Poly(I:C)-Encapsulating Nanoparticles Enhance Innate Immune

Oct 3, 2017 - Sandrine Luce , Sophie Guinoiseau , Alexis Gadault , Franck Letourneur , Bertrand Blondeau , Patrick Nitschke , Eric Pasmant , Michel Vi...
1 downloads 0 Views 2MB Size
Subscriber access provided by LAURENTIAN UNIV

Article

Poly(I:C)-encapsulating nanoparticles enhance innate immune responses to the tuberculosis vaccine Bacille-Calmette-Guérin (BCG) via synergistic activation of innate immune receptors Martin T. Speth, Urska Repnik, Elisabeth Müller, Julia Spanier, Ulrich Kalinke, Alexandre Corthay, and Gareth Griffiths Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00795 • Publication Date (Web): 03 Oct 2017 Downloaded from http://pubs.acs.org on October 5, 2017

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a free service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are accessible to all readers and citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

Molecular Pharmaceutics is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

1

Poly(I:C)-encapsulating nanoparticles enhance

2

innate immune responses to the tuberculosis vaccine

3

Bacille-Calmette-Guérin (BCG) via synergistic

4

activation of innate immune receptors

5 6

Martin T. Speth1, Urska Repnik1, Elisabeth Müller1,2, Julia Spanier3, Ulrich Kalinke3, Alexandre

7

Corthay2, Gareth Griffiths1*

8 9

10

1

2

Tumor Immunology lab, Department of Pathology, Rikshospitalet, Oslo University Hospital and University of Oslo, N-0424 Oslo, Norway,

11

12

Department of Biosciences, University of Oslo, N-0371 Oslo, Norway,

3

Institute for Experimental Infection Research, TWINCORE, Center for Experimental and

13

Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research,

14

Braunschweig, and the Hannover Medical School, D-30625 Hannover, Germany,

15

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

16

ABSTRACT

17

The attenuated live vaccine strain Bacille Calmette-Guérin (BCG) is currently the only available

18

vaccine against tuberculosis (TB), but is largely ineffective against adult pulmonary TB, the most

19

common disease form. This is in part due to BCG’s ability to interfere with the host innate

20

immune response, a feature that might be targeted to enhance the potency of this vaccine. Here,

21

we investigated the ability of chitosan-based nanoparticles (pIC-NPs) containing polyinosinic-

22

polycytidylic acid (poly(I:C)), an inducer of innate immunity via Toll-like receptor 3 (TLR3) to

23

enhance the immunogenicity of BCG in mouse bone marrow-derived macrophages (BMDM) in

24

vitro. Incorporation of poly(I:C) into NPs protected it against degradation by ribonucleases and

25

increased its uptake by mouse BMDM. Whereas soluble poly(I:C) was ineffective, pIC-NPs

26

strongly enhanced the pro-inflammatory immune response of BCG-infected macrophages in a

27

synergistic fashion, as evident by increased production of cytokines and induction of nitric oxide

28

synthesis. Using macrophages from mice deficient in key signaling molecules involved in the

29

pathogen recognition response, we identified combined activation of MyD88- and TRIF-

30

dependent TLR signaling pathways to be essential for the synergistic effect between BCG and

31

NP. Moreover, synergy was strongly dependent on the order of the two stimuli, with TLR

32

activation by BCG functioning as the priming event for the subsequent pIC-NP-stimulus, which

33

acted through an auto-/paracrine type I interferon (IFN) feedback loop. Our results provide a

34

foundation for a promising new approach to enhance BCG-vaccine immunogenicity by co-

35

stimulation with NPs. They also contribute to a molecular understanding of the observed

36

synergistic interaction between the pIC-NPs and BCG-vaccine.

ACS Paragon Plus Environment

Page 2 of 46

Page 3 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

37 38

Molecular Pharmaceutics

KEYWORDS: adjuvant, Bacille Calmette-Guérin (BCG), chitosan, nanoparticles, poly(I:C), Toll-like receptor synergy, type I interferons (IFNs)

39

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

40

INTRODUCTION

41

Tuberculosis (TB), which is caused by the bacterial pathogen Mycobacterium tuberculosis

42

(M.tb), is one of the most deadly infectious diseases globally. In pulmonary TB, which is the

43

most common form of the disease, M.tb infects host macrophages and dendritic cells in the lung

44

and resides intracellularly within maturation arrested, immature phagosomes. M.tb has developed

45

a plethora of strategies to evade the host immune system including interference with dendritic

46

cell (DC) maturation, antigen presentation and migration, resulting in a delayed and suboptimal

47

adaptive immune response.1 Treatment of TB requires daily intake of multiple antibiotics over

48

several months, which is expensive and challenged by patient non-compliance, M.tb drug

49

tolerance and resistance. Therefore, prophylactic vaccination against TB remains a high priority

50

in order to reduce the rate of infection.

51

The live attenuated vaccine strain Mycobacterium bovis Bacille Calmette-Guérin (BCG)

52

confers some protection against more severe forms of tuberculosis (TB) during childhood, but is

53

largely ineffective against pulmonary TB in adults.2 Despite its limited and variable efficacy in

54

adults, the BCG-vaccine remains to date the only one in use against TB. The BCG-strain is

55

considered to be one of the safest vaccines currently in use, but has retained some undesirable

56

properties from its virulent ancestor, such as interference with professional antigen presenting

57

cell (APC) activation and antigen presentation and DC migration.3-4 These are believed to

58

contribute to the suboptimal induction of T cell-mediated immunity by the BCG-vaccine.5-6 The

59

cell-mediated arm of the immune system, including type I helper CD4+ (TH1) and in particular

60

cytotoxic CD8+ T lymphocytes (CTL) is widely accepted to be very important for conveying

61

protection against intracellular pathogens in general, and M.tb in particular.7

62

Thus, a major challenge for either improving the BCG-vaccine or developing new TB vaccines

63

is to evoke immune responses that are able to generate a strong, broad and long-lived T cell-

ACS Paragon Plus Environment

Page 4 of 46

Page 5 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

64

mediated immunity. For this purpose, several new immune modulating or stimulating agents are

65

currently under investigation as adjuvants for vaccines against TB as well as other diseases.8-9

66

The most potent and promising candidates are agonists of Toll-like receptors (TLRs), which are

67

involved in pathogen recognition by APCs, especially macrophages and dendritic cells. TLRs

68

recognize a set of pathogen associated molecular patterns (PAMPs) and trigger an immune

69

response through two main intracellular signaling pathways, either involving the adaptor

70

molecule MyD88 (all TLRs except TLR3), the adaptor molecule TRIF (TLR3), or both (TLR4).

71

Activation of the MyD88-dependent pathway drives production of pro-inflammatory cytokines,

72

whereas the TRIF-dependent pathway induces the transcription of antiviral proteins and type I

73

interferons (IFN).

74

The TLR3 agonist polyriboinosinic acid-polyribocytidylic acid (poly(I:C)) is a synthetic double-

75

stranded RNA derivate, which induces a robust and strong TH1- and CTL polarized immune

76

response in vivo.10-11 As a consequence, poly(I:C) is considered to be a promising adjuvant

77

candidate for new vaccines against intracellular pathogens such as viruses and invasive bacteria.12

78

However, soluble poly(I:C) is prone to rapid degradation by ribonucleases, which are

79

ubiquitously present in serum and tissues in vivo and higher dosage can cause detrimental side-

80

effects, including fever and anemia.13

81

Nanotechnology has further propelled the development of vaccines by providing delivery

82

platforms for vaccines with multiple advantageous properties. Nano-and micro-particles can

83

protect both immune-stimulatory adjuvants and vaccine antigens against rapid degradation,

84

enhance their cellular uptake and increase their specificity for innate immune cells. Most

85

importantly, these delivery formulations often function as a localized depot for adjuvants and

86

antigens, enabling sustained immune stimulation.14 Nanoparticle- and liposome-based

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

87

formulations in particular have been considered promising with several candidates currently in

88

advanced clinical trials.15-16

89

In recent years, special focus has been put on combining multiple immune-stimulatory

90

adjuvants with delivery platforms to multi-adjuvanted vaccine formulations.17 This strategy holds

91

great potential not only for enhancing the strength, but also for optimizing the quality of the

92

immune responses induced by new vaccine formulations. Different combinations of TLR

93

agonists were shown to act synergistically, whereas others have modulating or even antagonistic

94

effects on immune activation. Especially strong synergistic immune cell activation can be

95

achieved by combining MyD88-dependent and TRIF-dependent TLR-agonists.18 However, due

96

to their potency, especially when combined with nanotechnology delivery systems, it is crucial to

97

understand the mechanisms underlying such adjuvant interactions in order to take advantage of

98

these for multi-adjuvanted vaccines.

99

We have recently shown that complementing the BCG-vaccine with poly(I:C) using

100

nanotechnology methods can efficiently enhance pro-inflammatory activation in mouse primary

101

macrophages.19 Nanocoating of poly(I:C) onto the surface of live BCG bacteria using layer-by-

102

layer (LbL) self-assembly both enhanced the production of pro-inflammatory cytokines and the

103

expression of molecules involved in macrophage – T-cell interactions including CD80, CD86 and

104

CD40. Strikingly, poly(I:C) nanocoated onto BCG, but not poly(I:C) co-delivered in its soluble

105

form along with BCG induced significant production of nitric oxide (NO) and bactericidal

106

activity in macrophages. These results are in accordance with earlier reports by others showing

107

strongly enhanced immunostimulatory potency of nanoparticle (NP) surface-bound poly(I:C)

108

compared to poly(I:C) in its soluble form.20-21 Together, poly(I:C)-nanocoated BCG were able to

109

induce a distinct pro-inflammatory macrophage phenotype including IL-12 production, which is

ACS Paragon Plus Environment

Page 6 of 46

Page 7 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

110

commonly associated with promotion of a strongly TH1-skewed adaptive immune response,

111

involving secretion of IFN-γ.22-23

112

In this study, we developed and characterized poly(I:C)-encapsulating nanoparticles (pIC-NPs)

113

using the simple method of polyelectrolyte complexation between the anionic poly(I:C) and

114

cationic chitosan, a non-toxic, biocompatible and biodegradable natural polymer.24 We tested

115

these pIC-NPs for their ability to induce and amplify a pro-inflammatory phenotype in mouse

116

bone marrow-derived macrophages (BMDM), in conjunction with (separately administered)

117

BCG bacteria. Based on experiments using macrophages derived from transgenic mice deficient

118

in adaptor molecules of central pathogen recognition pathways, we also provide a model for the

119

molecular mechanisms involved, including those responsible for the underlying synergistic

120

interaction between BCG and poly(I:C)-NPs. Our results provide a promising foundation for a

121

new approach that combines the BCG-vaccine with NP-based delivery of the adjuvant poly(I:C)

122

that has the potential to improve the potency of the BCG-vaccine.

123 124

EXPERIMENTAL SECTION

125

Preparation and characterization of pIC-NPs

126

To prepare poly(I:C) encapsulating chitosan-nanoparticles (pIC-NPs), endotoxin-free low

127

molecular chitosan (KiOmedine-CSU®, MW 60,000–120,000; Sigma-Aldrich) was dissolved in

128

1 % acetic acid, diluted with dH2O to a concentration of 2 mg/ml and adjusted to pH 6. An equal

129

volume of 1 mg/ml low molecular weight poly(I:C) (Invivogen) in 0.9 % NaCl was then added

130

dropwise to the chitosan solution under stirring, and the solution was left stirring for 20 min. The

131

pIC-NPs were then collected on a glycerol bed by centrifugation at 10,000 g for 20 min, re-

132

suspended in 0.9 % NaCl and dissociated by water bath sonication for 10 min before use. All

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

133

reagents used for pIC-NP production were certified endotoxin-free by the manufacturers, and all

134

steps of the procedure were done in a laminar flow hood to ensure aseptic conditions. Red

135

fluorescent pIC-NPs and soluble red-fluorescent poly(I:C) were produced by supplementing the

136

poly(I:C)-solution with rhodamine-labeled poly(I:C) (Invivogen) in a ratio of 50:1. Dual-labelled

137

fluorescent pIC-NPs were prepared by using red-fluorescent poly(I:C) in combination with

138

chitosan conjugated with near-infrared fluorochrome IRDye® 680RD (LI-COR Biosciences).

139

NPs encapsulating 15 nm gold particles in their matrix (Au-pIC-NP) were produced as follows:

140

15 nm colloidal gold particles were prepared as described previously.25 Unconjugated 15 nm

141

colloidal gold particles were diluted in 2 mg/ml chitosan and concentrated 10 times by

142

centrifugation at 17,000 g for 30 min. Au-pIC-NPs were then prepared using the chitosan-gold

143

particle solution as described above. Chitosan nanoparticles without poly(I:C) were produced by

144

replacing poly(I:C) with 1 mg/ml sodium tripolyphosphate (TPP; Sigma-Aldrich).

145

Hydrodynamic size and the ζ-potential, representing the surface charge of the particles, were

146

measured on a Nano ZS Zeta-sizer instrument (Malvern Instruments). Encapsulation efficiency

147

and the amount of poly(I:C) encapsulated in pIC-NPs was determined indirectly by measuring

148

unbound poly(I:C) after NP collection based on the absorbance of soluble poly(I:C) at 260 nm

149

using a Nanodrop (Thermo Fisher Scientific). Likewise, the amount of chitosan in pIC-NPs was

150

calculated indirectly from the amount of unbound chitosan in the supernatant. Free chitosan was

151

measured using a fluorescamine assay.26 Throughout the article, the indicated concentrations of

152

pIC-NPs refer to the concentration of poly(I:C) in form of pIC-NPs.

153

The morphology of pIC-NPs was analyzed by transmission electron microscopy (TEM). The

154

pIC-NPs were adsorbed to a formvar-coated TEM grid and briefly stained with 3% uranyl acetate

155

and imaged with a CM100 transmission electron microscope (FEI). Images were recorded with a

156

Quemesa camera using iTEM software (Olympus Soft Imaging Solutions).

ACS Paragon Plus Environment

Page 8 of 46

Page 9 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

157

For the nuclease protection assay, 10 U RNase (RNaseOne™; Promega) was added to a

158

solution of soluble poly(I:C) or pIC-NPs, both at a concentration of 20 µg/ml poly(I:C), and the

159

absorbance at 260 nm was measured at different time points using a spectrophotometer

160

(Eppendorf).

161 162

Bacterial cultures and preparation of BCG for infection

163

Bacille Calmette-Guérin (Pasteur strain) expressing GFP (BCG-GFP; generous gift of Michael

164

Niederweis) were grown in Middlebrook's 7H9 broth medium (BD Biosciences, Heidelberg,

165

Germany) supplemented with 10% OADC, 0.02% glycerol, 0.05% Tween 80 and 50 µg/ml

166

Hygromycin B at 37 °C without agitation. To prepare BCG-GFP cultures for infection of

167

BMDM, bacteria in the exponential growth phase with an optical density (OD600) of 0.6–0.9

168

were harvested by centrifugation at 3,000 g for 10 min, washed twice in 0.9% NaCl solution and

169

resuspended in RPMI-medium supplemented with 10% FCS. The bacterial suspension was then

170

sonicated for 10 min in a bath sonicator and passed 10 times through a 23-gauge needle to break

171

up clumps and to ensure a homogenous suspension of single bacteria.

172 173

Isolation and treatment of Bone Marrow-Derived Macrophages (BMDM)

174

Wild-type C57BL/6 (WT) mice were purchased from Taconic Biosciences and transgenic mice

175

deficient in MyD88 (MyD88-/-), TRIF (TRIF-/-), CARDIF (CARDIF-/-) or the IFNAR 1 chain

176

(IFNAR-/-) were reared at the central mouse facility of the Helmholtz Centre for Infection

177

Research, Braunschweig, and at TWINCORE, Centre for Experimental and Clinical Infection

178

Research, Hannover, Germany. Mice deficient in ASC (ASC-/-) came from the Oslo University

179

Hospital, Rikshospitalet.27 Bone Marrow-Derived Macrophages were prepared as described

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 10 of 46

180

previously.19 Briefly, bone marrow was flushed from the tibias and femurs of 8-12 week old mice

181

and red blood cells were removed by incubation in RBC lysis buffer. Bone marrow cells were

182

then incubated in RPMI-medium supplemented with 10% FCS, 30% L929 conditioned medium

183

and 50 µM β-mercaptoethanol in non-tissue culture treated dishes at 37 °C for 7 days with

184

medium changes after 3 and 6 days. Differentiated BMDM were then detached by incubating the

185

cell monolayer in Ca2+/Mg2+-free PBS at 4 °C and harvested by centrifugation. Throughout all

186

experiments, cells were then maintained in RPMI medium supplemented with 10% FCS, 10%

187

L929-conditioned medium and 50 µM β-mercaptoethanol.

188

For infection with live BCG, WT- and KO-BMDM cultures were incubated with BCG-GFP at

189

a MOI of 10 for 3 h and then washed twice with warm Ca2+/Mg2+-free PBS in order to remove

190

any extracellular bacteria. Uninfected or BCG-infected BMDM were then incubated with

191

medium only, soluble poly(I:C), pIC-NPs or Chi/TPP-NPs at the indicated concentrations for 24

192

h. For short term stimulation, BMDM were infected with BCG for 3 h in the presence of soluble

193

poly(I:C) or pIC-NPs at the indicated concentrations, washed and cultured for further 24 h in cell

194

culture medium.

195

For selective stimulation of TLR2, BMDM were treated with lipomannan or the synthetic

196

triacylated lipopeptide Pam3CSK4 (both Invivogen) at the indicated concentrations and for the

197

indicated time periods.

198

For stimulation experiments with recombinant IFN-β (rIFN-β), WT-BMDM were treated with

199

different concentrations (given in international units (IU) per ml) of mouse rIFN-β (R&D

200

Systems) for 24 h either before or after infection with BCG. Cell culture supernatants were then

201

collected for analysis 24 h after BCG infection. For rIFN-β rescue experiments, BCG-infected

202

WT- and KO-BMDM were treated with 2 µg/ml pIC-NPs alone or in presence of 3000 IU/ml

203

rIFN-β for 24 h.

ACS Paragon Plus Environment

Page 11 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

204

Molecular Pharmaceutics

Flow Cytometry (FACS) Assay

205

To analyze the uptake of poly(I:C) by BMDM, cells were incubated with 10 µg/ml red-

206

fluorescent pIC-NPs or soluble poly(I:C), which corresponds to a concentration of 0.2 µg/ml pure

207

rhodamine-labeled poly(I:C), for the indicated time periods. BMDM cultures were then washed 3

208

times with warm PBS, collected by incubation in Ca2+/Mg2+ free PBS at 4°C, fixed with 2%

209

paraformaldehyde for 15 min and analyzed using a BD FACsCalibur flow cytometer (BD

210

Biosciences).

211

To study the ability of BCG, lipomannan and pIC-NP to enhance the expression of TLR2 and

212

TLR3, BMDM cultures were treated with BCG or lipomannan for 3 h, washed and incubated for

213

additional 24 h in normal medium or they were incubated with 2 µg/ml pIC-NPs for 24 h. Cells

214

were then collected, washed with PBS and treated with a Fc receptor blocking solution (Macs).

215

To analyze TLR2 expression, cells were stained with a PE-conjugated antibody against TLR2

216

(clone CB225, Biolegend) or a matching isotype control using standard protocols for surface

217

staining. In contrast, TLR3 is exclusively expressed intracellularly. To analyze expression of

218

TLR3, which is exclusively located intracellularly, BMDM were fixed with 2%

219

paraformaldehyde after incubation with FC receptor blocking solution, permeabilized with 0.5%

220

Tween20 in PBS/0.5% BSA/2 mM EDTA and stained with a PE-conjugated antibody against

221

TLR3 (clone 11F8, Biolegend) or a matching isotype control. After staining, cells were fixed

222

with 2% paraformaldehyde for 15 min and analyzed using a BD FACsCalibur flow cytometer.

223 224

Confocal microscopy

225

For Confocal microscopy analysis, BMDM were seeded on poly-L-lysine coated glass

226

coverslips. To study pIC-NP uptake, cells were incubated with 10 µg/ml red fluorescent pIC-NPs

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 12 of 46

227

or soluble poly(I:C) for different time periods. For co-localization studies, BMDM cultures were

228

treated with 20 µg/ml dual-labeled pIC-NPs alone or together with BCG-GFP for 3 h, washed 3

229

times with warm Ca2+/Mg2+ free PBS and fixed immediately or after additional 24 h culture

230

with 2% paraformaldehyde for 15 min. Cells were finally imaged with an Olympus FluoView

231

1000 inverted IX81 confocal laser scanning microscope.

232 233

Transmission electron microscopy (TEM) of thin eponsections

234

To label lysosomes, BMDM were pulsed with 5nm colloidal gold for 2 h, washed and

235

incubated in fresh medium for additional 2 h. Cells were then incubated with BCG and 10 µg/ml

236

pIC-NPs encapsulating 15 nm gold (Au-pIC-NPs) for 3 h, washed 3 times with warm

237

Ca2+/Mg2+ free PBS and incubated in normal medium for additional 3 or 24 h. Cells were then

238

fixed with 1% glutaraldehyde in 0.2 M HEPES buffer, pH 7.4, overnight. Cell pellets were

239

embedded in 1% low melting point agarose and post-fixed with 2% osmium tetroxide for 2 h,

240

followed by staining with 2% uranyl acetate for 2 h, dehydration with a graded ethanol series

241

(70%, 80%, 90%, 96%, 100%), progressive infiltration with epoxy resin and finally heat

242

polymerization at 60 °C overnight. Thin sections (70 nm) were cut using an ultramicrotome

243

Ultracut UCT (Leica Microsystems) and a diamond knife (Diatome) and then contrasted with

244

0.2% lead citrate for 15 s. Samples were analyzed with JEM-1400 transmission electron

245

microscope (JEOL). Images were recorded with TemCam-F216 camera and EM-MENU software

246

(TVIPS).

247 248

Nitric oxide and cytokine detection

ACS Paragon Plus Environment

Page 13 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

249

Cell culture medium were collected from WT or KO-BMDM cultures after the indicated

250

treatments, centrifuged at 400 g for 10 min and the cell-free supernatants were used immediately

251

(NO measurement) or frozen at −80 °C until use (cytokine detection). NO production in

252

macrophage cultures was determined by measuring the accumulation of the stable NO metabolite

253

nitrite in the supernatants using the colorimetric Griess assay (Thermo Fisher Scientific)

254

according to the producer's protocol. Absorption at 570 nm was measured with a Wallac Victor2

255

plate reader (Perkin Elmer) and the concentration of nitrite was calculated from a nitrite standard

256

curve. For determination of cytokine production, cell culture supernatants were thawed and

257

multiplex bead array assays were used to measure the levels of IL-1β, IL-6, IL-10, IL-

258

12(p40/p70), GM-CSF, TNF-α (Thermo Fisher Scientific) and the type I IFNs, IFN-α and IFN-β

259

(eBioscience) according to the producer's instructions. Sample were then were analyzed using a

260

Luminex® xMAP platform (BD Biosciences).

261 262

Statistics

263

Multiple groups were compared by using one-way or two-way ANOVA where appropriate and

264

differences between groups were analyzed post hoc by Tuckey's pairwise comparisons. Statistical

265

tests were performed using Minitab 17 (Minitab).

266 267 268

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 14 of 46

269

RESULTS

270 271

Figure 1. Characterization of poly(I:C) encapsulating chitosan nanoparticles (pIC-NPs). (A)

272

Schematic illustration of the preparation process of pIC-NPs. (B) Transmission electron

273

microscopy (TEM) of pIC-NPs; scale bar 200 nm. (C) Nuclease protection assay with soluble

274

poly(I:C) and pIC-NPs. Degradation of poly(I:C) was measured as increase in absorbance at 260

275

nm (hyperchromatic effect). Data are presented as the mean ± SD of 3 independent experiments.

276 277 278

Preparation and characterization of poly(I:C)-encapsulating chitosan nanoparticles (pIC-

279

NPs)

280

Poly(I:C)-encapsulating

nanoparticles

(pIC-NPs)

were

prepared

by

polyelectrolyte

281

complexation, exploiting the ability of the anionic poly(I:C) to physically cross-link the cationic

282

polyelectrolyte chitosan via electrostatic interactions (Figure 1A). Nanoparticles (NPs) formed

283

spontaneously after mixing the polyelectrolytes in aqueous solution and were positively charged

284

(+37.7 ± 1.8 mV) with a hydrodynamic size of 384 ± 46 nm and a polydispersity index (PDI) of

285

0.145 ± 0.06. Poly(I:C) was efficiently incorporated into the pIC-NPs with an encapsulation

286

efficacy of 88.7 ± 2.6 %. By measuring the amount of both the unbound poly(I:C) and chitosan

ACS Paragon Plus Environment

Page 15 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

287

remaining in the supernatant after collection of pIC-NPs, the loading percentage of poly(I:C) in

288

pIC-NPs was determined. Poly(I:C) loading percentage was 58.7 ± 3.1 % based on weight. These

289

results are in accordance with other studies showing very similar characteristics for poly(I:C)-

290

encapsulating chitosan nanoparticles.28 Transmission electron microscopy analysis revealed that

291

pIC-NPs consist of a rather loose irregularly shaped core and fiber-like polymer-structures

292

protruding from the core (Figure 1B). A well-known, major disadvantage of poly(I:C) is its

293

vulnerability to rapid degeneration by ribonucleases, which are ubiquitously present in vivo.29 To

294

test if pIC-NP are able to protect poly(I:C) against ribonuclease degradation, both soluble

295

poly(I:C) and pIC-NPs were exposed to recombinant ribonuclease. Degradation of poly(I:C) was

296

analyzed by measuring the increase in UV-light absorption at 260 nm (hyperchromic effect).30

297

Whereas soluble poly(I:C) showed a rapid increase in absorbance within 2-3 min, indicating

298

degradation, absorbance of pIC-NPs did not increase but remained stable over the course of 60

299

min, indicating full protection of poly(I:C) against nuclease degradation (Figure 1C).

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 16 of 46

300 301

Figure 2. Uptake of soluble poly(I:C) or pIC-NPs and intracellular stability of pIC-NPs in

302

BMDM. (A) Flow cytometry analysis of the uptake of red-fluorescent pIC-NPs in comparison to

303

soluble red-fluorescent poly(I:C) (both 2 µg/ml) by BMDM at different time points. Uptake was

304

measured as percentage of cells positive for red fluorescence. Data are presented as the mean ±

305

SD of 3 independent experiments; *** p < 0.001. (B) Confocal microscopy images of the uptake

306

of red-fluorescent pIC-NPs compared to soluble red-fluorescent poly(I:C) by BMDM after

307

different time periods; scale bars: 20 µm. (C) Confocal microscopy images of dual-labeled pIC-

308

NPs with red-fluorescent poly(I:C) and IRDye680™-conjugated chitosan. (D) Co-localization of

309

poly(I:C) and chitosan directly or 24 h after intracellular uptake by BMDM; scale bars: 5 µm.

310 311 312

Uptake of poly(I:C) by mouse BMDM is enhanced by pIC-NPs

ACS Paragon Plus Environment

Page 17 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

313

NPs are in general preferentially and effectively taken up by APCs usually via the phagocytic

314

pathway or receptor-mediated endocytosis rather than by passive uptake via the fluid phase.31 To

315

investigate if pIC-NPs enhance uptake of poly(I:C) in primary macrophages, we followed the

316

uptake of fluorescently labelled poly(I:C) in soluble form or in pIC-NPs over time by flow

317

cytometry and confocal microscopy. Incorporation of poly(I:C) into NPs greatly increased uptake

318

of poly(I:C) by macrophages compared to soluble poly(I:C) and resulted in different intracellular

319

distribution patterns (Figure 2A and B). After a 12 h and 24 h incubation, pIC-NPs were

320

distinctly concentrated in numerous bright fluorescent foci, whereas foci representing

321

accumulated soluble poly(I:C) were considerably fainter. To study the stability of the poly(I:C)-

322

chitosan interaction in the pIC-NPs within macrophages, we combined rhodamine-labelled

323

poly(I:C) and chitosan conjugated with the near-infrared fluorochrome IRDye® to create dual-

324

labelled pIC-NPs. Using laser confocal microscopy, dual-labelled pIC-NPs revealed essentially

325

complete co-localization of poly(I:C) and chitosan in the NPs, which persisted for at least 24 h

326

after uptake by macrophages (Figure 2C and D).

327

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 18 of 46

328 329

Figure 3. Intracellular location of pIC-NPs and BCG in BMDM subcellular compartments. (A)

330

Confocal images of BMDM infected with green-fluorescent BCG (BCG-GFP) and treated with

331

dual-labeled pIC-NPs for 3 h and cultured for additional 24 h. The outline of the cell nucleus is

332

indicated with a stipulated line in the images; scale bar: 5 µm. (B) Schematic depiction of the

ACS Paragon Plus Environment

Page 19 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

333

preparation of 15 nm gold particle encapsulating pIC-NPs (Au-pIC-NPs). (C- E) Transmission

334

electron micrographs of BMDM at different time points after treatment with BCG and Au-pIC-

335

NPs for 3 h. (C) Uptake of Au-pIC-NPs by BMDM; 15 nm gold particles are indicated by

336

arrowheads. Inlet in (C): example of an Au-pIC-NP; arrowheads indicate 15 nm gold particles

337

within the nanoparticles matrix or closely associated with pIC-NPs; scale bar 200 nm.

338

Intracellular location of BCG and Au-pIC-NPs (labeled in the figure as NP, arrowheads indicate

339

15 nm gold) in BMDM 3 h (D) and 24 h (E) after treatment with Au-pIC-NPs. Areas indicated

340

with boxes in (D) are shown at high magnification (Di and Dii). Thick arrows indicate electron-

341

dense polyphosphate granules in the cytoplasm of BCG. Lysosomal compartments were labeled

342

by pre-loading the cells with 5 nm gold particles (thin arrows) before treatment; scale bars: 1 µm

343

(C, D); 500nm (E).

344 345 346

Intracellular BCG-bacteria and pIC-NPs are located to different compartments of the

347

endocytic pathway in BMDM

348

Aiming to study the interaction of BCG and pIC-NPs on the immunological response, we first

349

investigated if and how pIC-NPs might interact with BCG bacteria intracellularly with respect to

350

subsequent intracellular trafficking. By using dual-labelled pIC-NPs (red and magenta) and

351

fluorescent BCG-bacteria (green) we were able to visualize their intracellular distribution by

352

confocal fluorescence microscopy; which revealed that both were located within the same cells,

353

but without spatial overlap (Figure 3A). However, confocal fluorescence microscopy is limited in

354

terms of magnification and resolution, which makes it challenging to analyze subcellular

355

localization precisely. To circumvent these limitations, we exploited the electrostatic interaction

356

between cationic chitosan and negatively charged, 15 nm-sized gold particles to develop pIC-NPs

357

encapsulating gold particles (Au-pIC-NPs), which could be reliably identified within

358

macrophages by transmission electron microscopy (TEM) (Figure 3B and 3C, inlet). In addition,

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 20 of 46

359

to identify the endosomal compartments, in which Au-pIC-NPs and BCG accumulate, BMDM

360

were pulse-chased with 5 nm sized gold-particles to label late endosomal and lysosomal

361

compartments before they were incubated with Au-pIC-NPs and BCG. Shortly after their uptake,

362

Au-pIC-NPs were predominantly located in 5 nm gold-labelled late endosomes or lysosomes,

363

whereas 24 h after uptake, they were found to be exclusively in these compartments (Figure 3D,

364

Di and Dii). In contrast, 24 h after their uptake the majority of BCG-bacteria were neither found

365

in 5 nm gold containing compartments nor together with Au-pIC-NPs in the same compartment,

366

suggesting that they resided in phagosomes that have failed to fuse with lysosomes (Figure 3E).

367

ACS Paragon Plus Environment

Page 21 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

368

Figure 4. Production of nitric oxide (NO) and cytokines in BMDM co-stimulated with BCG and

369

pIC-NPs or soluble poly(I:C). (A) NO production by BMDM infected with BCG and

370

subsequently incubated with different concentrations of poly(I:C) in soluble form or in pIC-NPs

371

for 24 h. As controls, BMDM were left untreated, infected with BCG only or treated with 20

372

µg/ml poly(I:C) in soluble form or pIC-NPs for 24 h. Production of NO was measured by

373

determining the nitrite levels in the culture supernatants using the Griess assay (B) NO

374

production in BMDM infected with BCG and subsequently incubated for 24 h with 2 µg/ml pIC-

375

NPs or Chi/TPP-NPs, which were produced by replacing poly(I:C) with sodium triphosphate

376

(TPP). The concentration of Chi/TPP-NPs was matched to the chitosan content of the pIC-NPs

377

used. (C) Production of pro- and anti-inflammatory cytokines by BMDM infected with BCG and

378

subsequently incubated with 20 µg/ml poly(I:C) in soluble form or in pIC-NPs for 24 h.

379

Untreated cells, BMDM treated with 20 µg/ml soluble poly(I:C) or pIC-NPs alone, or cells only

380

infected with BCG were used as controls. Data are presented as means ± SD of three (B, C) or

381

four (A) independent experiments; nd: not detected; ns: not significant; * p < 0.05; ** p < 0.01;

382

*** p < 0.001.

383 384 385

BCG and pIC-NPs interact synergistically to stimulate pro-inflammatory activation of

386

BMDM

387

To test if pIC-NPs, or soluble poly(I:C) were able to enhance BCG-induced pro-inflammatory

388

activation of primary mouse macrophages, we measured the accumulation of nitrite, a stable

389

metabolite of nitric oxide (NO), in BMDM culture supernatants. Production of NO is a strong and

390

reliable marker for macrophage pro-inflammatory activation and usually correlates with

391

production of pro-inflammatory cytokines.32 BMDM were first infected with BCG and

392

immediately after treated with pIC-NP or soluble poly(I:C) for 24 h, when culture supernatants

393

were harvested and analyzed. There was a striking synergistic interaction between BCG and pIC-

394

NPs in inducing NO production (Figure 4A). But also simultaneous co-stimulation of BMDM

ACS Paragon Plus Environment

Molecular Pharmaceutics

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Page 22 of 46

395

with pIC-NPs and BCG for only a short period (3 h), followed by wash-out and 24 h culture in

396

the absence of pIC-NPs resulted in synergistic induction of significant NO levels (Supplementary

397

Figure S1A). Neither BCG nor pIC-NPs alone, even at the highest concentration, were able to

398

trigger significant NO production. Soluble poly(I:C) used at the same concentrations as pIC-NPs

399

likewise failed to induce any significant NO production in BCG-infected macrophages. Chitosan

400

itself had no effect on NO production, as neither chitosan alone nor in combination with BCG

401

induced NO production (data not shown). In agreement with this, poly(I:C) proved to be the

402

essential component of pIC-NPs as chitosan nanoparticles produced using the crosslinker sodium

403

triphosphate (TPP) instead of poly(I:C) were unable to induce NO production in WT

404

macrophages in conjunction with BCG (Figure 4B).

405

In a similar manner, pIC-NPs synergistically enhanced the production of the pro-inflammatory

406

cytokines TNF-α, IL-12(p40/p70), IL-6, IL-1β and GM-CSF in BCG-infected macrophages

407

(Figure 4C). Although soluble poly(I:C) also was able to increase the levels of some cytokines

408

(IL-6 and GM-CSF), it did so to a much lesser degree than pIC-NPs. The only exception was IL-

409

12(p40/p70), whose levels in BCG-infected macrophages were elevated by both soluble poly(I:C)

410

and pIC-NPs to a similar extent. Neither soluble poly(I:C) nor pIC-NPs alone induced any

411

significant cytokine production. The production of the anti-inflammatory cytokine IL-10 was

412

likewise enhanced by pIC-NPs in combination with BCG, although at a low level. Like the

413

induction of NO, already a short period of co-stimulation with pIC-NPs was sufficient to

414

significantly enhance cytokine production in BCG-infected macrophages (Supplementary Figure

415

S1B). Compared with short co-treatment for 3 h, stimulation with pIC-NPs for 24 h after BCG

416

infection further increased levels of TNF-α, IL-6 and IL-1β substantially, whereas the levels of

417

IL-12(p40/p70), GM-CSF and IL-10 remained unchanged (Supplementary Figure S1C). In

ACS Paragon Plus Environment

Page 23 of 46

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33 34 35 36 37 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60

Molecular Pharmaceutics

418

conclusion, the synergistic interaction between BCG and pIC-NPs, seen for NO production, was

419

also reflected in the enhanced production of pro-inflammatory cytokines.

420

421 422

Figure 5. Role of TLR- and non-TLR signaling pathways in synergistic induction of NO

423

production and enhancement of cytokine secretion in BMDM by pIC-NPs and BCG. (A) NO

424

production in BCG-infected WT and the indicated KO-BMDM macrophages in response to

425

stimulation with 2 µg/ml pIC-NPs for 24 h. (B) Secretion of pro-inflammatory cytokines in BCG-

426

infected WT- and the KO-BMDM stimulated with 2 µg/ml pIC-NPs for 24 h. Different letters

427

indicate statistically significant differences between groups (p ≤ 0.05). (C) NO production in WT-

428

BMDM after co-stimulation with 2 µg/ml pIC-NPs and different concentrations of lipomannan

429

for 24 h compared to co-stimulation with 2 µg/ml soluble poly(I:C) or to lipomannan treatment

430

alone. Data represent means ± SD of three (C) or four (A, B) independent experiments; ** p