JVI Accepts, published online ahead of print on 13 August 2014 J. Virol. doi:10.1128/JVI.02107-14 Copyright © 2014, American Society for Microbiology. All Rights Reserved.

1

Inhibition of breast cancer cell proliferation through disturbance of the

2

Calcineurin/NFAT pathway by Human Herpesvirus-6B U54 tegument

3

protein

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

Mathieu Iampietroa, Annie Gravela, Louis Flamanda,b #

a

Division of infectious and immune diseases, CHU de Quebec Research Center, Quebec

city, Canada; b Department of Microbiology, Infectious Diseases and Immunology, Faculty of Medicine, Laval University, Quebec city, Canada.

Running Title: HHV-6B U54 protein inhibits breast cancer progression # Address correspondence to:

Louis Flamand PhD MBA Room T1-49 Division of infectious and immune diseases CHU de Quebec Research Center, Quebec city, Canada G1V 4G2 Tel: 418-525-4444 (x46164) Fax: 418-654-2765 Email:[email protected]

Abstract: 69 words Text: 1029 words

38

1

39

ABSTRACT

40

Nuclear factor of activated T cell (NFAT) proteins are key regulators involved in multiple

41

physiological mechanisms such as immune response or cell growth. Selective calcineurin /NFAT

42

inhibitors already demonstrated their capacity to decrease NFAT-dependent cancer cell

43

progression, particularly in breast cancer. In this study, we report a role for the human

44

herpesvirus 6B (HHV-6B) U54 tegument protein in inhibiting MCF-7 breast cancer cell line

45

proliferation by inhibiting NFAT activation.

46

RESULTS

47

The NFAT transcription factors include five elements (NFAT1-NFAT5) (4, 12) in which

48

four of them are regulated by Ca2+ (NFAT1-NFAT4) (6, 7). In resting cells, NFAT proteins are

49

hyperphosphorylated and remain cytoplasmic. Upon cell stimulation and rises in intracellular

50

Ca2+ concentrations, the Ca2+/calmodulin-dependent serine phosphatase calcineurin (CaN) (10)

51

becomes active, recruits and dephosphorylates NFAT exposing a nuclear localization signal

52

allowing NFAT to migrate to the nucleus and enhance the expression of several genes (5, 13, 27,

53

29).

54

Disorders of CaN/NFAT pathway cause disturbances in adaptative immune responses,

55

cell differentiation or cell proliferation (15). Selective inhibitors known to prevent CaN/NFAT

56

interaction (30) and subsequent NFAT activation include drugs such as cyclosporine A (CsA)

57

(21) and FK506 (Tacrolimus) (9), inhibitory peptides (1, 2) or proteins expressed by pathogens

58

such as A238L protein of African swine fever virus (24, 25). Recently, Iampietro et al identified

59

the HHV-6B U54 tegument protein as being capable of inhibiting NFAT activation and

60

subsequent IL-2 gene expression, pointing out for a role of the U54 protein in immune evasion

2

61

(17). Considering that the functions of NFAT extend beyond the development of the adaptive

62

immune response, we evaluated the effects of U54 expression of in the proliferation of cells

63

whose growth is NFAT-dependent.

64

Breast cancer is the leading cause of cancer death in women worldwide (16), and is

65

caused by disturbance of NFAT activity (18, 32) by promoting cell transformation, proliferation,

66

invasion and tumor angiogenesis (20, 23). Relative expression of NFAT members in MCF-7 cells

67

is presented under table 1. We used the MCF-7 breast cancer cell line to test the possible

68

inhibitory effects of HHV-6B U54 protein on cell proliferation. To achieve this goal, MCF-7

69

cells (2x105) were transfected with expression vectors 4TO, 4TO-U54 (encoding WT U54), 4TO-

70

U54mut (IT296-297AA mutant with reduced NFAT inhibitory potential), 4TO-U11 (encoding

71

WT U11) and NFAT-Luc reporter plasmids, as described in Iampietro et al (17). After 48h, cells

72

were stimulated or not with TPA 25 ng/ml and ionomycin 0.5μM (TPA/ionomycin) to induce

73

CaN/NFAT pathway for an additional 24h. Luciferase activity was determined and normalized

74

with protein content (n=4) as described in Iampietro et al (17). Treatment with TPA/ionomycin

75

activated endogenous NFAT resulting in a 5x fold increase (P

76

while in cells expressing U54 showed a 70% reduction in luciferase activity (P

77

treated with CsA 5μg/ml were used as positive control. Expression of U54mut or U11, a second

78

HHV-6 tegument protein, had marginal effects on reporter activity. Protein expression was

79

monitored by western blot analysis with beta-actin as loading controls. Next, we wanted to

80

determine whether the U54 inhibitory activity would translate in a physiological effect such as

81

reduced cell growth. We transfected 293T and MCF-7 cells (1x105) with the same plasmids as

82

above and cultured the cells for 96h. CsA or FK506 5 μg/ml were used as positive inhibitory

83

controls. Transfection efficiencies were determined for several wells (n=6) using a GFP reporter

0.0001) in luciferase activity 0.0001). Cells

3

84

vector and found to be equivalent (not shown). Cells were counted every 24h for four days using

85

an automatic cellometer auto T4 cell counter (Nexcelcom, Lawrence, MA). After 72h and 96h,

86

4TO-transfected MCF-7 cells grew approximately 4.5x and 7.5x, respectively (P

87

(figure 2A). Cells transfected with 4TO-U54mut or 4TO-U11 showed proliferation equivalent as

88

4TO control cells. In contrast, at 72 and 96h post-transfection, MCF-7 proliferation was

89

significantly inhibited by U54 (figure 2A). Similar results were obtained with FK-506 (figure

90

2B). 293T cells, which do not rely on NFAT for proliferation (used as controls), were not

91

affected by CsA or U54 expression (figure 2C). Protein expression was monitored by western

92

blot analysis. We next determined how the U54 protein would cause NFAT inactivation, leading

93

to cell growth inhibition. To highlight this mechanism, we analyzed the phosphorylation status of

94

ectopically expressed NFAT1 detected with an antibody detecting the hyperphosphorylated forms

95

(140 kDa) of NFAT1. MCF-7 cells (1,5x105) were transfected with 4TO, 4TO-U54, 4TO-U11,

96

4TO-U54mut and REP-NFAT1 plasmids. After 48h, cells were stimulated or not with

97

TPA/ionomycin for 10 min. Cells pretreated with CsA 10 μg/ml were used as a positive control

98

for inhibition of NFAT dephosphorylation. As shown in figure 3, under resting condition,

99

NFAT1

was

hyperphosphorylated,

as

expected.

In

control-transfected

0.001) (n=4)

cells

(4TO),

100

TPA/ionomycin induced the dephosphorylation of NFAT1, as U11 and U54mut expressing cells.

101

In contrast, in the presence of U54 or CsA, NFAT1 remained hyperphosphorylated (figure 3A).

102

Densitometric analyses were used to quantify the relative levels of NFAT1 phosphorylation

103

(figure 3B).

104

Lastly, we studied whether U54 would impact the expression of a gene, such as COX-2,

105

whose expression partly depends on NFAT and whose role in cancer progression is well

106

documented (8, 28, 31). Under the same conditions tested in figure 3, we evaluated COX-2

4

107

mRNA levels following a 24h stimulation with TPA/ionomycin. A 12-14x fold increase in COX-

108

2 mRNA was recorded in 4TO, U11 and U54mut transfected cells (P

109

expressing cells or cells treated with CsA showed significantly reduced COX-2 mRNA. (P

110

0.0001) (n=4) (figure 4A). Protein expression was monitored by western blot analysis. These

111

results confirm the capacity of HHV-6B U54 protein to abrogate NFAT activation and

112

subsequent MCF-7 breast cancer cell line proliferation.

0.001) (n=4) while U54

113

In a previous work, we identified HHV-6B U54 tegument protein as a new viral protein

114

inhibiting the CaN/NFAT pathway (17), likely favoring immune escape and enhancing viral

115

infection. In this study, we have extended these observations by highlighting the capacity of U54

116

to inhibit breast cancer cell growth. Cancer immunotherapy protocols have demonstrated some

117

effectiveness against virally-induced disorders (14) such as in EBV post transplant

118

lymphoproliferative disorders (3, 22) but remain a big challenge against solid tumors such as

119

breast cancer. Immunodominant epitopes derived from the HHV-6B U54 protein have been

120

described recently (11, 26). Coupled to our observation that U54 can inhibit cancer cell growth,

121

expression of U54 in breast cancer cells could give a double advantage: first by liming the cancer

122

cell proliferations and second, by “marking” them for anti-U54 CD8 T cell recognition and

123

destruction. However, before such therapy can be envisionned, several more studies are needed

124

to identify a reliable gene therapy approach allowing the specific expression of U54 in breast

125

cancer cells.

126

REFERENCE LIST

127

1.

Aramburu, J., F. Garcia-Cozar, A. Raghavan, H. Okamura, A. Rao, and P. G.

128

Hogan. 1998. Selective inhibition of NFAT activation by a peptide spanning the

129

calcineurin targeting site of NFAT. Mol Cell 1:627-37. 5

130

2.

Aramburu, J., M. B. Yaffe, C. Lopez-Rodriguez, L. C. Cantley, P. G. Hogan, and A.

131

Rao. 1999. Affinity-driven peptide selection of an NFAT inhibitor more selective than

132

cyclosporin A. Science 285:2129-33.

133

3.

134 135

disorders: classification and treatment. Oncologist 13:577-85. 4.

136 137

5.

6.

Feske, S., R. Draeger, H. H. Peter, and A. Rao. 2000. Impaired NFAT regulation and its role in a severe combined immunodeficiency. Immunobiology 202:134-50.

7.

142 143

Crabtree, G. R., and E. N. Olson. 2002. NFAT signaling: choreographing the social lives of cells. Cell 109 Suppl:S67-79.

140 141

Chytil, M., and G. L. Verdine. 1996. The Rel family of eukaryotic transcription factors. Curr Opin Struct Biol 6:91-100.

138 139

Carbone, A., A. Gloghini, and G. Dotti. 2008. EBV-associated lymphoproliferative

Feske, S., J. Giltnane, R. Dolmetsch, L. M. Staudt, and A. Rao. 2001. Gene regulation mediated by calcium signals in T lymphocytes. Nat Immunol 2:316-24.

8.

Flockhart, R. J., B. L. Diffey, P. M. Farr, J. Lloyd, and N. J. Reynolds. 2008. NFAT

144

regulates induction of COX-2 and apoptosis of keratinocytes in response to ultraviolet

145

radiation exposure. FASEB J 22:4218-27.

146

9.

Fruman, D. A., C. B. Klee, B. E. Bierer, and S. J. Burakoff. 1992. Calcineurin

147

phosphatase activity in T lymphocytes is inhibited by FK 506 and cyclosporin A. Proc

148

Natl Acad Sci U S A 89:3686-90.

149

10.

Garcia-Cozar, F. J., H. Okamura, J. F. Aramburu, K. T. Shaw, L. Pelletier, R.

150

Showalter, E. Villafranca, and A. Rao. 1998. Two-site interaction of nuclear factor of

151

activated T cells with activated calcineurin. J Biol Chem 273:23877-83.

152 153

11.

Gerdemann, U., L. Keukens, J. M. Keirnan, U. L. Katari, C. T. Nguyen, A. P. de Pagter, C. A. Ramos, A. Kennedy-Nasser, S. M. Gottschalk, H. E. Heslop, M. K. 6

154

Brenner, C. M. Rooney, and A. M. Leen. Immunotherapeutic strategies to prevent and

155

treat human herpesvirus 6 reactivation after allogeneic stem cell transplantation. Blood

156

121:207-18.

157

12.

Graef, I. A., J. M. Gastier, U. Francke, and G. R. Crabtree. 2001. Evolutionary

158

relationships among Rel domains indicate functional diversification by recombination.

159

Proc Natl Acad Sci U S A 98:5740-5.

160

13.

Graef, I. A., F. Wang, F. Charron, L. Chen, J. Neilson, M. Tessier-Lavigne, and G.

161

R. Crabtree. 2003. Neurotrophins and netrins require calcineurin/NFAT signaling to

162

stimulate outgrowth of embryonic axons. Cell 113:657-70.

163

14.

Heslop, H. E. 2009. How I treat EBV lymphoproliferation. Blood 114:4002-8.

164

15.

Hogan, P. G., L. Chen, J. Nardone, and A. Rao. 2003. Transcriptional regulation by

165 166

calcium, calcineurin, and NFAT. Genes Dev 17:2205-32. 16.

167 168

Hutchinson, L. Breast cancer: challenges, controversies, breakthroughs. Nat Rev Clin Oncol 7:669-70.

17.

Iampietro, M., G. Morissette, A. Gravel, L. Flamand. 2014. Inhibition of Interleukin-2

169

Gene Expression by Human Herpesvirus 6B U54 Tegument Protein. Submitted for

170

publication.

171

18.

Jauliac, S., C. Lopez-Rodriguez, L. M. Shaw, L. F. Brown, A. Rao, and A. Toker.

172

2002. The role of NFAT transcription factors in integrin-mediated carcinoma invasion.

173

Nat Cell Biol 4:540-4.

174

19.

Jaworska, J., A. Gravel, K. Fink, N. Grandvaux, and L. Flamand. 2007. Inhibition of

175

transcription of the beta interferon gene by the human herpesvirus 6 immediate-early 1

176

protein. J Virol 81:5737-48.

7

177

20.

Jiang, K., B. He, L. Lai, Q. Chen, Y. Liu, Q. Guo, and Q. Wang. Cyclosporine A

178

inhibits breast cancer cell growth by downregulating the expression of pyruvate kinase

179

subtype M2. Int J Mol Med 30:302-8.

180

21.

Liu, J., J. D. Farmer, Jr., W. S. Lane, J. Friedman, I. Weissman, and S. L. Schreiber.

181

1991. Calcineurin is a common target of cyclophilin-cyclosporin A and FKBP-FK506

182

complexes. Cell 66:807-15.

183

22.

Lucas, K. G., T. N. Small, G. Heller, B. Dupont, and R. J. O'Reilly. 1996. The

184

development of cellular immunity to Epstein-Barr virus after allogeneic bone marrow

185

transplantation. Blood 87:2594-603.

186

23.

187 188

Mancini, M., and A. Toker. 2009. NFAT proteins: emerging roles in cancer progression. Nat Rev Cancer 9:810-20.

24.

Miskin, J. E., C. C. Abrams, and L. K. Dixon. 2000. African swine fever virus protein

189

A238L interacts with the cellular phosphatase calcineurin via a binding domain similar to

190

that of NFAT. J Virol 74:9412-20.

191

25.

192 193

Miskin, J. E., C. C. Abrams, L. C. Goatley, and L. K. Dixon. 1998. A viral mechanism for inhibition of the cellular phosphatase calcineurin. Science 281:562-5.

26.

Nastke, M. D., A. Becerra, L. Yin, O. Dominguez-Amorocho, L. Gibson, L. J. Stern,

194

and J. M. Calvo-Calle. Human CD4+ T cell response to human herpesvirus 6. J Virol

195

86:4776-92.

196

27.

197 198

Rao, A., C. Luo, and P. G. Hogan. 1997. Transcription factors of the NFAT family: regulation and function. Annu Rev Immunol 15:707-47.

28.

Robida, A. M., K. Xu, M. L. Ellington, and T. J. Murphy. 2000. Cyclosporin A

199

selectively inhibits mitogen-induced cyclooxygenase-2 gene transcription in vascular

200

smooth muscle cells. Mol Pharmacol 58:701-8. 8

201

29.

Serfling, E., F. Berberich-Siebelt, S. Chuvpilo, E. Jankevics, S. Klein-Hessling, T.

202

Twardzik, and A. Avots. 2000. The role of NF-AT transcription factors in T cell

203

activation and differentiation. Biochim Biophys Acta 1498:1-18.

204

30.

205 206

alternatives to CsA and FK506? Cell Commun Signal 7:25. 31.

207 208

Sieber, M., and R. Baumgrass. 2009. Novel inhibitors of the calcineurin/NFATc hub -

Yiu, G. K., and A. Toker. 2006. NFAT induces breast cancer cell invasion by promoting the induction of cyclooxygenase-2. J Biol Chem 281:12210-7.

32.

Yoeli-Lerner, M., G. K. Yiu, I. Rabinovitz, P. Erhardt, S. Jauliac, and A. Toker.

209

2005. Akt blocks breast cancer cell motility and invasion through the transcription factor

210

NFAT. Mol Cell 20:539-50.

211 212 213 214 215 216 217 218 219 220 221 9

222 223 224

LEGENDS

225

Table 1: Relative expression of NFAT transcripts in MCF7 and Jurkat cell lines

226

Expression of NFAT transcripts in MCF7 and Jurkat cell lines was evaluated by RT-PCR assay.

227

MCF7 and Jurkat cells were lysed using Qiazol lysis reagent. RNA was extracted and RT was

228

performed to obtain cDNAs that were tested with specific primers to evaluate expression of

229

mRNAa. Primers used are NFAT1 forward 5’- CGA AGA AGA GCC GAA TGC AC -3’ and

230

NFAT1 reverse 5’- AGA AAC TTC TGC GGC CCT AC -3’, NFAT2 forward 5’- CAC TCC

231

TGC TGC CTT ACA CA -3’ and NFAT2 reverse 5’- AAG ATG CGA GCA TGC GAC TA -3’,

232

NFAT3 forward 5’- CGG CCT CTA AGA GAG GTT GA -3’ and NFAT3 reverse 5’- CCT CCT

233

TTT CCT CCC CGA AC -3’ while primers used for GAPDH are described in Jaworska et al(19).

234

Figure 1: U54 inhibits endogenous NFAT transcriptional activity in MCF-7 cells

235

Transcriptional activity of endogenous-expressed NFAT factors was evaluated in MCF-7 cells by

236

luciferase assay. A) MCF-7 cells were transfected with 4TO, 4TO-U54, 4TO-U11, 4TO-U54mut

237

and NFAT-Luc reporter plasmids. As positive control, we pretreated 4TO condition with CsA.

238

Luciferase activity was determined and normalized to protein content (n=4) Western blot analysis

239

confirmed the expression of proteins of interest for each condition tested. Beta-actin was included

240

as loading control.

241

Figure 2: U54 specifically reduces MCF-7 breast cancer cells growth

242

A-B) MCF-7 cells and –C) 293T cells were transfected with 4TO, 4TO-U54, 4TO-U11 and 4TO-

243

U54mut plasmids. As control, we pretreated 4TO condition with CsA (A-C) or FK506 (B). Cells 10

244

were harvested and evaluated at 24h, 48h, 72h and 96h following transfection by cell counting

245

assay (n=4). Western blot analysis confirmed the expression of proteins of interest for each

246

condition tested. Beta-actin was included as loading control.

247

Figure 3: U54 inhibits dephosphorylation of NFAT1 protein.

248

NFAT1 dephosphorylation was evaluated in MCF-7 cells by Western blot assay. A) MCF-7 cells

249

were transfected with 4TO, 4TO-U54, 4TO-U11, 4TO-U54mut and REP-NFAT1 plasmids. As

250

positive control, 4TO transfected cells we pretreated with CsA. Western blot analyses were

251

performed by blotting each condition with a phospho specific anti-NFAT1 antibody (140 kDa)

252

(upper panel). Western blot analysis confirmed expression of U54, U54mut or U11 proteins

253

(middle panels). Beta-actin was included as loading control. B) Densitometric analysis of P-

254

NFAT1 was performed following western blot analysis. The P-NFAT1 level in resting cells

255

(4TO) was set at 100%. Following TPA/ionomycin stimulation, P-NFAT1 levels were compared

256

to their respective resting control (one representative experiment of three independent

257

experiments).

258

Figure 4: U54 reduces COX-2 gene transcription in MCF-7 cell line

259

A) MCF-7 cells were transfected with 4TO, 4TO-U54, 4TO-U11 and 4TO-U54mut plasmids for

260

48h before being stimulated or not with TPA/ionomycin for an additional 24h. RNA was

261

extracted and COX-2 mRNA levels determined by real-time RT-QPCR. The primers used for

262

COX-2 detection were COX-2 forward 5’- TGC ATT CTT TGC CCA GCA CT -3’ and COX-2

263

reverse 5’- AAA GGC GCA GTT TAC GCT GT -3’. The primers for GAPDH, used for

264

normalization were previously described (19). B) Western blot analysis confirmed the expression

265

of U54 and U54mut proteins. Beta-actin was included as loading control.

11

266 267 268 269 270 271 272 273 274

Table 1: Relative expression of NFAT1, 2, 3 in MCF7 and Jurkat T cells.

275 276

- denotes absence of expression; + denotes basal expression level; ++ denotes moderate

277

expression level; +++ denotes high expression level.

278

12

Figure 2 A

B

!

!

!

!

C

!

!

NFAT pathway by human herpesvirus 6B U54 tegument protein.

Nuclear factor of activated T cell (NFAT) proteins are key regulators involved in multiple physiological mechanisms, such as immune response and cell ...
664KB Sizes 2 Downloads 5 Views