JVI Accepts, published online ahead of print on 26 March 2014 J. Virol. doi:10.1128/JVI.00590-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Coregulatory Interactions between CD8α DCs, LAT, and PD-1 Contribute to higher HSV-1

2

Latency

3 4 5 6

Kevin R. Mott, Sariah J. Allen, Mandana Zandian, and Homayon Ghiasi*

7 8

Center for Neurobiology and Vaccine Development, Ophthalmology Research, Department of

9

Surgery, Cedars-Sinai Burns & Allen Research Institute, CSMC - D2066, 8700 Beverly Blvd.,

10 11 12 13 14

Los Angeles, CA.

*Corresponding author. Center for Neurobiology and Vaccine Development - D2066, Cedars-Sinai Burns and Allen Research Institute, 8700 Beverly Blvd., Los Angeles, CA 90048

15 16

PHONE: (310) 248-8582

17

Email: [email protected]

18 19 20

Running title: Factors contributing to HSV-1 latency.

21

Keywords: dendritic cells, HSV-1, depletion, T cells, exhaustion

22

1

23 24

Summary The latency-associated transcript (LAT) of HSV-1, CD8α+ dendritic cells (DCs), and

25

programmed death-1 (PD-1) have all been implicated in the HSV-1 latency-reactivation cycle.

26

It is not known, however, whether an interaction between LAT and CD8α+ DCs regulates

27

latency and T-cell exhaustion. To address this question, we used LAT(+) and LAT(-)viruses.

28

Depletion of DCs in mice ocularly infected with LAT(+) virus resulted in a reduction in the

29

number of T cells expressing PD-1 in the trigeminal ganglia (TG), whereas depletion of DCs in

30

mice similarly infected with LAT(-) virus did not alter PD-1 expression. CD8α+ DCs, but not

31

CD4+ DCs, infected with LAT(+) virus had higher levels of ICP-0, ICP-4, TK, and PD-L1

32

transcripts than those infected with LAT(-) virus. Coculture of infected bone marrow (BM)

33

derived DCs from wild-type (WT) mice, but not infected DCs from CD8α-/- mice, with WT naive T

34

cells contributed to an increase in PD-1 expression. Transfer of bone marrow from WT mice, but

35

not from CD8α-/- mice, to recipient Rag1-/- mice increased the number of latent viral genomes in

36

reconstituted mice infected with the LAT(+) virus. Collectively, this data indicated that a

37

reduction in latency correlated with a decline in the levels of CD8α+ DCs and PD-1 expression.

38

In summary, our results demonstrate an interaction among LAT, PD-1, and CD11cCD8α+ cells

39

that regulates latency in the TG of HSV-1 infected mice.

40

2

41

Importance

42

Very little is known regarding the inter-relationship of LAT, PD-1, and CD8α+ DCs, and how

43

such interactions might contribute to relative number of latent viral genomes. We have shown here

44

that: (1) In both in vivo and in vitro studies, deficiency of CD8α+ DCs significantly reduced T-cell

45

exhaustion in the presence of LAT(+) virus but not LAT(-) virus; (2) HSV-1 infectivity was

46

significantly lower in the LAT(-)-infected DCs than their LAT(+)-infected counterparts; and (3)

47

Adoptive transfer of bone marrow (BM) from WT but not CD8α-/- mice to recipient Rag1-/- mice

48

restored latency to the WT mice level following infection with LAT(+) virus.

49

These studies point to a key role for CD8α+ DCs in T-cell exhaustion in the presence of LAT,

50

which leads to higher numbers of latent viral genomes. Thus, altering this negative function of

51

CD8α+ DCs can potentially be used to generate a more effective vaccine against HSV infection.

52

3

53 54

INTRODUCTION A characteristic feature of infection with HSV-1 is the ability of the virus to establish

55

latency in sensory neurons of an infected host (1-4). Individuals who have acquired a latent

56

infection are subject to episodic recurrences and serve as permanent carriers who are

57

intermittently infectious (5-7). The recurrences are caused by reactivation of the virus, which

58

results in its transit back to the original site of infection (8, 9). More than 80 HSV-1 genes are

59

expressed in neurons during lytic infection. This expression of HSV-1 genes is drastically

60

curtailed during latency. Indeed, the latency-associated transcript (LAT) is the only gene product

61

consistently detected in abundance during latency in infected mice, rabbits, and humans (1, 2,

62

4, 10, 11).

63

Using LAT(+) and LAT(-) viruses, we recently demonstrated that the presence of LAT

64

leads to the generation of dysfunctional T-cell responses in the trigeminal ganglia (TG) of

65

latently infected mice (12). Both LAT expression and enhanced latency correlated with

66

increased mRNA levels of CD8 and the inhibitory receptor programmed death-1 (PD-1) in the

67

TG. These results suggested that TG that are latently infected with LAT(+) virus contain both

68

more CD8+ T cells and more CD8+ T cells expressing the exhaustion marker, PD-1, than TG

69

that are latently infected with LAT(-) virus. This was confirmed by flow cytometry analyses of

70

expression of CD3/CD8/PD-1, HSV-1 gC, the gB498-505-specific CD8+ T-cell pentamer, IL-2, IFN-

71

γ, and TNF-α. The functional significance of PD-1 and its ligands in HSV-1 latency was

72

indicated by the significantly lower levels of HSV-1 latency in mice that were deficient in PD-1 or

73

PD-L1 as compared to WT mice. The levels of HSV-1 latency were unaffected in PD-L2-deficent

74

mice. We also have shown that latency is enhanced by immunization of infected WT mice with

75

FMS-like tyrosine kinase 3 ligand (Flt3L) DNA, which increases the number of dendritic cells

76

(DCs) (13, 14). Conversely, depletion of DCs was associated with reduced latency. Latency

77

was also significantly lower in infected Flt3L-/- and CD8-/- mice than infected WT mice.

78

Interestingly, however, although immunization of Flt3L-/- mice with Flt3L DNA enhanced latency, 4

79

immunization of CD8-/- mice with Flt3L DNA did not. Transfer experiments using DCs expanded

80

ex vivo in the presence of Flt3L or granulocyte-macrophage colony-stimulating factor (GM-CSF)

81

suggested that enhanced latency was associated with the presence of lymphoid-related

82

(CD11c+CD8α+) DCs, whereas reduced latency was associated with myeloid-related

83

(CD11c+CD8α-) DCs. Modulation of DC numbers by Flt3L DNA immunization or by depletion

84

did not, however, alter acute virus replication in the eye and TG or eye disease in ocularly-

85

infected mice (13, 14). It has been shown that CD8+ T cells infiltrate TG by 3 days post ocular

86

infection and it has been postulated that they act to inhibit HSV-1 reactivation (15). During

87

latency, a subset of CD8+ T cells remain in direct contact with infected neurons (16). These

88

cells can block HSV-1 reactivation from latency in ex vivo cultures of TG from latently-infected

89

mice (15, 16). These results suggest a role for CD8+ T cells in HSV-1 latency, while our

90

published studies indicate an involvement of DCs in the efficiency by which HSV-1 establishes

91

latency (13, 14). Using a combination of knockout mouse and adoptive transfer approaches, we

92

have now demonstrated clearly that CD8α+ DCs, rather than CD8+ T cells, are responsible for

93

enhanced HSV-1 latency (17).

94

Collectively our published studies show that: (1) Mice latently infected with WT HSV-1 have

95

higher levels of LAT RNA, CD8 mRNA and PD-1 mRNA in the TG than in mice depleted of DCs (13,

96

14, 18); (2) HSV-1 latency was significantly lower in PD-1-/- and PD-L1-/- mice as compared to WT

97

mice (12); and (3) The presence of CD11c+CD8α+ directly increased the number of HSV-1 latent

98

viral genomes in mice TG (13, 14). However, very little is known regarding the inter-relationship of

99

LAT, PD-1, and CD8α+ DCs, and how such interactions might contribute to latency. We therefore

100

undertook studies to test the hypothesis that CD11c+CD8α+ DCs in the presence of LAT are

101

harboring more non-productive virus, which contributes to an increase in T-cell exhaustion and,

102

thus, enhances latency. We report here that deficiency of CD8α+ DCs significantly reduced T-cell

103

exhaustion in LAT(+) infected mice but not in LAT(-) infected mice. Additionally, in vitro assays

5

104

revealed that incubation of HSV-1-infected WT DCs, but not similarly infected CD8α- DCs, with T

105

cells isolated from naive WT mice promoted T-cell exhaustion. Furthermore, adoptive transfer of

106

bone marrow (BM) from WT but not CD8α-/- mice to recipient Rag1-/- mice restored latency to the

107

WT mice level following infection with LAT(+) virus. These studies point to a key role for CD8α+

108

DCs in T-cell exhaustion in the presence of LAT, which leads to higher levels of virus.

109

6

110

Materials and Methods

111

Virus and mice. Plaque-purified virulent HSV-1 strains McKrae (WT, LAT(+)) and

112

dLAT2903 (LAT(-)) were grown in rabbit skin (RS) cell monolayers in minimal essential medium

113

(MEM) containing 5% fetal calf serum (FCS), as we described previously (19, 20). Throughout

114

the study, McKrae (wild type) and dLAT2903 viruses are referred to as LAT(+) and LAT(-),

115

respectively. dLAT2903 was derived from McKrae in which both copies of the LAT promoter

116

(one in each viral long repeat) and the first 1,667 nucleotides (nt) of the LAT transcript were

117

deleted (20).

118

WT C57BL/6, C57BL/6-CD8α-/-, C57BL/6-β2m-/-, C57BL/6-DTR, C57BL/6-PD-L1-/-, and

119

C57BL/6-Rag1-/- were used. All animal procedures adhered to the Association for Research in

120

Vision and Ophthalmology (ARVO) statement for the Use of Animals in Ophthalmic and Vision

121

Research and were conducted according to institutional animal care and use guidelines.

122

Ocular infection. Mice were infected ocularly with 2 x 105 PFU per eye of LAT(+) or

123

LAT(-) virus. Each virus was suspended in 2 μl of tissue culture media and administered as an

124

eye drop without prior corneal scarification.

125

DC culture. Six week-old mice were used as a source of bone marrow (BM) for the

126

generation of mouse DCs in culture. BM cells were isolated by flushing femurs with PBS.

127

Pelleted cells were briefly resuspended in red blood lysing buffer (Sigma-Aldrich, St. Louis, MO)

128

to lyse red blood cells and stabilized by adding complete medium (RPMI 1640, 10% fetal bovine

129

serum, 100 U/ml penicillin, 100 μg/ml streptomycin, 2 mM L-glutamine). The cells were

130

centrifuged and resuspended in complete medium supplemented with GM-CSF (100 ng/ml;

131

Peprotech, NJ) to enhance replication of DCs (21). Afterwards, cells were plated in non-tissue

132

culture plastic Petri dishes (1 bone per 10 cm dish) for 5 days at 37oC with CO2. After 5 days,

133

the media was removed and the adherent cells were recovered by incubating them for 5 min at

7

134

37oC with Versene (Invitrogen, San Diego, CA). Cells were washed, counted, and plated onto

135

tissue-culture dishes for use the following day.

136

Infection of DCs. Monolayers of DCs from the different strains of mice described above

137

were infected with 1 PFU/cell of LAT(+) or LAT(-) virus. One hr after infection at 37oC, virus was

138

removed and the infected cells were washed three times with fresh media and infected cells

139

were incubated with growth medium for 24 hr. Infected DCs were used for the following studies:

140

A) To measure differences in the infectivity of WT, β2m-/-, CD8α-/-, or PD-L1-/- DCs following

141

infection with LAT(+) or LAT(-). Infected DCs monolayers were harvested and the presence of

142

gB transcript was determined by qRT-PCR. B) To measure the differences in the infectivity of

143

CD8α+ DCs versus CD4+ DCs, infected DCs were harvested and were separated into CD4+

144

DCs or CD8α+ DCs as described below. The presence of ICP0, ICP4, TK, and PD-L1

145

transcripts in each cell population were determined by qRT-PCR as described below. C) To

146

measure the activation state of DCs following HSV-1 infection, DCs isolated from WT mice were

147

mock-infected or infected with 1 PFU/cell of HSV-1 strain McKrae for 24 or 48 hr. Mock-infected

148

and infected DCs were stained and flow cytometric analyses were performed as described

149

below. D) Finally, to determine if DCs contributed to T cell exhaustion, mock-infected or HSV-1

150

infected DCs from WT or CD8α-/- mice were co-cultured with different ratios of naive T cells from

151

WT mice for 24, 48, 72, or 96 hr. The monolayers were harvested at various times post co-

152

cultivation and stained for flow cytometric analyses as described below.

153

Isolation of CD4+ and CD8α+ DCs. Cultured DCs were grown as above, harvested on

154

day 5 and infected with 1 PFU/cell of LAT(+) or LAT(-) HSV-1 for 24 hr. After 24 hr PI, isolation

155

of CD8α+ and CD4+ DCs was performed using a two-step magnetic cell sorting kit (Kit # 130-

156

091-169 and 130-091-262, Miltenyi Biotec, Auburn CA) per the manufacturer’s protocol. Briefly,

157

harvested cells were directly labeled with CD8 Microbeads and CD8α+ DCs were isolated by

158

positive selection from the enriched DC fraction. The magnetically-labeled CD8α+ DCs were

8

159

retained on a column and eluted after removal of the column from the magnetic field.

160

Subsequently, the remaining enriched DCs that passed through the column were directly

161

labeled with CD4 Microbeads, and CD4+ DCs were isolated by the same method of positive

162

selection in a magnetic field.

163

Adoptive transfer of BM to recipient mice. BM from WT C57BL/6 or CD8α-/- mice

164

was isolated and each recipient Rag1-/- mouse received BM from one donor mouse

165

intravenously (IV). Two weeks post-transfer, the Rag1-/- recipient mice were ocularly infected

166

with LAT(+) virus. TG from infected mice were isolated for detection of LAT RNA and gB DNA

167

on day 30 PI.

168

Depletion of DCs. C57BL/6-DTR mice were depleted of their DCs using 100 ng of

169

diphtheria toxin (DT) in 100 μl of PBS via an IP route 24 hr prior to ocular infection and at days

170

2, 5, 10, and 14 PI. As a negative control, a subset of mice was similarly injected with PBS

171

alone, and is therefore referred to as mock-depleted. Efficiency of DC depletion in corneas and

172

spleens was monitored by flow cytometric analysis before ocular infection and at day 5 PI. After

173

the first depletion, more than 90% of DCs had been depleted.

174

Confocal Microscopy and Image Analysis. DCs isolated from different strains of mice

175

were grown on Lab-Tex chamber slides (Sigma-Alderich) as we described previously (22). Briefly,

176

cells were fixed by incubating slides in methanol for 10 min followed by acetone for 5 min at -20oC.

177

Afterwards, slides were rinsed three times for 5 min each at ambient temperature in PBS containing

178

0.05% v/v Tween-20 (PBS-T). Slides were then blocked for 30 min at ambient temperature in PBS-T

179

containing 1% w/v BSA. Rat anti-CD8α (clone 53-6.7, eBioscience, San Diego, CA), rat anti-CD4

180

(clone Gk1.5, eBioscience), rat anti-CD8β (clone YTS156.7.7, BioLegend), and hamster anti-CD11c

181

(clone HL3, BD Biosciences) were used for IHC. Immunostaining was done using CD11c/CD4,

182

CD11c /CD8α, or CD11c /CD8β antibody combinations and staining for 1 hr at 25°C. After three

183

rinses for 5 min each in PBS, slides were incubated for 1 hr at 25°C with secondary antibodies

9

184

labeled with anti-hamster FITC or anti-Rat TRITC (Invitrogen). Slides were washed three times with

185

PBS, air-dried and mounted with Prolong Gold DAPI mounting media (Invitrogen). Images were

186

captured at 1024 x 1024 pixels (original magnification = 20X) in independent fluorescence channels

187

using a Nikon C1 eclipse inverted confocal microscope.

188

Flow Cytometric Analysis. Infected or mock-infected cells were harvested at various times

189

PI and stained with anti-CD3, anti-CD4, anti-CD8α, anti-PD-1, and anti-CD11c obtained from BD

190

PharMingen (San Diego, CA) and BioLegend (San Diego, CA), and then analyzed by flow cytometry

191

as we previously described (23).

192

DNA extraction and PCR analysis for HSV-1 genomic DNA. DNA was isolated from

193

homogenized individual TG using the commercially available DNeasy Blood &Tissue Kit

194

(Qiagen, Stanford, CA) according to the manufacturer's instructions. PCR analyses were done

195

using gB specific primers (Forward - 5'-AACGCGACGCACATCAAG-3'; Reverse - 5'-

196

CTGGTACGCGATCAGAAAGC-3'; and Probe - 5'-FAM-CAGCCGCAGTACTACC-3'). The

197

amplicon length for this primer set was 72 bp. Relative copy numbers for gB DNA were

198

calculated using standard curves generated from the plasmid pAc-gB1 (24). In all experiments,

199

GAPDH was used for normalization of transcripts.

200

RNA Extraction, cDNA Synthesis, and TaqMan RT-PCR. RNA was extracted from

201

latent TG or infected DCs as we have described previously (22, 25, 26). Following RNA

202

extraction, 1 μg of total RNA was reverse-transcribed using random hexamer primers and

203

Murine Leukemia Virus (MuLV) Reverse Transcriptase from the High Capacity cDNA Reverse

204

Transcription Kit (Applied Biosystems, Foster City, CA), in accordance with the manufacturer's

205

recommendations. The levels of various transcripts were evaluated using commercially

206

available TaqMan Gene Expression Assays (Applied Biosystems) with optimized primer and

207

probe concentrations. Primer-probe sets consisted of two unlabeled PCR primers and the

208

FAM

TM

dye-labeled TaqMan MGB probe formulated into a single mixture. Additionally, all

10

209

cellular amplicons included an intron-exon junction to eliminate signal from genomic DNA

210

contamination. The HSV-1 ICP0, ICP4, TK, gB, LAT, and PD-L1 primers and probe used were

211

as follows: 1) ICP0: forward primer, 5'- CGGACACGGAACTGTTCGA-3'; reverse primer, 5'-

212

CGCCCCCGCAACTG-3'; and probe, 5'-FAM-CCCCATCCACGCCCTG-3' -- Amplicon length =

213

111 bp; 2) ICP4: forward primer, 5'- GCGTCGTCGAGGTCGT-3'; reverse primer, 5'-

214

CGCGGAGACGGAGGAG-3'; and probe, 5'-FAM-CACGACCCCGACCACC-3' -- Amplicon

215

length = 69 bp; 3) TK: forward primer, 5'- CAGTAGCGTGGGCATTTTCTG-3'; reverse primer,

216

5'-CCTCGCCGGCAACAAAA-3'; and probe, 5'-FAM-CTCCAGGCGGACTTC-3' -- Amplicon

217

length = 59 bp; 4) gB: forward primer, 5'-AACGCGACGCACATCAAG-3', reverse primer, 5'-

218

CTGGTACGCGATCAGAAAGC-3'; and probe, 5'-FAM-CAGCCGCAGTACTACC-3' – Amplicon

219

length = 72 bp; 5)LAT: forward primer 5'-GGGTGGGCTCGTGTTACAG-3'; reverse primer, 5'-

220

GGACGGGTAAGTAACAGAGTCTCTA-3'; and probe, 5'- FAM-ACACCAGCCCGTTCTTT-3'–

221

Amplicon Length =81 bp, corresponding to LAT nts 119553-119634; and 6) PD-L1- ABI ASSAY

222

I.D. Mm00452054_m1 – Amplicon length = 77 bp. As an internal control, a set of GAPDH

223

primers from Applied Biosystems (ASSAY I.D. m999999.15_G1 - Amplicon Length = 107 bp)

224

was used.

225

Quantitative real-time PCR (qRT-PCR) was performed using an ABI PRISM 7900HT

226

Sequence Detection System (Applied Biosystems, Foster City, CA) in 384-well plates as we

227

described previously (25, 26). Real-time PCR was performed in triplicate for each tissue

228

sample. The threshold cycle (Ct) values, which represents the PCR cycle at which there is a

229

noticeable increase in the reporter fluorescence above baseline, were determined using SDS

230

2.2 Software. GAPDH transcript was used for normalization purposes.

231

Statistical analysis. Student’s t test and chi-squared tests were performed using the

232

computer program Instat (GraphPad, San Diego). Results were considered statistically

233

significant when the P value was < 0.05.

234 11

235

RESULTS

236

DCs play an indispensable role in regulating T-cell exhaustion in mice latently

237

infected with HSV-1. We have shown that depletion of DCs reduces the number of HSV-1

238

latent genomes in ocularly infected mice (13, 14) and that a decrease in latency correlates with

239

a decrease in T-cell exhaustion in the TG of latently infected mice (12). However, very little is

240

known regarding the role of DCs in T-cell exhaustion. As a proof-of-principal experiment, we

241

depleted DCs in diphtheria toxin receptor (DTR) transgenic mice by treatment with DT one day

242

before ocular infection with LAT(+) or LAT(-) HSV-1 virus and at various times post-infection

243

(PI). As controls, DTR mice were mock-depleted of DCs and ocularly infected; DTR mice were

244

both mock-depleted and mock-infected (designated “naive”); and WT C57BL/6 mice were

245

infected with the viruses. Since 30 days PI is accepted as a point in time by which latency has

246

been established (27), the TG were isolated from individual mice at day 30 PI and single-cell

247

suspensions of the TG from each mouse were stained for CD3, CD8, and PD-1. The

248

percentages of CD3+CD8+PD-1+ T cells in the TG of DC-depleted, mock-depleted, and WT mice

249

infected with LAT(+) virus, as well as naive mice are shown in Figure 1A. As would be

250

expected, there were few CD3+CD8+PD-1+ T cells in the TG from naive mice and the highest

251

numbers of CD3+CD8+PD-1+ cells were found in the TG of the WT and mock-depleted DTR

252

control mice. The numbers of CD3+CD8+PD-1+ cells in the TG of the DC-depleted mice was

253

significantly lower than in these two virus-infected controls (Figure 1A). Moreover, the numbers

254

of CD3+CD8+PD-1+ cells in the TG of DC-depleted mice that were infected with LAT(-) virus did

255

not differ significantly from the numbers of CD3+CD8+PD-1+ cells in the TG of mock-depleted

256

controls (Figure 1B). These results are consistent with a previously reported study for the

257

number of CD8-positive T cells in TG of mice infected with LAT(-) virus versus LAT(+) virus (12).

258

We next analyzed the kinetics of CD3/CD8/PD-1 expression during both lytic (days 5 and

259

10) and latent (day 30) in the TG of DC-depleted or mock-depleted DTR mice infected with

260

LAT(+) virus (Figure 1C). During both lytic and latent phases of HSV-1 infection, the numbers of 12

261

CD3+CD8+PD-1+ T cells were significantly lower in the TG of DC-depleted mice than in the TG

262

of the mock-depleted mice. In both groups, the numbers of CD3+CD8+PD-1+ cells increased

263

through day 10 PI and had declined by day 30 PI. Similar to this study, previously it was shown

264

that by 3 days PI, CD8+ T cells begin infiltrating the TG, peaking at 14 days (virus clear by day

265

11 PI), declined significantly after clearance of lytic infection and persisting as latent virus (12,

266

16, 27-29). In addition, in line with our results, previously it was shown that the level of PD-1

267

expression during early stage of human hepatitis C virus (HCV) infection is significantly higher

268

from subjects who progress to chronic HCV infection than from those who clear infection (30).

269

Furthermore, during acute HCV infection, the loss of PD-1 expression by CD8+ T cells was

270

shown to correlate with viral clearance (31). These data support our overall hypothesis that the

271

presence of LAT and DCs affects the level of T-cell exhaustion during both lytic and latent

272

cycles of HSV-1 infection.

273

Expression of CD4 and CD8α transcripts in BM-derived DCs isolated from different

274

strains of mice before HSV-1 infection. Recently, we reported that the absence of CD8α+ DCs

275

correlated with reduced latency (17) . To determine if lower levels of latency associated with a

276

reduction in CD8α+ transcripts, we quantified CD4 and CD8α transcripts in BM-derived DCs isolated

277

from WT, β2m-/-, PD-L1-/-, and CD8α-/- naive mice. The CD4 copy numbers were similar in the DCs

278

from all four strains of mice (Figure 2A, P>0.05). Although the copy numbers of CD8α transcripts in

279

DCs isolated from WT mice were similar to the copy numbers found in β2m-/- mice, the copy numbers

280

of the CD8α transcripts were significantly lower in the DCs isolated from the PD-L1-/- mice (Figure

281

2B, P

Coregulatory interactions among CD8α dendritic cells, the latency-associated transcript, and programmed death 1 contribute to higher levels of herpes simplex virus 1 latency.

The latency-associated transcript (LAT) of herpes simplex virus 1 (HSV-1), CD8α(+) dendritic cells (DCs), and programmed death 1 (PD-1) have all been ...
1MB Sizes 0 Downloads 3 Views