JGV Papers in Press. Published January 6, 2014 as doi:10.1099/vir.0.058313-0

1

Human Cytomegalovirus Suppresses FAS Expression and Function

2 3

Sepehr Seirafian, Virginie Prod’homme, Daniel Sugrue, James Davies, Ceri Fielding, Peter

4

Tomasec and Gavin W. G. Wilkinson*

5

Institute of Infection & Immunity, Cardiff University School of Medicine, Heath Park, Cardiff

6

CF14 4XN, UK.

7 8 9 10 11 12 13 14 15 16 17

Corresponding author: Professor Gavin W.G. Wilkinson Department of Medical Microbiology Cardiff Institute for Infection and Immunity Tenovus Building Cardiff University School of Medicine Cardiff CF14 4XN Email: [email protected] Telephone: 029-20687349

18

Running Title: HCMV downregulates Fas

19

Category: Animal viruses- large DNA

20 21 22

Word Count. Summary: 150 words Main Text: 2164 words

23

Number of Figures: 3

24

Keywords: Human cytomegalovirus, Fas, CD95, APO-1, TNFRSF6

25 26 27

28

Summary

29

Human cytomegalovirus (HCMV) is known to evade extrinsic pro-apoptotic pathways not only

30

by downregulating cells surface expression of the Death Receptors (DRs) TNFR1, TRAIL

31

receptor 1 (TNFRSF10A) and TRAIL receptor 2 (TNFRSF10B), but also by impeding

32

downstream signalling events. Fas (CD95/APO-1/TNFRSF6) also plays a prominent role in

33

apoptotic clearance of virus-infected cells so its fate in HCMV infected cells needed to be

34

addressed. Here, we show that cell surface expression of Fas was suppressed in HCMV-

35

infected fibroblasts from 24 h onwards through the late phase of productive infection, and was

36

dependent on de novo virus-encoded gene expression but not virus DNA replication. Significant

37

levels of the fully glycosylated (endoglycosidase H-resistant) Fas were retained within HCMV

38

infected cells throughout the infection within intracellular membranous structures. HCMV

39

infection provided cells with a high level of protection from Fas-mediated apoptosis.

40

Downregulation of Fas was observed with HCMV strains AD169, FIX, Merlin and TB40.

41

42

Human cytomegalovirus (HCMV), the prototype member of the subfamily Betaherpesviridae, is

43

ubiquitous in human populations worldwide. HCMV establishes a lifelong persistent infection

44

that is normally controlled by continuous host immune surveillance. Although the vast majority of

45

infections in the immunocompetent host appear to be benign, HCMV is a major cause of severe

46

morbidity and mortality following congenital transmission, and in immunocompromised

47

individuals. Studies using murine and rhesus cytomegaloviruses have shown that efficient

48

infection, super-infection and long-term persistence in vivo are dependent on effective viral

49

immune-evasion functions (Babić et al., 2011; Früh et al., 2013; Vidal et al., 2013). HCMV also

50

possesses an impressive array of immunomodulatory functions that are instrumental in avoiding

51

T cells, natural killer (NK) cells, the interferon response and apoptosis.

52

HCMV UL36 (vICA) and MCMV m36 are positional homologues (no overt amino acid sequence

53

homology) that suppress death receptor-mediated apoptosis by inhibiting caspase-8 activation

54

and promoting virulence in vivo respectively (Ebermann et al., 2012; Skaletskaya et al., 2001).

55

In HCMV, cellular DRs are also targeted directly during infection. The laboratory strain AD169

56

downregulates TNFR1 from the cell surface (Baillie et al., 2003) more efficiently than low

57

passage strains (Montag et al., 2006). The inconsistency was explained when UL138 was found

58

to stimulates surface expression of TNFR1; strain AD169 has suffered a deletion of the 15 kb

59

region UL/b’ that encompasses UL138 (Le et al., 2011; Montag et al., 2011). HCMV thus

60

appears to encode functions capable of acting post-transcriptionally to suppress and ‘potentiate’

61

TNFR1 expression. Recently, we demonstrated HCMV also regulates expression of a second

62

DR: TNF-related apoptosis-inducing ligand (TRAIL) receptor (Smith et al., 2013). Although

63

HCMV infection stimulates expression of TRAIL receptor 2 (TR2) in fibroblasts, gpUL141 binds

64

TR2 directly to sequester the DR in the endoplasmic reticulum, thereby protecting HCMV-

65

infected cells against both soluble TRAIL and TRAIL-dependent NK cell-mediated killing

66

(Nemčovičová et al., 2013; Smith et al., 2013).

67

Fas is another member of the tumour necrosis factor receptor superfamily (TNFRSF)

68

recognised to play a major role in controlling virus infections (Itoh et al., 1991; Trauth et al.,

69

1989; Yonehara et al., 1989). While Fas is expressed on most cell types, its cognate ligand

70

(FasL) is restricted to activated T, NK and dendritic cells (Nagata, 1999; Nagata & Golstein,

71

1995). The upregulation of FasL and TRAIL on HCMV-infected dendritic cells promotes direct

72

killing of activated T lymphocytes, an action that may preferentially delete HCMV-specific T cells

73

(Raftery et al., 2001). Moreover, the activation of FasL on HCMV-infected retinal pigment

74

epithelial cells may subvert neutrophil function in HCMV retinitis (Chiou et al., 2001; Cinatl et al.,

75

2000). Although HCMV may exploit FasL to dampen immune responses, FasL has the potential

76

to kill HCMV-infected cells. FasL acts by inducing a conformational change in Fas, leading to

77

recruitment of FADD and procaspase-8, and assembly of the death-inducing signalling complex

78

(DISC) (Kischkel et al., 1995; Scott et al., 2008). Caspase-8 released from DISC induces

79

cleavage of downstream substrates including effector caspases 3 and 7, resulting in proteolysis

80

of critical cellular components and culminating in apoptosis (Barnhart et al., 2003; Salvesen &

81

Dixit, 1997). While UL36 inhibits caspase-8, the fate of Fas during HCMV infection was not clear

82

(Chaudhuri et al., 1999).

83

Human foetal foreskin fibroblasts (HFFF-hTERTs) (McSharry et al., 2001) were therefore

84

infected with HCMV strain Merlin and cell surface expression of Fas tracked over the course of

85

infection. Fas was unaffected by HCMV infection until 24 h p.i.; the cell surface downregulation

86

detected at this time-point persisted through the late phase of infection (48 and 72 h) (Fig. 1a).

87

Consistent with Fas downregulation occurring with early kinetics, cells treated with the viral DNA

88

replication inhibitor Ganciclovir showed comparable levels of Fas downregulation,

89

demonstrating that viral DNA replication is not required for the function (72 h p.i., Fig. 1b).

90

Latent carriage of HCMV has been shown to protect CD34+ progenitor cells from FasL-mediated

91

apoptosis through increased cIL10 secretion (Poole et al., 2011). However, transfer of

92

supernatants from strain Merlin-infected cells did not result in substantial downregulation of Fas

93

at the cell surface, indicating the function is not carried out by a soluble factor (Fig. 1c). In

94

addition, virus inactivated by gamma-irradiation did not modulate Fas expression, thus

95

suggesting the function was attributable to a de novo expressed virus-encoded function rather

96

than input virions (Fig. 1c). MHC class-I was included as an infection control; downregulation of

97

classical MHC class-I expression is achieved by four HCMV genes (US2, US3, US6, US11) that

98

are expressed with immediate early and early kinetics (Ahn et al., 1996; Hengel et al., 1996;

99

Hesse et al., 2013; Lehner & Cresswell, 1996; Park et al., 2002). Downregulation of Fas and

100

MHC class-I exhibit similar kinetics (Fig. 1).

101 102

Since HCMV exhibits an exceptionally high level of inter-strain sequence variation (Dolan et al.,

103

2004), we were interested in determining whether Fas regulation was a conserved function. The

104

level of Fas downregulation was similar in cells infected with HCMV strains Merlin, AD169, FIX

105

and TB40 (Fig. 1d). Comparable results were also obtained using HFFF cells and primary

106

dermal fibroblasts (data not shown). Variation in the efficiency of MHC class-I downregulation

107

was attributable to the fact that strains FIX (ΔUS2, ΔUS3 and ΔUS6) and TB40 (ΔUS3 and

108

ΔUS6) are derived from BAC clones, and were deleted in the US segment to facilitate genome

109

manipulation (Murphy et al., 2003; Sinzger et al., 2008).

110

The sensitivity of HCMV-infected cells to Fas-mediated apoptosis was ascertained by

111

measuring the activation of effector caspases 3 and 7. Cells were infected with HCMV strains

112

Merlin, AD169 or mock-infected and treated with FasL or a cross-linking Fas mAb; soluble TR2

113

or an IgM control antibody. Caspase 3/7 activity was then measured at 16 or 72 h p.i. by their

114

capacity to cleave a luminogenic substrate in presence of a recombinant luciferase (Fig. 2). At

115

16 h p.i., prior to Fas downregulation at the cell surface, there was no significant difference in

116

caspase 3/7 activity between mock-infected and HCMV-infected cells in any of the treatment

117

groups (Fig. 2a). However, at 72 h p.i., cells infected with strains Merlin or AD169 became less

118

sensitive to Fas signalling induced by either FasL or Fas mAb (Fig. 2b). In addition, there was

119

no significant difference in the level of protection imparted on cells by strains Merlin and AD169.

120

This is interesting, since the AD169 variant that was used in this experiment carries a single

121

amino acid substitution in the UL36 gene that abolishes the antiapoptotic function of vICA

122

(Skaletskaya et al., 2001). HCMV infection therefore renders cells less sensitive to Fas-

123

mediated apoptosis. This function correlates with Fas downregulation from the surface of

124

infected cells, and can occur independently of vICA function.

125 126

Fas mRNA levels, as assessed by quantitative reverse transcriptase PCR, were not significantly

127

affected by HCMV infection at 24, 48 or 72 h p.i. (Fig. 3a). Nevertheless, levels of Fas in total

128

cell lysates appeared modestly reduced following infection with HCMV strains Merlin, AD169,

129

Fix or TB40 (Fig. 3b). HCMV is known to suppress the cell surface expression of specific

130

proteins (e.g. CD112, CD155, MHC-I, MICB, TR2, ULBP2) often by sequestering them within

131

the cell (Cosman et al., 2001; Jones et al., 1996; Nemčovičová et al., 2013; Prod'homme et al.,

132

2010; Smith et al., 2013; Tomasec et al., 2005). N-linked glycoproteins acquire resistance to

133

endoglycosidase-H (EndoH) during maturation in the Golgi apparatus. Fas was clearly heavily

134

glycosylated as evidenced by its sensitivity to peptide N-glycosydase-F (PNGaseF), and

135

resistant to EndoH treatment ± HCMV infection (Fig. 3c). Consequently, HCMV does not appear

136

to be retaining newly synthesised Fas in pre-Golgi compartments. Immunofluorescence showed

137

Fas to illuminate the surface of uninfected fibroblasts in addition to a diffuse cytoplasmic

138

staining pattern (Fig. 3d). In cells infected with HCMV, Fas appeared largely excluded from the

139

plasma membrane; rather the protein localised to extended membranous perinuclear structures

140

(Fig. 3d).

141 142

Fas joins the impressive list of immunomodulatory proteins that HCMV downregulates from the

143

cell surface by post-translational regulation; others include MHC class-I, MICA, MICB, ULBP2,

144

CD155, CD112, TR1, TR2 and TNFR1 (Baillie et al., 2003; Dunn et al., 2003; Nemčovičová et

145

al., 2013; Prod'homme et al., 2010; Smith et al., 2013; Stern-Ginossar et al., 2007; Tomasec et

146

al., 2005). While MHC class-I, CD112 and MICA (Fielding, personal communication) are

147

targeted for efficient proteolytic degradation; CD155, TNFR1, TR2 and Fas are maintained at

148

significant levels within infected cells.

149 150

HCMV infection induces resistance to Fas-mediated apoptosis, yet the extent to which this can

151

be attributed to cell surface suppression of Fas will ultimately require the identification of the

152

HCMV gene(s) responsible. Despite a systematic screen of an expression library encoding the

153

canonical HCMV genes, the function responsible has yet to be mapped (Seirafian, 2013). In this

154

context, multiple HCMV genes can be expected to impact Fas signalling. The UL36 and UL37

155

gene products efficiently inhibit Fas-mediated apoptosis by inhibiting capsase-8 activation and

156

cytochrome-c release respectively (Arnoult et al., 2004; Goldmacher et al., 1999; Skaletskaya et

157

al., 2001). Moreover, IE2 is known to upregulate c-FLIP, a protease-deficient procaspase-8

158

homologue (Chiou et al., 2006) whilst the tegument protein UL45 suppresses Fas-mediated

159

killing in the context of HCMV infection by an uncharacterized mechanism (Patrone et al., 2003).

160

These functions operate at or downstream of the DISC, and are thus likely to impact on both

161

TRAIL and Fas mediated signalling to similar degrees. Also, since UL141 downregulation of

162

TR2 had marked impacts on TRAIL-mediated cell death (Smith et al., 2013), it is likely that

163

HCMV downregulation of Fas too is an important component of HCMV immune evasion.

164 165

Autoimmune lymphoproliferative syndrome (ALPS) is a rare disorder characterised by abnormal

166

lymphocyte survival resulting from a defect in Fas function. A study of two brothers with ALPS

167

experiencing HCMV disease following neonatal exposure documented the development of

168

disseminated infections that were eventually controlled (Arkwright et al., 2000). That Fas-

169

mediated apoptosis is not critical for the control of HCMV disease is consistent with the virus

170

having evolved effective countermeasures to evade Fas-mediated killing. The immune evasion

171

functions of HCMV are a realistic target for therapeutic intervention.

172 173

Acknowledgements

174

This work was supported by funds from the Wellcome Trust WT090323MA and MRC

175

G1000236. We are grateful to Victor Goldmacher for advice on setup of the apoptosis assay.

176

References

177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193 194 195 196 197 198 199 200 201 202 203 204 205 206 207 208 209 210 211 212 213 214 215 216 217 218 219 220 221 222

Ahn, K., Angulo, A., Ghazal, P., Peterson, P. A., Yang, Y. & Früh, K. (1996). Human cytomegalovirus

inhibits antigen presentation by a sequential multistep process. Proceedings of the National Academy of Sciences 93, 10990-10995. Arkwright, P., Rieux‐Laucat, F., Le Deist, F., Stevens, R., Angus, B. & Cant, A. (2000). Cytomegalovirus infection in infants with autoimmune lymphoproliferative syndrome (ALPS). Clinical & Experimental Immunology 121, 353-357.

Arnoult, D., Bartle, L. M., Skaletskaya, A., Poncet, D., Zamzami, N., Park, P. U., Sharpe, J., Youle, R. J. & Goldmacher, V. S. (2004). Cytomegalovirus cell death suppressor vMIA blocks Bax-but not Bak-

mediated apoptosis by binding and sequestering Bax at mitochondria. Proceedings of the National Academy of Sciences of the United States of America 101, 7988-7993. Babić, M., Krmpotić, A. & Jonjić, S. (2011). All is fair in virus–host interactions: NK cells and cytomegalovirus. Trends in molecular medicine 17, 677-685. Baillie, J., Sahlender, D. & Sinclair, J. (2003). Human cytomegalovirus infection inhibits tumor necrosis factor alpha (TNF-α) signaling by targeting the 55-kilodalton TNF-α receptor. Journal of virology 77, 7007-7016. Barnhart, B. C., Alappat, E. C. & Peter, M. E. (2003). The CD95 type I/type II model. In Seminars in immunology, pp. 185-193: Elsevier. Chaudhuri, A. R., Jeor, S. S. & Maciejewski, J. P. (1999). Apoptosis induced by human cytomegalovirus infection can be enhanced by cytokines to limit the spread of virus. Experimental hematology 27, 1194-1203.

Chiou, S.‐H., Liu, J.‐H., Hsu, W.‐M., Chen, S. S.‐L., Chang, S.‐Y., Juan, L.‐J., Lin, J.‐C., Yang, Y.‐T., Wong, W.‐W. & Liu, C.‐Y. (2001). Up-regulation of Fas ligand expression by human cytomegalovirus immediate-early gene product 2: a novel mechanism in cytomegalovirus-induced apoptosis in human retina. The Journal of Immunology 167, 4098-4103.

Chiou, S.‐H., Yang, Y.‐P., Lin, J.‐C., Hsu, C.‐H., Jhang, H.‐C., Yang, Y.‐T., Lee, C.‐H., Ho, L. L., Hsu, W.‐M. & Ku, H.‐H. (2006). The immediate early 2 protein of human cytomegalovirus (HCMV) mediates the apoptotic control in HCMV retinitis through up-regulation of the cellular FLICE-inhibitory protein expression. The Journal of Immunology 177, 6199-6206.

Cinatl, J., Blaheta, R., Bittoova, M., Scholz, M., Margraf, S., Vogel, J.‐U., Cinatl, J. & Doerr, H. W. (2000). Decreased neutrophil adhesion to human cytomegalovirus-infected retinal pigment epithelial cells is mediated by virus-induced up-regulation of Fas ligand independent of neutrophil apoptosis. The Journal of Immunology 165, 4405-4413.

Cosman, D., Müllberg, J., Sutherland, C. L., Chin, W., Armitage, R., Fanslow, W., Kubin, M. & Chalupny, N. J. (2001). ULBPs, novel MHC class I–related molecules, bind to CMV glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 14, 123-133. Dolan, A., Cunningham, C., Hector, R. D., Hassan‐Walker, A. F., Lee, L., Addison, C., Dargan, D. J., McGeoch, D. J., Gatherer, D. & Emery, V. C. (2004). Genetic content of wild-type human cytomegalovirus. Journal of General Virology 85, 1301-1312. Dunn, C., Chalupny, N. J., Sutherland, C. L., Dosch, S., Sivakumar, P., Johnson, D. C. & Cosman, D. (2003). Human cytomegalovirus glycoprotein UL16 causes intracellular sequestration of NKG2D ligands, protecting against natural killer cell cytotoxicity. The Journal of experimental medicine 197, 1427-1439.

Ebermann, L., Ruzsics, Z., Guzmán, C. A., van Rooijen, N., Casalegno‐Garduño, R., Koszinowski, U. & Čičin‐Šain, L. (2012). Block of Death-Receptor Apoptosis Protects Mouse Cytomegalovirus from Macrophages and Is a Determinant of Virulence in Immunodeficient Hosts. PLoS pathogens 8, e1003062.

223 224 225 226 227 228 229 230 231 232 233 234 235 236 237 238 239 240 241 242 243 244 245 246 247 248 249 250 251 252 253 254 255 256 257 258 259 260 261 262 263 264 265 266 267 268 269

Früh, K., Malouli, D., Oxford, K. L. & Barry, P. A. (2013). Non-Human-Primate Models of

Cytomegalovirus Infection, Prevention, and Therapy. In Cytomegaloviruses: From Molecular Pathogenesis to Intervention, pp. 463-496. Edited by M. J. Reddehase: Caister Academic Press.

Goldmacher, V. S., Bartle, L. M., Skaletskaya, A., Dionne, C. A., Kedersha, N. L., Vater, C. A., Han, J.‐w., Lutz, R. J., Watanabe, S. & McFarland, E. D. C. (1999). A cytomegalovirus-encoded

mitochondria-localized inhibitor of apoptosis structurally unrelated to Bcl-2. Proceedings of the National Academy of Sciences 96, 12536-12541. Hengel, H., Flohr, T., Hämmerling, G. J., Koszinowski, U. H. & Momburg, F. (1996). Human cytomegalovirus inhibits peptide translocation into the endoplasmic reticulum for MHC class I assembly. Journal of general virology 77, 2287-2296. Hesse, J., Ameres, S., Besold, K., Krauter, S., Moosmann, A. & Plachter, B. (2013). Suppression of CD8+ T-cell recognition in the immediate-early phase of human cytomegalovirus infection. Journal of General Virology 94, 376-386.

Itoh, N., Yonehara, S., Ishii, A., Yonehara, M., Mizushima, S.‐I., Sameshima, M., Hase, A., Seto, Y. & Nagata, S. (1991). The polypeptide encoded by the cDNA for human cell surface antigen Fas can mediate apoptosis. Cell 66, 233-243. Jones, T. R., Wiertz, E., Sun, L., Fish, K. N., Nelson, J. A. & Ploegh, H. L. (1996). Human cytomegalovirus US3 impairs transport and maturation of major histocompatibility complex class I heavy chains. Proceedings of the National Academy of Sciences 93, 11327-11333.

Kischkel, F., Hellbardt, S., Behrmann, I., Germer, M., Pawlita, M., Krammer, P. & Peter, M. (1995).

Cytotoxicity-dependent APO-1 (Fas/CD95)-associated proteins form a death-inducing signaling complex (DISC) with the receptor. The EMBO journal 14, 5579. Le, V. T. K., Trilling, M. & Hengel, H. (2011). The Cytomegaloviral Protein pUL138 Acts as Potentiator of Tumor Necrosis Factor (TNF) Receptor 1 Surface Density To Enhance ULbE-Encoded Modulation of TNF-α Signaling. Journal of virology 85, 13260-13270. Lehner, P. J. & Cresswell, P. (1996). Processing and delivery of peptides presented by MHC class I molecules. Current opinion in immunology 8, 59-67. McSharry, B., Jones, C., Skinner, J., Kipling, D. & Wilkinson, G. (2001). Human telomerase reverse transcriptase-immortalized MRC-5 and HCA2 human fibroblasts are fully permissive for human cytomegalovirus. Journal of General Virology 82, 855-863. Montag, C., Wagner, J., Gruska, I. & Hagemeier, C. (2006). Human cytomegalovirus blocks tumor necrosis factor alpha-and interleukin-1β-mediated NF-κB signaling. Journal of virology 80, 11686-11698. Montag, C., Wagner, J. A., Gruska, I., Vetter, B., Wiebusch, L. & Hagemeier, C. (2011). The LatencyAssociated UL138 Gene Product of Human Cytomegalovirus Sensitizes Cells to Tumor Necrosis Factor Alpha (TNF-α) Signaling by Upregulating TNF-α Receptor 1 Cell Surface Expression. Journal of virology 85, 11409-11421.

Murphy, E., Yu, D., Grimwood, J., Schmutz, J., Dickson, M., Jarvis, M. A., Hahn, G., Nelson, J. A., Myers, R. M. & Shenk, T. E. (2003). Coding potential of laboratory and clinical strains of human cytomegalovirus. Proceedings of the National Academy of Sciences 100, 14976-14981. Nagata, S. (1999). Fas ligand-induced apoptosis. Annual review of genetics 33, 29-55. Nagata, S. & Golstein, P. (1995). The Fas death factor. Science 267, 1449-1456. Nemčovičová, I., Benedict, C. A. & Zajonc, D. M. (2013). Structure of Human Cytomegalovirus UL141 Binding to TRAIL-R2 Reveals Novel, Non-canonical Death Receptor Interactions. PLoS pathogens 9, e1003224. Park, B., Oh, H., Lee, S., Song, Y., Shin, J., Sung, Y. C., Hwang, S.‐Y. & Ahn, K. (2002). The MHC class I homolog of human cytomegalovirus is resistant to down-regulation mediated by the unique

270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285 286 287 288 289 290 291 292 293 294 295 296 297 298 299 300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316

short region protein (US) 2, US3, US6, and US11 gene products. The Journal of Immunology 168, 3464-3469.

Patrone, M., Percivalle, E., Secchi, M., Fiorina, L., Pedrali‐Noy, G., Zoppé, M., Baldanti, F., Hahn, G., Koszinowski, U. H. & Milanesi, G. (2003). The human cytomegalovirus UL45 gene product is a

late, virion-associated protein and influences virus growth at low multiplicities of infection. Journal of general virology 84, 3359-3370. Poole, E., Dallas, S. R. M., Colston, J., Joseph, R. S. V. & Sinclair, J. (2011). Virally induced changes in cellular microRNAs maintain latency of human cytomegalovirus in CD34+ progenitors. Journal of General Virology 92, 1539-1549.

Prod'homme, V., Sugrue, D. M., Stanton, R. J., Nomoto, A., Davies, J., Rickards, C. R., Cochrane, D., Moore, M., Wilkinson, G. W. & Tomasec, P. (2010). Human cytomegalovirus UL141 promotes

efficient downregulation of the natural killer cell activating ligand CD112. Journal of General Virology 91, 2034-2039. Raftery, M. J., Schwab, M., Eibert, S. M., Samstag, Y., Walczak, H. & Schönrich, G. (2001). Targeting the function of mature dendritic cells by human cytomegalovirus: a multilayered viral defense strategy. Immunity 15, 997-1009. Salvesen, G. S. & Dixit, V. M. (1997). Caspases: intracellular signaling by proteolysis. Cell 91, 443. Scott, F. L., Stec, B., Cristina Pop, M. & lstrok (2008). The Fas–FADD death domain complex structure unravels signalling by receptor clustering. Nature 457, 1019-1022. Seirafian, S. (2013). An analysis of Human Cytomegalovirus gene usage: Cardiff University, PhD thesis.

Sinzger, C., Hahn, G., Digel, M., Katona, R., Sampaio, K. L., Messerle, M., Hengel, H., Koszinowski, U., Brune, W. & Adler, B. (2008). Cloning and sequencing of a highly productive, endotheliotropic virus strain derived from human cytomegalovirus TB40/E. Journal of General Virology 89, 359368.

Skaletskaya, A., Bartle, L. M., Chittenden, T., McCormick, A. L., Mocarski, E. S. & Goldmacher, V. S. (2001). A cytomegalovirus-encoded inhibitor of apoptosis that suppresses caspase-8 activation. Proceedings of the National Academy of Sciences 98, 7829-7834. Smith, W., Tomasec, P., Aicheler, R., Loewendorf, A., Nemčovičová, I., Wang, E. C., Stanton, R. J., Macauley, M., Norris, P. & Willen, L. (2013). Human Cytomegalovirus Glycoprotein UL141 Targets the TRAIL Death Receptors to Thwart Host Innate Antiviral Defenses. Cell Host & Microbe 13, 324-335.

Stern‐Ginossar, N., Elefant, N., Zimmermann, A., Wolf, D. G., Saleh, N., Biton, M., Horwitz, E., Prokocimer, Z., Prichard, M. & Hahn, G. (2007). Host immune system gene targeting by a viral miRNA. Science Signaling 317, 376. Tomasec, P., Wang, E. C., Davison, A. J., Vojtesek, B., Armstrong, M., Griffin, C., McSharry, B. P., Morris, R. J., Llewellyn‐Lacey, S. & Rickards, C. (2005). Downregulation of natural killer cell– activating ligand CD155 by human cytomegalovirus UL141. Nature immunology 6, 181-188. Trauth, B. C., Klas, C., Peters, A. M., Matzku, S., Möller, P., Falk, W., Debatin, K.‐M. & Krammer, P. H. (1989). Monoclonal antibody-mediated tumor regression by induction of apoptosis. Science (New York, NY) 245, 301-305. Vidal, S., Krmpotić, A., Pyzik, M. & Jonjić, S. (2013). Innate Immunity to Cytomegalovirus in the Murine

Model. In Cytomegaloviruses: From Molecular Pathogenesis to Intervention, pp. 192-214 Edited by M. J. Reddehase: Caister Academic Press. Yonehara, S., Ishii, A. & Yonehara, M. (1989). A cell-killing monoclonal antibody (anti-Fas) to a cell surface antigen co-downregulated with the receptor of tumor necrosis factor. The Journal of experimental medicine 169, 1747-1756.

317

Figure Legends

318

Fig. 1. Modulation of Fas cell surface expression in cells infected with HCMV

319

a) HFFF-hTERTs were infected with HCMV strain Merlin (MOI=10) or mock-infected and

320

analysed at indicated time-points by flow cytometry for cell surface Fas expression (R&D

321

Systems, mab142, n= at least 3). b) HFFFs were infected with HCMV strain Merlin (MOI=10, 72

322

h) in the presence or absence of 100µM Ganciclovir and analysed by flow cytometry for cell

323

surface Fas expression (n= 3). c)HFFF-hTERTs were incubated with supernatants (sn) of strain

324

Merlin-infected cells (MOI=10, 72 h p.i.) from which virions had been removed using a 0.1µm

325

filter, or were infected with HCMV strain Merlin (MOI=10, 72 h) or equivalent γ-irradiated

326

preparation (2500 Gγ) and analysed by flow cytometry for cell surface Fas expression (n=3). d)

327

HFFF-hTERTs were infected with HCMV strains Merlin, AD169, FIX or TB40 (MOI=10, 72 h)

328

and analysed by flow cytometry for cell surface Fas expression (n= at least 3). Control IgG

329

staining is shown in black and grey lines for mock-infected and HCMV-infected cells

330

respectively.

331 332

Fig. 2. HCMV infection renders cells less sensitive to Fas-mediated apoptosis

333

HFFFs were infected with strain Merlin or AD169 (MOI=10), or mock-infected. At 4 (a) or 60 (b)

334

h p.i. cells were treated with cycloheximide (Sigma, C6255) at 10 µg/ml concentration, and:

335

FasL (IBA-Lifesciences, 23901002), Fas mAb (Beckman-Coulter, IM2387), sTRAIL-R2 (control

336

for Fas ligand) or IgM isotype control. Apoptosis was then measured at indicated time-points as

337

caspase 3/7 activation using the Caspase-Glo 3/7 kit (Promega, G8091). Results are presented

338

as mean relative light units (RLU) ± SEM (n=4). P values were calculated using a one-way

339

ANOVA test and a Bonferroni post test.

340 341

Fig. 3. Analysis of Fas expression in HCMV-infected cells

342

a) HFFF cells were infected with strain Merlin (MOI=10) or mock-infected and Fas mRNA levels

343

analysed at indicated times by qRT-PCR: total cell RNA was extracted (Qiagen) followed by RT-

344

PCR using random hexamer primers (Amersham Biosciences). Resulting cDNA was analysed

345

by qPCR using primers specific to Fas or GAPDH and SYBR green dye (applied Biosciences).

346

Relative quantity (RQ) values were calculated by the comparative CT method. Fas RQ values

347

are shown normalised to GAPDH and relative to the ‘24 h p.i.’ mock sample (± SEM, n=3). P

348

values were calculated using a one-way ANOVA test and a Bonferroni post test. b) HFFF-

349

hTERT cells were infected with strains Merlin, AD169, FIX and TB40 (MOI=10, 72 h) or mock-

350

infected. Total cell lysates were analysed by western blot (Abcam, ab133619, n= at least 3). c)

351

Cell lysates equivalent to b) were treated with EndoH (NEB, P0702) or PNGaseF (NEB, P0704)

352

and analysed by western blot (n= at least 3). d) HFFF-hTERT cells were infected with strains

353

Merlin or AD169 (MOI=10, 72 h) or mock-infected and Fas expression visualised by

354

immunofluorescence (R&D Systems, mab142, n= at least 3, shown in red in top panel). Outlines

355

of cells were visualised with phalloidin-AF488 (phall, Invitrogen, A12379) and are overlaid with

356

Fas staining in bottom panel. Scale bars correspond to 10µm.

357 358

Fas

a

MHC class-I

Fas

2h

MHC class-I

count

count

+Ganciclovir

6h count

102103104105106

102103104105106

c count

Merlin/Merlin sn

count

12 h

count

γ-irradiated

count

18 h

102103104105106

102103104105106

d Merlin

count

count

24 h

AD169

count

count

48 h

FIX

count

count

72 h

TB40 102103104105106

count

102103104105106

b count

-Ganciclovir

102103104105106

102103104105106

102103104105106

cIgG cIgG cIgG

102103104105106

Mock +HCMV +HCMV supernatant

Fig. 1. Modulation of Fas cell surface expression in cells infected with HCMV a) HFFF-hTERTs were infected with HCMV strain Merlin (MOI=10) or mock-infected and analysed at indicated timepoints by flow cytometry for cell surface Fas expression (R&D Systems, mab142, n= at least 3). b) HFFFs were infected with HCMV strain Merlin (MOI=10, 72 h) in the presence or absence of 100µM Ganciclovir and analysed by flow cytometry for cell surface Fas expression (n= 3). c)HFFF-hTERTs were incubated with supernatants (sn) of strain Merlin-infected cells (MOI=10, 72 h p.i.) from which virions had been removed using a 0.1µm filter, or were infected with HCMV strain Merlin (MOI=10, 72 h) or equivalent γ-irradiated preparation (2500 Gγ) and analysed by flow cytometry for cell surface Fas expression (n=3). d) HFFF-hTERTs were infected with HCMV strains Merlin, AD169, FIX or TB40 (MOI=10, 72 h) and analysed by flow cytometry for cell surface Fas expression (n= at least 3). Control IgG staining is shown in black and grey lines for mock-infected and HCMV-infected cells respectively.

a

b

16 h p.i. P>0.05

1.0x105 8.0x104

72 h p.i. 8.0x104

P0.05

4.0x104 2.0x104

0

2.0x104 0

Fig. 2. HCMV infection renders cells less sensitive to Fas-mediated apoptosis

HFFFs were infected with strain Merlin or AD169 (MOI=10), or mock-infected. At 4 (a) or 60 (b) h p.i. cells were treated with cycloheximide (Sigma, C6255) at 10 µg/ml concentration, and: FasL (IBA-Lifesciences, 23901002), Fas mAb (Beckman-Coulter, IM2387), sTRAIL-R2 (control for Fas ligand) or IgM isotype control. Apoptosis was then measured at indicated time-points as caspase 3/7 activation using the Caspase-Glo 3/7 kit (Promega, G8091). Results are presented as mean relative light units (RLU) ± SEM (n=4). P values were calculated using a one-way ANOVA test and a Bonferroni post test.

Mock

a

1.5

b

RQ 0.0

-

+

-

-

+

-

PNGaseF -

-

+

-

-

+

Merlin

Fas

40

0.5

EndoH

TB40

FIX

1.0

AD169

P>0.05

c Merlin

P>0.05

Mock

P>0.05

Merlin

Mock

40 40 24 h

48 h

Fas

GAPDH 30

72 h

d Mock

Merlin

AD169

Isotype CTRL

Fas

Isotype CTRL

Fas

Isotype CTRL

Fas

Isotype + phall

Fas + phall

Isotype + phall

Fas + phall

Isotype + phall

Fas + phall

Fig. 3. Analysis of Fas expression in HCMV-infected cells a) HFFF cells were infected with strain Merlin (MOI=10) or mock-infected and Fas mRNA levels analysed at indicated times by qRT-PCR: total cell RNA was extracted (Qiagen) followed by RT-PCR using random hexamer primers (Amersham Biosciences). Resulting cDNA was analysed by qPCR using primers specific to Fas or GAPDH and SYBR green dye (applied Biosciences). Relative quantity (RQ) values were calculated by the comparative C T method. Fas RQ values are shown normalised to GAPDH and relative to the ‘24 h p.i.’ mock sample (± SEM, n=3). P values were calculated using a one-way ANOVA test and a Bonferroni post test. b) HFFF-hTERT cells were infected with strains Merlin, AD169, FIX and TB40 (MOI=10, 72 h) or mock-infected. Total cell lysates were analysed by western blot (Abcam, ab133619, n= at least 3). c) Cell lysates equivalent to b) were treated with EndoH (NEB, P0702) or PNGaseF (NEB, P0704) and analysed by western blot (n= at least 3). d) HFFF-hTERT cells were infected with strains Merlin or AD169 (MOI=10, 72 h) or mock-infected and Fas expression visualised by immunofluorescence (R&D Systems, mab142, n= at least 3, shown in red in top panel). Outlines of cells were visualised with phalloidinAF488 (phall, Invitrogen, A12379) and are overlaid with Fas staining in bottom panel. Scale bars correspond to 10µm.

Human cytomegalovirus suppresses Fas expression and function.

Human cytomegalovirus (HCMV) is known to evade extrinsic pro-apoptotic pathways not only by downregulating cell surface expression of the death recept...
1022KB Sizes 1 Downloads 0 Views