JGV Papers in Press. Published March 24, 2015 as doi:10.1099/vir.0.000117

Journal of General Virology Detection of exosomal prions in blood by immunochemistry techniques --Manuscript Draft-Manuscript Number:

JGV-D-14-00185R2

Full Title:

Detection of exosomal prions in blood by immunochemistry techniques

Short Title:

Exosomal prions in blood

Article Type:

Short Communication

Section/Category:

TSE Agents

Corresponding Author:

Maurizio Pocchiari Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità Roma, ITALY

First Author:

Francesca Properzi

Order of Authors:

Francesca Properzi Mariantonia Logozzi Hanin Abdel-Haq Cristina Federici Luana Lugini Tommaso Azzarito Ilaria Cristofaro Daniela di Sevo Elena Ferroni Franco Cardone Massimo Venditti Marisa Colone Emmanuel Comoy Valérie Durand Stefano Fais Maurizio Pocchiari

Abstract:

In most forms of prion diseases blood is infectious, but the detection by immunochemistry techniques of the only available marker of infection (the misfolded prion protein, PrPTSE) in blood remains elusive. We developed a novel method for the detection of PrPTSE in blood of prion-infected rodents based on the finding that PrPTSE is associated with plasma exosomes. However, further purification of exosome on sucrose gradient was necessary for removing plasma immunoglobulins, which interfere with PrPTSE masking its detection by immunochemistry. Finally, we report that about 20% of plasma infectivity is associated with exosomes.

Powered by Editorial Manager® and ProduXion Manager® from Aries Systems Corporation

Manuscript Including References (Word document) Click here to download Manuscript Including References (Word document): Properzi_text_revised_clean.docx

1

Detection of exosomal prions in blood by immunochemistry techniques

2

Running title: Exosomal prions in blood

3

Francesca Properzi, Mariantonia Logozzi, Hanin Abdel-Haq, Cristina Federici, Luana Lugini, Tommaso

4

Azzarito, Ilaria Cristofaro, Daniela di Sevo, Elena Ferroni, Franco Cardone, Massimo Venditti, Marisa Colone,

5

Emmanuel Comoy, Valérie Durand, Stefano Fais and Maurizio Pocchiari

6

Affiliations

7

Francesca Properzi, Hanin Abdel-Haq, Ilaria Cristofaro, Daniela di Sevo, Elena Ferroni, Franco Cardone,

8

Massimo Venditti, and Maurizio Pocchiari, Department of Cell Biology and Neurosciences, Istituto

9

Superiore di Sanità, Viale Regina Elena 299, 00161, Rome, Italy.

10

Mariantonia Logozzi, Cristina Federici, Luana Lugini, Tommaso Azzarito, and Stefano Fais, Department of

11

Therapeutic Research and Medicines Evaluation, Istituto Superiore di Sanità, 00161 Rome, Italy;

12

Marisa Colone, Department of Technologies and Health, Istituto Superiore di Sanità, 00161 Rome, Italy

13

Emmanuel Comoy and Valérie Durand, Institute of Emerging Diseases and Innovative Therapies, CEA,

14

Fontenay-aux-Roses, France

15 16

Corresponding author

17

Maurizio Pocchiari

18

Email: [email protected]

19

Telephone: +39 06 49903203

20

Fax: +39 06 49903805

21 22 23

2 24

Word Count Summary: 93

25

Word Count Text:

2411

26

Tables:

0

27

Figures:

3

28

Suppl. Figures:

4

29

3 30

Summary paragraph

31

In most forms of prion diseases blood is infectious, but the detection by immunochemistry techniques of

32

the only available marker of infection (the misfolded prion protein, PrPTSE) in blood remains elusive. We

33

developed a novel method for the detection of PrPTSE in blood of prion-infected rodents based on the

34

finding that PrPTSE is associated with plasma exosomes. However, further purification of exosome on

35

sucrose gradient was necessary for removing plasma immunoglobulins, which interfere with PrPTSE masking

36

its detection by immunochemistry. Finally, we report that about 20% of plasma infectivity is associated with

37

exosomes.

38

4 39

Transmissible spongiform encephalopathy (TSE) or prion diseases are fatal neurodegenerative disorders

40

affecting humans and animals caused by unique infectious particles (prions), which are mainly composed

41

by misfolded forms of the cellular prion protein (PrPC). PrPC is constitutively expressed, but converts to the

42

misfolded noxious structure (PrPTSE) either spontaneously or upon exposure to prions (Weissmann, 2004).

43

Prions are efficiently transmitted via blood in most animal models of TSE diseases (Brown et al., 2001)

44

(Houston et al., 2008) and in humans with variant and possibly sporadic CJD (Llewelyn et al., 2004) (Wroe

45

et al., 2006) (Puopolo et al., 2011) (Douet et al., 2014). Prion infectivity in blood is associated with both

46

cellular and non cellular components (Gregori et al., 2006) with estimated titres in plasma ranging from 1

47

to 10 infectious unit/ml at disease onset (Brown et al., 2001) (Douet et al., 2014). Although plasma has

48

been shown to carry infectivity, the identification of PrPTSE in infected animals and humans has been elusive

49

and limited to the use of highly sensitive (Castilla et al., 2005) (Saá et al., 2006) (Pan et al., 2007) (Orrú et

50

al., 2011) or complex (Edgeworth et al., 2011) methodologies. Because blood components likely interfere

51

with PrPTSE detection (Hartwell et al., 2005) (Abdel-Haq, 2014), we thought to bypass this difficulty by

52

looking at plasma exosomes. Exosomes are nanovesicles that carry RNAs, proteins, lipids, other metabolites

53

(Théry et al., 2009) (Fais et al., 2013), PrPC and, in prion-infected hosts, PrPTSE and infectivity (Fevrier et al.,

54

2004) (Alais et al., 2008) (Coleman et al., 2012) (Saá et al., 2014).

55

We first collected blood from intracerebrally 263K scrapie-infected hamsters (n=125) (Kimberlin and

56

Walker, 1977) or from uninfected and healthy animals as controls. Plasma was separated from blood cells

57

by centrifugation at 5,000 g for 30 minutes at room temperature. Single plasma fractions from infected

58

hamsters were pooled to a final volume of 250 ml and used for the measurement of infectivity and

59

purification of exosomes for the detection of PrPTSE by immunochemistry assays (Fig. 1). Bioassay was

60

performed by intracerebral inoculation of 124 six weeks-old female Syrian hamsters with 50 µl of thawed

61

plasma, under anesthesia with ketamine/xylazine (75 and 10 mg/kg respectively) mixture. Animals were

62

housed in accordance to the national guidelines in level-3 animal care facilities (agreement numbers A 92-

63

032-02 for animal care facilities, 92-189 for animal experimentation, CEA, France) and regularly monitored.

64

Animals with clinical signs or with no disease up to 600-700 days were euthanized, the brain removed and

65

tested for the presence of PrPTSE by the ELISA test as previously described (Barret et al., 2003). Briefly, PrPTSE

5 66

was extracted from brains by the Bio-Rad purification kit (USA), digested with proteinase K, and added into

67

anti-PrP SAF53 (Bertin Pharma, France) monoclonal antibody (mAb)-coated ELISA wells. Bound PrPTSE was

68

detected by incubation with the 11C6 (Bertin Pharma, France) anti-PrP mAb coupled with

69

acetylcholinesterase, and revealed using the Ellman substrate at 414 nm optical wavelength.

70

Eighteen animals resulted positive for scrapie (time to death, mean, 265 days; s.e.m., 23 days) with a

71

corresponding infectivity titre of 3.1±0.7 infectious dose (ID) per ml (Supplementary Fig. 1), in line with

72

previously published results on plasma samples (Gregori et al., 2006) (Brown et al., 2001). Infectivity titre

73

(ID) was calculated by the limiting dilution method according to the Poisson distribution (Gregori et al.,

74

2006).

75

From the same pool of plasma, exosomes were isolated from 5 ml aliquots of control and infectious plasma

76

pool by a previously published protocol (Caby et al., 2005). Briefly, exosome plasma pellets were prepared

77

by serial centrifugations; the resulting pellets were washed in PBS, filtered through a 0.22 μm filter

78

(Millipore, USA) and ultracentrifuged at 110,000g for 1 h. The exosome pellets were re-suspended in PBS,

79

lysis buffer or in 2.5 M sucrose buffer, for further analyses as indicated in Fig. 1. Antibodies used for

80

exosomal marker identification were the anti-Flotillin, anti-Tsg101, and anti-mouse CD81 (Santa Cruz,

81

Germany). For PrP identification the primary antibodies used were SAF84 and biotinylated-SAF84 (Bertin

82

Pharma, France), 3F4 (Millipore, Italy), and D18 (gift from R. A. Williamson, The Scripps, Florida). Western

83

blot analyses were performed as described (Lee et al., 2000) and the consistency of these preparations

84

confirmed by the detection of exosomal markers (Fig. 2a). Assuming a similar ratio between infectivity and

85

PrPTSE molecules in blood and in brain tissue (about 0.1 to 1 pg PrPTSE/infectious unit) (Brown et al., 2001),

86

we estimated that 25 µl of exosomal plasma pellets (EPPs), corresponding to 5 ml of the original plasma

87

volume and 15.5 IDs, were suitable for detecting PrPTSE by standard western blot analyses (Lee et al., 2000).

88

In EPPs from control and prion-infected samples, we found that anti-PrP antibodies recognised the Ig light

89

chain (26 kD), and the IgG (53 kD) and IgM (65 kD) heavy chains as previously reported (Hartwell et al.,

90

2005). These bands were resistant to an amount (50 µg/ml) of proteinase K (PK) that is largely sufficient to

91

remove the bulk of tissue proteins in brain samples (Fig. 2). The atypical bands were seen irrespectively of

92

the anti-PrP primary antibody used (Supplementary Fig. 2) and were not detected in the absence of primary

6 93

antibodies (Fig. 2a). They were specific to plasma (Fig. 2a) and observed in EPPs of other species including

94

humans (data not shown) and wild type mice (C57BL/10) (Fig. 2a and supplementary Fig. 2). They were also

95

reproduced in EPPs of PrP knock-out 129/Ola mice (gift from Dr Di Bari and Dr Agrimi, Istituto Superiore di

96

Sanità, Rome, Italy) (Fig. 2a and supplementary Fig. 2) clearly indicating a cross-reaction of the primary

97

antibodies with plasmatic PK-resistant material, unrelated to PrP epitopes. Finally, when primary anti-PrP

98

antibodies were pre-absorbed with an excess of Syrian hamster 23-231 recombinant PrP (recPrP) (5:1),

99

western blot membranes were negative, demonstrating that pre-absorbed primary antibodies are unable

100

to bind PK-resistant Igs (IgRES). These data suggest that the levels of endogenous PrPC in plasma (in the

101

range of pg/ml) is not sufficient to replace the non-specific binding of largely represented plasma proteins

102

(in the range of mg/ml).

103

Stringent PK conditions (100 µg/ml), however, digested high molecular weight IgRES bands (Fig. 2a)

104

allowing the recognition of three bands of the same molecular weight of full length prion protein with a

105

glycoform ratio similar to that observed in hamster brain (Fig. 2a) (Meade-White et al., 2009). We then

106

further purified EPPs on a sucrose cushion starting from 15 ml of plasma to verify that these bands

107

effectively belong to full-length prion protein. Continuous sucrose gradient for the isolation and purification

108

of exosomes was performed as published (Raposo et al., 1996), with some modifications. Briefly, purified

109

exosomes were re-suspended in 2 ml of 2.5 M sucrose, 20 mM Hepes/NaOH, pH 7.2. A linear sucrose

110

gradient (2.0-0.25 M sucrose) was layered on top of the exosomes suspension in a UltraClear™ tube and

111

the sample was centrifuged at 110,000 g for 16 h at 4oC in a Beckman-Coulter (USA) SW41Ti rotor. Gradient

112

fractions (12 x 1 ml) were collected from the top of the tube and washed with PBS. Fractions density was

113

evaluated using an Abbé Refractometer (Carl Zeiss, Germany).

114

Silver

115

http://imagej.nih.gov/ij/) showed that after sucrose gradient ultracentrifugation the bulk of the

116

immunoglobulins was retained at high-density fractions (1.64 to 1.70 g/ml), while exosomal fractions (1.11

117

to 1.13 g/ml) had significantly lower content of IgRES. On sucrose gradient purified EPPs, di-glycosylated

118

PrPC bands (37 kD) were detected at densities of 1.13 g/ml (Fig. 3) and disappeared after PK treatment (Fig.

119

3). This fraction was also positive on western blot to standard exosomal markers (Tsg101, Flotillin and

staining

after

PAGE

and

quantification

of

bands

using

ImageJ

(NIH,

available

at

7 120

CD81) and negative to ER and nucleus markers (calnexin and nucleoporin) indicating the purity of exosomal

121

preparation after sucrose gradient isolation (Fig. 3). As controls, ER and nucleus markers resulted positive in

122

the brain homogenate (Fig. 3).

123

In infectious EPPs, 37 kD PrP bands were observed in exosomal fractions at densities of 1.13 g/ml (Fig. 3).

124

After treatment with PK, PrP-positive bands showed the characteristic shift in molecular weights of PrPTSE

125

(27-30 kD) with a glycoform ratio similar to that observed in 263K-infected brain homogenate (Fig. 3). The

126

presence of PrPC on plasma exosomes was further ascertained by electron microscopy. Immunoelectron

127

microscopy was performed on isolated sucrose gradient fractions of hamster plasma exosomes. A drop of

128

exosome suspension was placed onto a formvar-carbon-coated grid. After primary antibody (SAF84)

129

incubation exosomes were labelled with anti-rabbit IgG or anti-mouse IgG 5nm diameter-gold particle-

130

conjugated antibody (Sigma-Aldrich, USA). Grids were negatively stained with uranyl acetate solution and

131

examined with a Philips 208 transmission electron microscope (FEI Company, The Netherlands). In the 1.13

132

g/ml fraction there was presence of nanovesicles, which after immunogold staining, were PrP-positive

133

particles on the surface of nanovesicles (~50 nm) (Supplementary Fig. 3a and 3b). On these samples,

134

nanovesicles of the same size were positive to the standard exosomal marker Rab5b and no PrP staining

135

was observed in fractions of lower and higher densities (Supplementary Fig. 3c).

136

Finally, to further confirm the presence of prion in plasma exosomes we measured scrapie infectivity.

137

However, for this experiment we used the mouse-adapted 139A scrapie strain (Dickinson, 1976), because

138

139A, but not 263K infectivity can be quickly and accurately measured by using the End Point Scrapie Cell

139

Assay (SCEPA) (Klöhn et al., 2003). Susceptible CAD5 cells (gift from Prof. Charles Weissmann, The Scripps,

140

Florida, USA) were seeded in wells of 96-well plate and exposed to 12 or more aliquots of the 139A prion-

141

containing samples for 3 days, grown to confluence and split 1:3 three times and 1:10 twice. When the cells

142

reached confluence, 25,000 cells were transferred to membranes of ELISpot plates and PrPTSE-containing

143

cells were identified as spots using the D18 antibody (Supplementary Fig. 4). A well is considered positive

144

when the following two criteria are fulfilled. Firstly, the number of positive spots must be higher then the

145

mean plus 5 times standard deviations of unseeded wells (background value). Secondly, prion propagation,

146

revealed by an increased number of positive spots after subsequent passages, must be evident. The ID was

8 147

calculated considering the number of negative wells over positive by using a Poisson distribution. A titred

148

brain homogenate is used as positive control to monitor variations caused by antibodies. Nanovesicles

149

were isolated from 3 ml of plasma of 139A-infected mice at terminal stage. We found 1 of 6 positive wells

150

for exosomal samples and 4 of 48 positive wells for the 1:5 dilution of plasma supernatant. Accordingly to

151

the Poisson distribution for rare events, the IDs contained in the processed volume (3 ml) were 1.1 for the

152

exosomal pellet and 4.3 for plasma supernatant. These findings confirm that in vivo prions are associated to

153

nanovesicles similarly to what previously observed in vitro (Fevrier et al., 2004) (Alais et al., 2008) (Coleman

154

et al., 2012) (Fauré et al., 2006) (Vella et al., 2007) and that in plasma about 20% of total infectivity is

155

associated to exosomes leaving open the issue on how the remaining infectivity is distributed.

156 157

The most important finding of our work is the successful identification of PrP TSE in plasma of scrapie-

158

infected hamsters by standard western blot technique performed on exosomal fractions. This result

159

enforces previous data showing the presence of PrPTSE in blood exosomes by the protein misfolding cyclic

160

amplification (PMCA) method (Saá et al., 2014), in plasma by the PMCA (Castilla et al., 2005) and RT-QuiC

161

assays (Orrú et al., 2011) and in human vCJD patients by the solid-state binding matrix assay (Edgeworth et

162

al., 2011), but adds the important novel information that PrPTSE, similarly to infectivity (Brown et al., 2001)

163

is detectable in blood without using amplification techniques. The detection of PrP TSE by the standard

164

western blot technique was only possible after reducing the interference of IgRES by exosomal pellet

165

fractionation on a sucrose gradient. IgRES compete for the primary antibody binding with endogenous PrP

166

epitopes crucially interfering with PrPTSE detection in blood. This result confirms that blood components

167

interfere with prion diagnosis (Hartwell et al., 2005) (Abdel-Haq, 2014) reducing the sensitivity and

168

specificity of the various tested assays. Moreover, these findings might explain the discrepancy between

169

the excellent performances of tests accomplished on spiked blood and their failure on endogenously

170

infected blood that is likely related to different interaction with plasma Igs between exogenous and

171

endogenous PrPTSE.

172 173

9 174

Acknowledgments

175

We thank Dr Anna Ladogana for critical revision of the manuscript, Giuseppe Loreto for the preparations of

176

the figures, Dr Agnese Molinari for support with EM, Nick Verity for supplying the SCEPA samples, and Dr

177

Alessandra Garozzo for editorial assistance.

178

This work was partially supported by the Istituto Superiore di Sanità, Rome, Italy and by the Alliance

179

Biosecure Project “Plasmasecure”.

180 181

10 182

REFERENCES

183

Abdel-Haq, H. (2014) Factors intrinsic and extrinsic to blood hamper the development of a routine blood

184

test for human prion diseases. J Gen Virol doi: 10.1099/vir.0.070979-0

185

Alais, S., Simoes, S., Baas, D., Lehmann, S., Raposo, G., Darlix, J. L. & Leblanc, P. (2008). Mouse

186

neuroblastoma cells release prion infectivity associated with exosomal vesicles. Biol Cell 100, 603–615.

187

Barret, A., Tagliavini, F., Forloni, G., Bate, C., Salmona, M., Colombo, L., De Luigi, A., Limido, L., Suardi, S.

188

& other authors. (2003). Evaluation of quinacrine treatment for prion diseases. J Virol 77, 8462–8469.

189

Brown, P., Cervenáková, L. & Diringer, H. (2001). Blood infectivity and the prospects for a diagnostic

190

screening test in Creutzfeldt-Jakob disease. J Lab Clin Med 137, 5–13.

191

Caby, M.-P., Lankar, D., Vincendeau-Scherrer, C., Raposo, G. & Bonnerot, C. (2005). Exosomal-like vesicles

192

are present in human blood plasma. Int Immunol 17, 879–887.

193

Castilla, J., Saá, P. & Soto, C. (2005). Detection of prions in blood. Nat Med 11, 982–985.

194

Coleman, B. M., Hanssen, E., Lawson, V. A. & Hill, A. F. (2012). Prion-infected cells regulate the release of

195

exosomes with distinct ultrastructural features. FASEB J 26, 4160–4173.

196

Dickinson, A.G. (1976) Scrapie in sheep and goats. In Slow Virus Diseases of Animals and Man. Frontiers of

197

Biology series, vol. 44, chapter 10, pp. 209-241. Edited by R. H. Kimberlin. Amsterdam and New York: North

198

Holland.

199

Douet, J.Y., Zafar, S., Perret-Liaudet, A., Lacroux, C., Lugan, S., Aron, N., Cassard, H., Ponto, C., Corbière, F.

200

& other authors. (2014). Detection of infectivity in blood of persons with variant and sporadic Creutzfeldt-

201

Jakob disease. Emerg Infect Dis 20, 114-117.

202

Edgeworth, J. A., Farmer, M., Sicilia, A., Tavares, P., Beck, J., Campbell, T., Lowe, J., Mead, S., Rudge, P. &

203

other authors. (2011). Detection of prion infection in variant Creutzfeldt-Jakob disease: a blood-based

204

assay. Lancet 377, 487–493.

11 205

Fais, S., Logozzi, M., Lugini, L., Federici, C., Azzarito, T., Zarovni, N. & Chiesi, A. (2013). Exosomes: the ideal

206

nanovectors for biodelivery. Biol Chem 394, 1–15.

207

Fauré, J., Lachenal, G., Court, M., Hirrlinger, J., Chatellard-Causse, C., Blot, B., Grange, J., Schoehn, G.,

208

Goldberg, Y. & other authors. (2006). Exosomes are released by cultured cortical neurones. Mol Cell

209

Neurosci 31, 642–648.

210

Fevrier, B., Vilette, D., Archer, F., Loew, D., Faigle, W., Vidal, M., Laude, H. & Raposo, G. (2004). Cells

211

release prions in association with exosomes. Proc Natl Acad Sci USA 101, 9683–9688.

212

Gregori, L., Gurgel, P. V., Lathrop, J. T., Edwardson, P., Lambert, B. C., Carbonell, R. G., Burton, S. J.,

213

Hammond, D. J. & Rohwer, R. G. (2006). Reduction in infectivity of endogenous transmissible spongiform

214

encephalopathies present in blood by adsorption to selective affinity resins. Lancet 368, 2226–2230.

215

Hartwell, R. C., Nelson, M. S., Kislan, M. M., Stenland, C. J., Miller, J. L. C., Pifat, D. Y., Petteway, S. R., Jr &

216

Cai, K. (2005). An improved Western blot assay to assess the clearance of prion protein from plasma-

217

derived therapeutic proteins. J Virol Methods 125, 187–193.

218

Houston, F., McCutcheon, S., Goldmann, W., Chong, A., Foster, J., Sisó, S., González, L., Jeffrey, M. &

219

Hunter, N. (2008). Prion diseases are efficiently transmitted by blood transfusion in sheep. Blood 112,

220

4739–4745.

221

Kimberlin RH, Walker C. (1977). Characteristics of a short incubation model of scrapie in the golden

222

hamster. J Gen Virol 34, 295-304.

223

Klöhn, P.-C., Stoltze, L., Flechsig, E., Enari, M. & Weissmann, C. (2003). A quantitative, highly sensitive cell-

224

based infectivity assay for mouse scrapie prions. Proc Natl Acad Sci USA 100, 11666–11671.

225

Lee, D. C., Stenland, C. J., Hartwell, R. C., Ford, E. K., Cai, K., Miller, J. L., Gilligan, K. J., Rubenstein, R.,

226

Fournel, M. & Petteway, S. R., Jr. (2000). Monitoring plasma processing steps with a sensitive Western blot

227

assay for the detection of the prion protein. J Virol Methods 84, 77–89.

12 228

Llewelyn, C. A., Hewitt, P. E., Knight, R. S. G., Amar, K., Cousens, S., Mackenzie, J. & Will, R. G. (2004).

229

Possible transmission of variant Creutzfeldt-Jakob disease by blood transfusion. Lancet 363, 417–421.

230

Meade-White, K. D., Barbian, K. D., Race, B., Favara, C., Gardner, D., Taubner, L., Porcella, S. & Race, R.

231

(2009). Characteristics of 263K scrapie agent in multiple hamster species. Emerging Infect Dis 15, 207–215.

232

Orrú, C. D., Wilham, J. M., Raymond, L. D., Kuhn, F., Schroeder, B., Raeber, A. J. & Caughey, B. (2011).

233

Prion disease blood test using immunoprecipitation and improved quaking-induced conversion. MBio 2,

234

e00078–00011.

235

Pan, T., Sethi, J., Nelsen, C., Rudolph, A., Cervenakova, L., Brown, P. & Orser, C. S. (2007). Detection of

236

misfolded prion protein in blood with conformationally sensitive peptides. Transfusion 47, 1418–1425.

237

Puopolo, M., Ladogana, A., Vetrugno, V. & Pocchiari, M. (2011). Transmission of sporadic Creutzfeldt-

238

Jakob disease by blood transfusion: risk factor or possible biases. Transfusion 51, 1556–1566.

239

Raposo, G., Nijman, H. W., Stoorvogel, W., Liejendekker, R., Harding, C. V., Melief, C. J. & Geuze, H. J.

240

(1996). B lymphocytes secrete antigen-presenting vesicles. J Exp Med 183, 1161–1172.

241

Saá, P., Castilla, J. & Soto, C. (2006). Presymptomatic detection of prions in blood. Science 313, 92–94.

242

Saá, P., Yakovleva, O., De Castro, J., Vasilyeva, I., De Paoli, S. H., Simak, J. & Cervenakova, L. (2014). First

243

demonstration of PrPTSE in extracellular vesicles from plasma of mice infected with mouse-adapted variant

244

Creutzfeldt-Jakob disease by in vitro amplification of misfolded prion protein. J Biol Chem 289, 29247-

245

29260.

246

Théry, C., Ostrowski, M. & Segura, E. (2009). Membrane vesicles as conveyors of immune responses. Nat

247

Rev Immunol 9, 581–593.

248

Vella, L. J., Sharples, R. A., Lawson, V. A., Masters, C. L., Cappai, R. & Hill, A. F. (2007). Packaging of prions

249

into exosomes is associated with a novel pathway of PrP processing. J Pathol 211, 582–590.

250

Weissmann, C. (2004). The state of the prion. Nat Rev Microbiol 2, 861–871.

13 251

Wroe, S. J., Pal, S., Siddique, D., Hyare, H., Macfarlane, R., Joiner, S., Linehan, J. M., Brandner, S.,

252

Wadsworth, J. D. F. & other authors. (2006). Clinical presentation and pre-mortem diagnosis of variant

253

Creutzfeldt-Jakob disease associated with blood transfusion: a case report. Lancet 368, 2061–2067.

254

14 255

Figure Legends

256

Figure 1

257

Infectivity of plasma pool was measured by bioassay. Positive animals were identified by ELISA detection of

258

PrPTSE on brain samples. Prion infected exosomal plasma pellets (EPPs) were prepared, treated with or

259

without proteinase K (PK) and analysed by western blot before and after sucrose gradient fractionation. IC,

260

intracerebral

Diagram of the main experimental procedure

261 262

Figure 2. Western blots of PrP in infected and control exosomal plasma pellets

263

a PrP positive immunoglobulin (Ig) bands were resistant to standard (50µg/ml; +) proteinase K (PK)

264

digestion (arrowheads) in both exosomal plasma pellets from control (cEPP) and infected (iEPP)

265

samples. The same conditions digested 37 kD PrP (arrow) of normal brain homogenate (NBH). After

266

stringent treatment with 100 µg/ml of PK (++), PrP-like bands were also seen, possibly due to

267

degradation (asterisk). EPPs of PrP knock-out (KO) mouse are positive to anti PrP SAF84 antibody. CD81

268

positive cEPP of both wild type (WT) and KO mice contain PrP positive Ig bands (arrowheads) that are

269

resistant to PK. No staining is observed without (W/O) the primary SAF84 antibody and in NBH. The

270

exosomal marker CD81 is shown in the lower panels. (n=3).

271

b Pre-absorption of anti-PrP SAF84 antibody with recombinant PrP (recPrP) (5:1) removes the recPrP

272

band, PrP bands (arrows) of NBH, and Ig bands of EPPs (arrowheads). No staining was observed in

273

absence of the primary SAF84 antibody. (n=3).

274

Numbers on the left indicate molecular weights in kD.

275 276 277

Figure 3

Silver staining and western blots of sucrose gradient fractions of infected and control exosomal plasma pellets

278

PAGE silver staining reveals that the bulk of plasma proteins are retained in the high-density fractions of

279

both exosomal plasma pellets from control (cEPP) and infected (iEPP) samples.

280

Western blots with SAF84 antibody show the presence of PrPC in fraction 6 (1.13 g/ml) of cEPPs (n=3),

281

which is abolished by the treatment with proteinase K (PK). PK resistant PrPTSE is shown in fraction 6 of

15 282

iEPPs. Western blots of sucrose fractions for exosomal markers (Tsg101, flotillin and CD81) show that

283

exosomes float in fractions 5 to 8. Absence of calnexin and nucleoporin indicates no cellular debris

284

contamination. nBH, normal hamster brain homogenate; sBH, scrapie-infected hamster brain homogenate.

Figure 1 Click here to download high resolution image

Figure 2 Click here to download high resolution image

Figure 3 Click here to download high resolution image

Supplementary Material Files Click here to download Supplementary Material Files: Supplementary Figure Legends.pdf

Detection of exosomal prions in blood by immunochemistry techniques.

In most forms of prion diseases, blood is infectious, but detection by immunochemistry techniques of the only available marker of infection (the misfo...
1MB Sizes 1 Downloads 4 Views