Articles in PresS. Am J Physiol Cell Physiol (December 17, 2014). doi:10.1152/ajpcell.00321.2014

1

The topoisomerase II catalytic inhibitor ICRF-193 preferentially targets

2

telomeres that are capped by TRF2

3

Lianxiang Chen1#, Xiaowei Zhu1#, Yaru Zou1, Jun Xing1, Eric Gilson2, Yiming Lu1*

4

and Jing Ye1*

5

1

6

Medicine; Pôle Sino-Français de Recherches en Sciences du Vivant et Génomique,

7

Shanghai Ruijin Hospital, Shanghai 200025, China. 2.Institut for Research on Cancer

8

and Aging, Nice (IRCAN), Nice University, CNRS UMR7284/INSERM U1081,

9

Faculty of Medicine, Nice, France; department of Medical Genetics, Archet 2

10

Hospital, CHU of Nice, France; Pôle Sino-Français de Recherches en Sciences du

11

Vivant et Génomique, Shanghai Ruijin Hospital, Shanghai 200025, China

Shanghai Ruijin Hospital affiliated to Shanghai Jiaotong University, School of

12 13

# These authors contributed equally to this work

14 15

Running Title: TOP II inhibitor regulates telomere function

16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 33

*Address correspondence to: Dr. Jing Ye Shanghai Ruijin Hospital Shanghai Jiaotong University School of Medicine 197 Ruijin Second Road (Rm. 1110, Building 11) Luwan District, Shanghai, China Phone: 86-13585625087; Fax: 86-21 64671676; Email: [email protected] or Dr. Yiming Lu Shanghai Ruijin Hospital Shanghai Jiaotong University School of Medicine 197 Ruijin Second Road (Rm. 1110, Building 11) Luwan District, Shanghai, China Phone: 86 21 64335180; Fax: 86 21 64335180; Email: [email protected]

Copyright © 2014 by the American Physiological Society.

34

ABSTRACT

35

The increased level of chromosome instability in cancer cells is not only a driving

36

force for oncogenesis but also can be the Achille’s heel of the disease since many

37

chemotherapies kill cells by inducing a non tolerable rate of DNA damage. A wealth

38

of published evidence showed that telomere stability can be more affected than the

39

bulk of the genome by several conventional anti-neoplasic drugs. In the present study,

40

HT1080 cell lines compromised for either TRF2 or POT1 were treated with ICRF-193

41

(3uM, 24H) or Bleomycin (1uM, 24H). DNA damage was assayed by combining

42

telomeric DNA staining of a (CCCTAA) n PNA probe with immuno-fluorescence of

43

53BP1 in order to score the rate of telomeres colocalizing with 53BP1 foci. We found

44

that ICRF-193, but not bleomycin, leads to DNA damage preferentially at telomeres,

45

which can be rescued by TRF2 inhibition. POT1 inhibition exacerbates telomere

46

dysfunction induced by ICRF-193. Thus, ICRF-193 induces damage at telomere

47

properly capped by TRF2 but not by POT1. These findings are expected to broaden

48

our view on the mechanism by which conventional therapeutic molecules act to

49

eliminate cancer cells and how to use TRF2 and POT1 level as surrogate markers for

50

anti-topoisomerase II sensitivity.

51 52 53

Keywords: Topoisomerase II, ICRF-193, telomere, TRF2, POT1

54

INTRODUCTION

55

Telomeres are key features of chromosome termini that preserve genome integrity(5) .

56

In many organisms, their protective functions depend upon at least two pathways. The

57

first relies on telomerase, which can compensate for replicative erosion(8); the second

58

relies on the binding to telomeric DNA of the shelterin protein complex composed of

59

six polypeptides (TRF1, TRF2, RAP1, Tin2, TPP1 and Pot1)(11). Three of the

60

shelterin components recognize directly telomeric DNA; TRF1 and TRF2 bind

61

telomeric DNA duplexes, while Pot1 binds single-stranded 3’ overhangs. Among the

62

shelterin subunits, TRF2(3) is at the heart of the molecular events that maintain

63

telomere integrity in mammals by preventing at telomeres different events normally

64

occurring during DNA damage response : activation of ATM pathway(15),

65

non-homologous

66

Moreover, TRF2 helps folding the telomeric DNA into t-loop, which might contribute

67

to telomere protection by masking the 3’ overhang (1, 12).

end-joining

(NHEJ)(2)

and

homologous

recombination(9).

68

A correct function of telomere seemingly displays antagonistic functions as far as

69

tumorigenesis is concerned(7). On one hand, reactivation of telomerase in cancer cells

70

is crucial for tumour progression. On the other hand, telomere shortening cooperates

71

with p53 deficiency to favour carcinogenesis in aged mice(16). TRF2 appears also to

72

play an important role in oncogenesis as it is up-regulated in several human tumors

73

(4,14) and has been found to exert oncogenic properties when overexpressed in mouse

74

keratinocytes(6). Recently, we showed that the high level of TRF2 found in cancer

75

cells activates the expression of positively regulates the transcription of HS3ST4, a

76

gene encoding an heparan sulfate (glucosamine) 3-O-sulphotransferase that is

77

involved in the recruitment of NK-cells, revealing that TRF2 upregulation can have

78

oncogenic properties through cell non-autnomous mechanisms (4).

79

Inhibition of telomerase provides an interesting “universal” strategy for cancer

80

therapy: immunological and pharmacological inhibition of telomerase is currently

81

used in a series of clinical protocols. However, with many but not all telomerase

82

therapeutic approaches, growth arrest has been observed only when telomeres reach a

83

critically short length. An alternative approach, although still poorly developed, is to

84

target key components of telomere structure such as the use of G4 ligand(17).

85

Interestingly, although DNA damages induced by drugs used in conventional

86

chemotherapy are usually believed to occur stochastically in the genome, a wealth of

87

evidence indicate that telomere stability is more affected than the bulk of the genome

88

by several genotoxic molecules (e.g. cisplatin, spindle poisons as well as

89

anti-topoisomerase I and II and replication inhibitors) (13). For instance, hydroxyurea

90

(HU) is a chemotherapeutic agent commonly used for various malignancies and

91

hematological disorders, including chronic myelogenous leukemia and sickle cell

92

anemia. Chronic, low-level treatment with HU preferentially decreased the rate of

93

telomere DNA synthesis and dissociated TRF2 from telomere DNA. All these indicate

94

that a broad spectrum of genotoxic drugs can have specific DNA damage at telomeres

95

either by binding directly to the DNA structure, or by occupancy the interactive

96

domain of telomeres to specific telomeric nucleoproteins.

97

Collectively, these findings linking conventional chemotherapies to telomeres

98

open new avenues for innovative combinations of chemotherapy drugs, for the use of

99

telomere parameters as surrogate markers for therapy response and toxicity as well as

100

for the introduction of future drugs against telomerase and telomere structural

101

components. Therefore, there is a need for a precise understanding of the impact of

102

genotoxic drugs on telomere functions. In this context, this work was designed to

103

investigate the effects of the topoisomerase II catalytic inhibitor ICRF-193 (meso-2,

104

3-bis (2, 6-dioxopiperazin-4-yl) butane) on telomere functions and conversely to

105

study the impact of specific telomere dysfunctions on ICRF-193 effects. We found

106

that dysfunctional telomeres as a consequence of TRF2 inhibition, but not of POT1

107

inhibition, are less sensitive to ICRF-193 than control ones. This might be due to the

108

fact that TRF2 has seemingly opposed roles regarding telomere topology: it can create

109

topological barriers, rendering telomeres more sensitive to topoisomerase depletion,

110

and it can cooperate with topoisomerase II to prevent replicative damage at

111

telomeres(19). These findings are expected to broaden our view on the mechanism by

112

which conventional therapeutic molecules act to eliminate cancer cells and how to use

113

TRF2 and POT1 level as surrogate markers for anti-topoisomerase II sensitivity.

114

115

MATERIALS AND METHODS

116

Cell Culture, Transfections and Lentiviral or retrovirus production and

117

infections

118

Human Embryonic Kidney 293 phoenix cells and HT1080 fibrosacoma cells were

119

grown in DMEM with 10% fetal calf serum and penicillin-streptomycin at 37ºC.

120

Transfections of plasmid DNA were performed with lipofectamine 2000 reagent

121

(Invitrogen) as described by the protocol. Empty vector or expressing TRF2∆B∆M or

122

shPOT1 vector, were produced by transient transfection of Human Embryonic Kidney

123

293 Phenix cell lines with two other packaging plasmids, p8.91 and pVSVg. 48 h and

124

72 h post transfection, the infectious supernatant was collected and applied to the

125

target cells. The efficiency of infection was determined by flow cytometry analysis of

126

GFP expression. The stable HT1080 cell line with POT1 knockdown induced by

127

shPOT1 was selected by100nM puromycin for 3 three weeks. HT1080 cells treated

128

with ICRF-193 (3uM, 24H), or Bleomycin (1uM, 24H) before performing further

129

experiments.

130 131

Plasmid constructs and siRNAs

132

TRF2∆B∆M was subcloned in pWPIR-GFP lentiviral vector upstream of the IRES in

133

order to allow individual translation of the bicistronic mRNA containing both

134

myc-TRF2∆B∆M and GFP. pLKO.1-puro lentivirus vectors expressing shPOT1 or

135

empty vector as a control were purchased from Sigma.

136 137

Immuno- fluorescence detection

138

Slides were either fixed by MeOH at -20°C, or by 4% formaldehyde at room

139

temperature10 to 15 min, then incubated 1h with blocking buffer (0,8x PBS, 50mM

140

NaCl, 0,5% Triton X100, 3% milk),

followed by 1h of incubation at 37°C with lst

141

antibody Rabbit polyclonal to 53BP1 (NOVUS BIOLOGICALS – NB 100-305) in

142

blocking buffer. Cells were then washed with 0.8xPBS, 50 mM NaCl, 0.1% Trition

143

X-100, followed by Donkey polyclonal anti mouse ALEXA488 (A21202;

144

MOLECULAR PROBES) and Donkey polyclonal anti rabbit ALEXA555 (A-31572;

145

MOLECULAR PROBES) . After washing with 0.8xPBS, 50 mM NaCl, 0.1% Trition

146

X-100, the nucleus was labelled with DAPI and/or TOTO3.

147 148

Combined immuno-fluorescence detection and PNA or DNA fluorescence in situ

149

hybridisation

150

Cells were permeabilised in Triton X-100 buffer (20 mM Tris-HCl pH 8.0, 50 mM

151

NaCl, 3 mM MgCl2, 0.5% Triton X-100 and 300 mM sucrose) at room temperature

152

(RT) for 5 min, followed by 15 min fixation in 3% (para) formaldehyde and 2%

153

sucrose, then again permeabilized in Triton X-100 buffer at RT for 10 min. Cells were

154

blocked in 2% BSA in PBS overnight at 4°C. Then followed IF steps shown above.

155

After final washes, slides were fixed for 2 min. at RT in 4% (para) formaldehyde,

156

followed by dehydration through an ethanol series (50%, 75% and 100%, 5 min. each).

157

Fluorochrome-coupled PNA probe (AATCCC)

158

PNA probe/µl, dissolved in 70% formamide, 10 mM Tris pH 7.2, 1% Blocking

159

Reagent (ROCHE BIOCHEMICALS) first denaturing at 80°C for 3 min, then

160

hybridisation for 2h at RT. Alternatively, slides were hybridised either with the

161

digoxigenin-labeled ITS probe (8ng/µl in 50% formamide / 2xSSC / 10% dextrane

162

sulphate) or with a probe labeling the subtelomeric region of the long arm of

163

chromosome 4 (TelVysion DNA Probes, VYSIS). Washes were the following: 2x15

164

min. at RT in 70% formamide, 10 mM Tris pH 7.2, 3x5 min. in 50 mM Tris pH 7.5,

was then applied to the slides (0.3 ng

165

150 mM NaCl,0.05% Tween-20. Finally, slides were counterstained with DAPI,

166

mounted in VECTASHIELD (VECTOR LABORATORIES).

167 168

Microscopy.

169

Images of metaphase preparations and Cytokinesis-Block micronucleus cytome assay

170

were recorded on an AxioPlan microscope from ZEISS, equipped with a

171

Plan-Apochromat 63x, NA 1.4, oil immersion lens and a cooled CCD camera

172

(CoolSNAP HQ, PHOTOMETRICS). Image acquisition, processing and analysis

173

software were from MetaMorph (MOLECULAR DEVICES).

174

Confocal images were made with the ZEISS LSM-510 confocal scanning laser

175

microscope. Optical sections were recorded at an interval of less than 200nm, with a

176

Plan-Apochromat 63x, NA 1.4, oil immersion lens. Excitation wavelengths were

177

488nm, 543nm and 633nm for the FITC-labeled PNA and DNA probes, for the

178

Alexa555-labeled secondary antibodies and for the TOTO3-stained DNA respectively.

179

Co-localisations were scored in each optical section by scrolling through the z-stack.

180

The appearance of the images was improved by whole-image operations with

181

Photoshop (ADOBE).

182 183

Statistical analysis

184

According to the results of univeriate test, continuous normal variables were

185

expressed as mean value±SD. Categorical variables were expressed as percentage;

186

comparison between these groups were analyzed by Chi-Square test. Parametric

187

variables of normal distribution were analyzed either by 2-tail T-test, or by F test of

188

ANVOA followed by Duncan test for each two group comparison. Results were

189

considered significant at p

The topoisomerase II catalytic inhibitor ICRF-193 preferentially targets telomeres that are capped by TRF2.

The increased level of chromosome instability in cancer cells is not only a driving force for oncogenesis but also can be the Achille's heel of the di...
609KB Sizes 0 Downloads 9 Views