; Daniel Y Bargieri2; Bruna O Carvalho1,3; Letusa Albrecht1,4;

7

Stefanie CP Lopes1,5; Ana Carolina AV Kayano1; Alessandro S Farias1;

8

Wan Ni Chia6; Carla Claser6; Benoit Malleret6; Bruce Russell6,#;

9

Catarina Castiñeiras1; Leonilda MB Santos1; Marcelo Brocchi1;

10

Gerhard Wunderlich2; Irene S Soares7; Mauricio M Rodrigues8;

11

Laurent Rénia6; Fabio TM Costa1*.

12 13 14

1

15

UNICAMP. Campinas, SP. Brazil.

16

2

17

Brazil.

18

3

19

RJ, Brazil.

20

4

21

Brazil.

22

5

23

Manaus, AM, Brazil.

24

6

25

Research (A*STAR), Singapore, Singapore.

Department of Genetics, Evolution and Bioagents. University of Campinas –

Department of Parasitology, University of São Paulo – USP. São Paulo, SP,

Instituto Oswaldo Cruz, Fundação Oswaldo Cruz – FIOCRUZ. Rio de Janeiro,

Instituto Carlos Chagas, Fundação Oswaldo Cruz – FIOCRUZ. Curitiba, PR,

Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz – FIOCRUZ.

Singapore Immunology Network, Agency for Science, Technology and

1

26

7

27

University of São Paulo - USP, São Paulo, SP, Brazil.

28

8

29

São Paulo, São Paulo, SP, Brazil.

Department of Clinical and Toxicological Analysis, Pharmaceutical Sciences,

Center for Cellular and Molecular Therapy - CTCMol, Federal University of

30 31

Running head: MAEBL, M2 domain, malaria vaccine, Plasmodium yoelii.

32 33

#

34

Singapore – NUS, Singapore, Singapore.

Present address: Department of Microbiology, National University of

35 36

*Address correspondence to: Fabio T M. Costa, [email protected]

37 38

Abstract

39

Malaria remains a world threatening disease largely due to the lack of a

40

long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane

41

molecule with a chimeric cysteine-rich ectodomain homologous to regions of

42

DBL-EBP and AMA1 antigens. Although MAEBL does not appear to be

43

essential for the survival of blood-stage forms, the ectodomains M1 and M2,

44

homologous to AMA1, seem to be involved in parasite attachment to

45

erythrocytes, especially M2. MAEBL is necessary for sporozoite infection of

46

mosquito salivary glands and is expressed in liver stages. Here, the

47

Plasmodium yoelii MAEBL-M2 domain was expressed in a prokaryotic vector.

48

C57BL/6J mice were immunized with doses of the P. yoelii recombinant

49

protein (rPyM2-MAEBL). High levels of antibodies, with balanced IgG1/IgG2c

50

subclasses, were achieved. rPyM2-MAEBL antisera were capable of

2

51

recognizing the native antigen. Anti-MAEBL antibodies recognized different

52

MAEBL fragments expressed in CHO cells showing stronger IgM and IgG

53

response against the M2 domain and repeat region, respectively. After

54

challenge with P. yoelii YM (lethal strain)-infected erythrocytes, up to 90% of

55

immunized animals survived, and a reduction of parasitemia was observed.

56

Moreover, splenocytes harvested from immunized animals proliferated in a

57

dose-dependent manner in the presence of rPyM2-MAEBL. Protection was

58

highly dependent on CD4+, but not CD8+ T cells, towards Th1. rPyM2-

59

MAEBL antisera were also able to significantly inhibit parasite development as

60

observed in ex vivo P. yoelii erythrocyte invasion assays. Collectively, these

61

findings support the use of MAEBL as a vaccine candidate and open

62

perspectives to understand the mechanisms involved in protection.

63 64

Introduction

65

Malaria remains one of the most devastating infectious diseases in

66

inter-tropical countries, affecting mainly children under the age of five and

67

pregnant women. Approximately 600,000 deaths occur every year (1). People

68

repeatedly exposed to malarial infections in endemic areas develop immunity

69

against clinical disease and subsequently against parasitemia (2-5).

70

Antibodies have been shown to be responsible for naturally acquired immunity,

71

since passive transfer of immune IgG from adults can protect against

72

Plasmodium falciparum blood-stage infection (2, 6-8), suggesting that a

73

malaria vaccine based on asexual antigens is feasible. Unfortunately, none of

74

the vaccines currently tested achieved a convincing rate of protected

3

75

individuals (9-11) and the observed protection was often short–lived or highly

76

strain-specific (12-16).

77

The stakes for blood stage vaccines are even higher when malaria

78

eradication is the goal because the vaccines must not only reduce disease but

79

also reduce the parasitic burden to a degree that reduces the transmission

80

(17). Despite considerable efforts, none of the blood stage vaccine candidates

81

have exhibited satisfactory clinical and sterile protection in field tests (18, 19).

82

Many of the current vaccine candidates were encountered based on

83

the finding that partly immune individuals possess high titers of antibodies

84

against the tested antigens. Recently, the finding that antibodies against

85

PfRH5 are highly effective in blocking merozoite reinvasion but are rarely

86

detected in significant quantities in semi-immune carriers was reported (20).

87

This finding suggests that other merozoite-exposed antigens against which no

88

significant response is developed in natural infections may also be effective

89

as vaccines.

90

MAEBL is a 200-kDa type 1 membrane protein that belongs to the

91

erythrocyte binding protein (ebl) family (21-24). It has a carboxy cysteine-rich

92

region homologous to region IV of the DBL-EBP (Duffy binding like –

93

Erythrocyte binding protein). Additionally, the MAEBL amino cysteine-rich

94

domains (M1 and M2 ligand domains) exhibit partial similarity with the apical

95

membrane antigen 1 (AMA1) (23, 25). The M1 and M2 domains from P. yoelii

96

have been shown to be functionally equivalent to the DBL ligand domain, as

97

they bind to mouse erythrocytes (25).

98

MAEBL is essential for the development of the parasite during

99

sporozoite infection of mosquito salivary glands (26, 27) and is also

4

100

expressed in the salivary gland sporozoite and during the late liver stage (28).

101

Weak expression of MAEBL can also be detected in blood merozoite forms,

102

although maebl deletion has no impact on blood parasite development (26).

103

Coincidently, only few antibodies are found in naturally infected individuals

104

from areas with low transmission rates (29). MAEBL is a highly conserved

105

gene between evolutionarily distinct species of Plasmodium (25). Among the

106

clones of P. falciparum and field isolates, there is little amino acid sequence

107

variation on M1 and M2 domains (21). Because MAEBL is a well conserved

108

gene and expressed at different parasite stages, it is considered an

109

interesting potential vaccine candidate (30).

110

The current knowledge of the mechanisms and interactions during

111

Plasmodium invasion of the erythrocytes are still limited, which impairs the

112

development of approaches to block this essential step in Plasmodium biology.

113

As blocking erythrocyte invasion strategies are part of the rational of several

114

vaccine based on merozoite antigens, approaches designed to elucidate the

115

invasion phenomenon might facilitate validation and identification of potential

116

antigens that could be used as vaccine targets.

117

In this study, we investigated the immunogenicity of the domain M2-

118

MAEBL of P. yoelii. We amplified, cloned and expressed M2-MAEBL as a

119

prokaryotic fusion protein (rPyM2-MAEBL). Mice received four doses of the

120

recombinant antigen emulsified in Freund´s adjuvant. Immunization induced

121

robust protection against a lethal challenge with asexual forms of P. yoelii YM.

122

Protection was dependent on CD4+, but not CD8+ T cells, towards Th1. By

123

adapting an ex vivo invasion assay we could show that sera from immunized

124

mice inhibited invasion of parasite blood-stage forms. These results

5

125

demonstrate that MAEBL can be used as antigen in anti-malarial vaccine

126

formulations.

127 128

Methods

129

Parasites and animals

130

Six to seven week-old C57BL/6J mice were purchased from the

131

University of Campinas Animal Center (CEMIB-UNICAMP). Animals were

132

kept in a mouse pathogen free facility. All experiments and procedures were

133

approved by the Ethical Committee for Animal Research of the University of

134

Campinas (Protocol No. 1437-1). Blood forms of P. yoelii were obtained after

135

intraperitoneal (i.p.) injection of thawed glycerin frozen stocks.

136 137

Generation of the recombinant P. yoelii M2-MAEBL

138

DNA fragments encoding M2-MAEBL were obtained by PCR

139

amplification using Platinum Taq high-fidelity DNA polymerase (Invitrogen),

140

using P. yoelii genomic DNA from infected mice as a template. For the

141

amplification of the sequence corresponding to the M2 domain of the MAEBL

142

antigen, we used synthetic oligonucleotides based on the sequence from

143

amino acid 589 to 992 from the P. yoelii yoelii MAEBL gene (GenBank,

144

Accession number AF031886). For this, the following oligonucleotides were

145

used: (I) pET28aM2F 5’-CGC GGA TCC CTT AAC AAA TAT ATG AAA TCT

146

AAT GTT GAA CTT -3' and (II) anti-sense pIgSPM2R 5’-CTC GAA TTC CTA

147

CGA TTC ATC GGT ATT TCT TGT AG-3’; containing BamHI and EcoRI

148

sites, purchased from Invitrogen, USA. The resulting PCR product was

149

purified and digested with both restriction enzymes and inserted into the

6

150

pET28a vector (Novagen). The resulting plasmid was designated pET28aM2.

151

To verify that the reading frame was correct, inserts were sequenced using

152

vector-specific oligonucleotides covering the entire cloned sequence.

153

The recombinant protein (rPyM2-MAEBL) was expressed and purified

154

as previously described (31) with slight modifications. E. coli BL21 DE3

155

(Novagen) were transformed with the pET28aM2 plasmid and cultivated at

156

37°C in Luria-Bertani (LB) broth in the presence of 30 μg/ml kanamycin

157

(Sigma). Protein expression was induced at OD600 with 0,1 mM IPTG

158

(Invitrogen) for 4 h. After centrifugation, the bacterial pellet was resuspended

159

and sonicated in phosphate-buffered saline (PBS) supplemented with 1 mg/ml

160

lysozyme (Sigma) and 100 mM of PMSF (Sigma). The sonicated material was

161

then centrifuged, and the supernatant was separated from the pellet. Under

162

these conditions, rPyM2-MAEBL was found in the insoluble fraction. For

163

solubilization and purification, the pellet was resuspended in 100 mM Tris-HCl,

164

8 M urea, and 100 mM DTT, pH 8.0. Complete solubilization was achieved by

165

the passage of the solution through a 21-gauge needle. The solution was then

166

centrifuged and the supernatant applied to a column with N2+-NTA-agarose

167

resin (Qiagen). After extensive washing the recombinant protein was eluted

168

with a buffer with pH 5.0, and the eluted protein was dialyzed against PBS

169

with 2 M urea. Protein expression and concentration was determined by SDS-

170

PAGE and immunoblotting analysis.

171 172 173

7

174

Generation of a pdisplay library containing fragments of the MAEBL

175

immunogen

176

Primers were designed to be in-frame, complementary and specific to

177

different regions of MAEBL, without self-annealing sequences using the

178

software Amplify3x. Three different regions (MAEBL-3, MAEBL-4 and

179

MAEBL-5) encompassing the M2, repeat and C-terminal cysteine regions

180

respectively were designed. Six smaller, overlapping regions of the MAEBL-3

181

region (which contains different segments of the M2 region, of the region

182

between inter-domain between M2 and/or the repeats or repeats) were

183

designed and these were subcloned into the pDisplay vector (Invitrogen)

184

together with MAEBL-4 and MAEBL-5 constructs. The primer sequences used

185

to clone these fragments of MAEBL are all reported in Table S1.

186

Genes-of-interest were amplified with Phusion Hot Start II polymerase

187

(Finnzymes) according to manufacturer’s instructions, using the Biorad Peltier

188

thermal cycler. The PCR products were ligated into the XmaI and SacII (NEB)

189

linearized pDisplay vector according to manufacturer’s recommendations (In-

190

Fusion cloning, Clontech) before heat shock transformation into XL10 gold

191

bacteria (Agilent). Bacteria clones were screened by colony PCR using T7

192

and BgH primers for positive transformants. They were then sent for

193

sequencing to ensure codons were in-frame. The plasmids were purified

194

using Nucleobond AX Endotoxin-Free Midiprep kit (Macherey-Nagel) before

195

transfection. CHO (Chinese hamster cell) cells were used for the expression

196

of recombinant MAEBL proteins. Cells were grown in F12 complete medium

197

containing 100 units/ml penicillin (Gibco), 100 μg/ml streptomycin (Gibco) and

198

10% heat-inactivated fetal bovine serum (FBS) (Gibco) in a 37oC incubator

8

199

with 5% CO2. Sterile ethylene-diamine-tetra-acetic acid (EDTA) diluted to a

200

final concentration of 2 mM in PBS was used to detach transfected

201

cells/stable cell lines. Endofectin (Genecopeia) was used to transfect the

202

recombinant pDisplay plasmid into CHO cells. CHO cells were seeded into 6-

203

cm cell culture dishes one day before transfection in a density that would give

204

approximately 70 - 80% confluency on the day of transfection. To transfect the

205

cells, 9 μl of Endofectin was added dropwise with vortexing to 3 μg of plasmid

206

diluted in F12 supplemented with 100 units/ml penicillin and 100 μg/ml

207

streptomycin for 6-cm dishes. The DNA-Endofectin complex was incubated at

208

room temperature for 25 min before it was added dropwise to the cells. The

209

plates or dishes were swirled gently to distribute the transfection mixture

210

before returning them to the 37oC incubator overnight. The transfection

211

medium was replaced with fresh F12 complete medium the following day.

212 213

Immunoblotting analysis

214

Recombinant protein samples were loaded on SDS-PAGE under

215

denaturating conditions (2-mercaptoethanol). Proteins were then transferred

216

onto a nitrocellulose membrane (GE Healthcare). Immunoblotting was

217

conducted with anti-His6 (Life Technologies) at a 1:1500 dilution. The antibody

218

binding was detected by incubation with peroxidase-coupled goat anti-mouse

219

IgG (Sigma) at a 1:2000 dilution. Bands were detected using a solution

220

containing 50 mM Tris-HCl pH 7.5, 0.6 mg/mL of 3,3’ diaminobenzidine and 1

221

μL/mL of 0,03% H2O2 (Sigma) solution.

222 223

9

224

Immunization regimen

225

Groups of six to ten C57BL/6J male mice (Six- to 7-week-old) were

226

injected subcutaneously four times at three-week intervals with 5 μg of

227

rPyM2-MAEBL 1:1 (vol/vol) emulsified in complete Freund’s adjuvant (Sigma)

228

(CFA) for the first dose or incomplete Freund’s adjuvant (IFA) in the

229

subsequent doses. As a control group, animals were injected with 1:1 (vol/vol)

230

adjuvant (CFA/IFA) in PBS. Sera from immunized mice were collected

231

immediately before each dose and 3 weeks after the last dose (Figure 2A).

232 233

ELISA

234

Anti-rPyM2-MAEBL antibodies titers were determined by ELISA as

235

previously described (32). Plates were coated with 200 ng/well of recombinant

236

protein. Mice sera were tested at serial dilutions starting from 1:100. A dilution

237

of 1:2000 peroxidase-coupled goat anti-mouse IgG conjugated (Sigma) was

238

applied as a secondary antibody. Specific anti-MAEBL titers were determined

239

as the highest dilution yielding an OD492 higher than 0.1. Detection of IgG

240

subclasses was performed as described above, except that the secondary

241

antibody used was specific for mouse IgG1, IgG2b and IgG2c (Southern

242

Technologies).

243 244

Epitope mapping

245

Transfected cells (150 000 per well) were transiently transfected and seeded

246

in 96 well plates (Nunc). They were stained with serum diluted in FACS buffer

247

(PBS+10% FBS) for 1 h with agitation at 4oC. The different MAEBL constructs

248

are flanked by a myc epitope on the N-terminus and HA epitope on the C-

10

249

terminus when expressed on the surface of CHO cells, and both epitopes

250

were utilized to assess for protein expression on CHO cells using rabbit anti-

251

myc IgG (Upstate) or rabbit anti-HA IgG (Sigma) diluted 1:100. Untransfected

252

parental CHO cells were stained using the same sera and used as negative

253

control. For determination of serum recognition, the different test sera from

254

mice immunized with MAEBL protein or control vehicle DMSO were diluted

255

1:100 and incubated with the cells for 1 h with agitation at 4oC and then

256

washed once with FACS buffer. Secondary antibody – either goat anti-rabbit

257

IgG-AlexaFluor488 (Molecular Probes, Invitrogen) diluted 1:500 for checking

258

of protein expression or goat anti-mouse IgM or IgG-AlexaFluor488

259

(Molecular Probes) diluted 1:500, mixed with propidium iodide diluted to 0.001

260

mg/ml for the serum screening assay were incubated for 30 min with agitation

261

at 4oC. Labeled samples were analyzed using the high throughput flow

262

cytometer Accuri (Becton Dickinson). Flowjo (Tree Star) was used for all flow

263

cytometry analysis. Serum response was determined as the percentage of

264

cells recognized by serum divided by cells expressing the construct of interest

265

as illustrated in this formula:

266

Serum response = [(% cells recognized by immune serum – % cells

267

recognized by naïve serum) / (% Protein of interest POI expressing cells)] x

268

100%.

269

The 20% Cut-off for positivity was determined as the mean + 6 standard

270

deviation of the quadruplicate serum recognition values obtained for sera from

271

naïve animals. This high threshold was uses to reduce the possibility of false

272

positives.

273

Cellular proliferation and cytokines quantification

11

274

Ten

million

splenic cells

collected

from

animals

of

different

275

immunization groups (n=3) were cultured in 96-well plate in a final volume of

276

200 μL. rPyM2-MAEBL protein was added to the culture at a final

277

concentration of 1 or 10 µg/mL. As a control, some wells received ConA (50

278

µg/ml). Lymphocyte proliferation was assessed by the addition of 0.5 mCi/well

279

of methyl-[3H] thymidine ([3H] TdR (GE Healthcare) to the splenocytes after

280

48 h of culture. Splenocytes were cultured for an additional 18 h, and plates

281

were frozen. For later analysis, plates were thawed, and the culture was

282

collected on filter paper using a cell harvester. Tritium incorporation was

283

measured as radioactivity using Beta 1450 Microscintillator (Perkin Elmer).

284

The results are expressed as the average of the triplicate culture.

285

In parallel, culture supernatants were collected after 120 h, and the

286

amount of secreted IFN-γ, TNF, IL-2, IL-4 and IL-5 in cell culture supernatant

287

were simultaneously detected using the Cytometric Bead Array (CBA) Mouse

288

th1/th2 Cytokine Kit (BD). Cytokine concentrations in each sample were

289

determined with standard curves of known concentration of all five

290

recombinant proteins. Samples were read in a FACS Calibur flow cytometer

291

(Becton Dickinson). The detection limit of the assay was 0.5 pg/ml.

292 293

Immunofluorescence assays

294

Slides were prepared by collecting blood from BALB/c mice infected

295

with P. yoelii YM on day 5 post-infection (p.i.). The selection of mature stages

296

of the parasite was performed using LS-MACS columns (Miltenyi Biotec), and

297

thin blood smears were made, air-dried and stored until immunofluorescence

298

(IF) preparation and microscopy analysis. Immunofluorescence assays were

12

299

performed after fixing the blood smears with ice-cold acetone for 20 min and

300

air-dried. Well diameters were established with the aid of a Dako-PenTM

301

(Dako), and blocking was performed by 30-min incubation at 37°C with PBS

302

containing 3% BSA (USB). Pooled sera from the different immunization

303

groups and P. yoelii hyperimmune sera were diluted 1:100 in PBS

304

supplemented with 3% BSA and applied to the slides for 1 h at 37°C. Slides

305

were washed 3 X in PBS and incubated with Alexa-568 goat anti-mouse IgG

306

(Invitrogen) for 1 h at 37°C in the dark, then washed 3 X in PBS and

307

incubated

308

(Invitrogen) diluted in ultrapure (Millipore) water for 10 min at room

309

temperature. After another round of washing, Fluorosave (Caliobiochem) was

310

added, and slides were sealed with coverslips. Parasites were visualized with

311

the aid of a Nikon TS100 epifluorescence microscope. P. yoelii hyperimmune

312

sera were obtained from BALB/c mice infected with P. yoelii 17XNL clone 1.1

313

(non-lethal strain) once a month for a total period of 7 months. In parallel, to

314

confirm specificity, we also conducted immunofluorescence assays using

315

Leishmania amazonensis promastigote forms (gently donated by Dr. Selma

316

Giorgio, UNICAMP) and anti-rPyM2-MAEBL antisera.

with

DAPI

(4′,6-diamidino-2-phenylindole,

dihydrochloride-

317 318

Mice challenge and parasitemia assessment

319

The protection levels against blood forms were determined by

320

immunized mice survival against lethal challenge after intraperitoneal injection

321

of 106 P. yoelii YM-infected erythrocytes. Parasitemia was monitored,

322

according to Figure 2A, between 3 and 10 days post infection and determined

13

323

by counting at least 1,000 erythrocytes from thin blood smears stained with

324

the Panótico Rápido Kit (Laborclin).

325 326

CD4+ and CD8+ T lymphocyte depletion

327

Groups of 15 C57BL/6J mice were immunized as detailed in Figure 6A.

328

Three weeks after the fourth dose, five animals were injected with 0.5

329

mg/animal (Bioxcell) anti-CD4 (clone GK1.5) or anti-CD8 (clone YTS169.4)

330

monoclonal antibodies for the depletion of the corresponding lymphocyte

331

populations. The efficacy of In vivo depletion efficacy for this dose of

332

antibodies was determined in a preliminary experiment, as assessed by flow

333

cytometry of CD8+T and CD4+T cell using specific monoclonal antibodies

334

recognizing different epitopes in the CD4 or CD8 molecules. As a control, a

335

group of five immunized animals were injected with 0.5 mg per animal of

336

purified unrelated mouse IgG (Sigma, USA). On the following day, animals

337

were challenged with 106 P. yoelii YM IEs (lethal strain). The survival of

338

animals was monitored and parasitemia was evaluated as described

339

previously.

340 341

Ex vivo invasion assays of P. yoelii YM merozoites

342

To obtain high levels of reticulocytes to be used as target cells in the

343

invasion assay, reticulocytemia was induced in groups of four to six naive

344

mice by repeated daily retro-orbital bleeding for 6 days. Leukocyte removal

345

from the reticulocyte-enriched blood was achieved after two cycles of CF11

346

filtration (33).

347

Infected erythrocytes were enriched and maturated to schizonts.

14

348

Enrichment was achieved by bleeding P. yoelii YM-infected mice (n= 3) on

349

day 5 post-infection (>60% parasitemia) and isolating mature forms (15-18 h

350

of development) by magnetic concentration as previously described (34). A

351

blood smear was made after the procedure to determine the concentration of

352

parasites and verify the complete elimination of leukocytes. Next, to

353

synchronize parasites to at least 80% at the schizont stage, the pellet

354

corresponding to the mature forms of P. yoelii was resuspended in McCoy's

355

5A medium (Sigma) supplemented with 2.4 g/l glucose, 40 mg/ml gentamycin

356

(Sigma) and 20% human AB serum (UNICAMP’s blood bank), inactivated and

357

adsorbed on murine red blood cells and cultivated 3-6 h in 5% CO2 and at

358

37°C for maturation.

359

Finally, invasion assays were adapted from an assay for P. vivax [38]

360

performed by mixing P. yoelii concentrated schizonts and target cells

361

(enriched reticulocyte rich blood) at a 1:10 ratio corresponding to

362

approximately 10% of the initial parasitemia. The mixture was diluted to a 4%

363

hematocrit in 100 µl of McCoy's 5A medium supplemented with 2.4 g/l

364

glucose, 40 mg/ml gentamycin and then cultured for approximately 20 h in a

365

candle jar (35).

366

Immunized sera used in the invasion assays were heat inactivated by

367

incubation at 56ºC for 40 min. Pool sera from animals immunized with four

368

doses of the rPyM2-MAEBL diluted 1:100 were added to the culture. As

369

controls, the pool sera of animals injected four times with CFA/IFA at the

370

same dilution were used, and E64 (Sigma) at 100 μM was used as a positive

371

inhibition control because it ensures no schizont rupture. As a negative control

372

of inhibition, the assay was performed without addition of any drug or serum.

15

373

Freshly infected erythrocytes were determined by light microscopy and

374

parasitemia of young forms (rings and young trophozoites) was determined as

375

previously described. The standardized methodology for the P. yoelii invasion

376

assays is depicted in detail in Figure 7.

377 378

Statistical analysis

379

Survival curves of immunized animals after lethal challenge was

380

evaluated by the Log-Rank test. Comparison of the means of three or more

381

groups was performed using one-way ANOVA, and significance between

382

every two groups was assessed by the Bonferroni multiple comparison test

383

when data assumed Gaussian distribution, as assessed by the D’Agostino

384

and Pearson normality test. For samples of no Gaussian distribution, group

385

comparison was analyzed using the Kruskal-Wallis test followed by Dunn’s

386

test.

387

Whitney U test was used. All tests were performed using Prism GraphPad

388

software version 5.0a. The results were considered significant when P

Immunization with the MAEBL M2 Domain Protects against Lethal Plasmodium yoelii Infection.

Malaria remains a world-threatening disease largely because of the lack of a long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane...
1MB Sizes 0 Downloads 6 Views