Accepted Manuscript Novel insights into the Apoptosis mechanism of DNA topoisomerase I inhibitor isoliquiritigenin on HCC tumor cell Ze-xin Li, Jian Li, Yan Li, Kun You, Hongwei Xu, Jianguo Wang PII:

S0006-291X(15)30234-5

DOI:

10.1016/j.bbrc.2015.07.003

Reference:

YBBRC 34211

To appear in:

Biochemical and Biophysical Research Communications

Received Date: 25 June 2015 Accepted Date: 1 July 2015

Please cite this article as: Z.-x. Li, J. Li, Y. Li, K. You, H. Xu, J. Wang, Novel insights into the Apoptosis mechanism of DNA topoisomerase I inhibitor isoliquiritigenin on HCC tumor cell, Biochemical and Biophysical Research Communications (2015), doi: 10.1016/j.bbrc.2015.07.003. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT

Novel insights into the Apoptosis mechanism of DNA

2

topoisomerase I inhibitor isoliquiritigenin on HCC tumor

3

cell

4

Ze-xin Li, Jian Li, Yan Li, Kun You, Hongwei Xu, Jianguo Wang*

RI PT

1

5

SC

6 7

M AN U

8 9 10

14 15 16 17 18 19

EP

13

AC C

12

TE D

11

20

1

ACCEPTED MANUSCRIPT Department of General Surgery, the First Affiliated Hospital of Xinxiang Medical University,

22

453100

23

*Corresponding Author: Jianguo Wang

24

Tel. /fax: +86 18001075725

25

E-mail addresses: [email protected]

26

ABSTRACT

SC

RI PT

21

The inhibitory effect of DNA topoisomerase (Top I) by isoliquiritigenin(ISO)

28

were investigated and their interaction mechanism was evaluated using methods

29

including UV–vis absorption, fluorescence, coupled with molecular simulation, and

30

using

31

proliferation assay, finally, the interaction of ISO with calf thymus DNA was

32

investigated by melting measurements and molecular docking studies. It was found

33

that isoliquiritigenin reversibly inhibited DNA Top I in a competitive manner with

34

the concentrations of ISO resulting in 50% activity lost (IC50) were estimated to be

35

0.178 ± 0.12 mM. Isoliquiritigenin exhibited a strong ability to quench the intrinsic

36

fluorescence of Top I through a static quenching procedure. The positive values of

37

enthalpy change and entropy change suggested that the binding of isoliquiritigenin to

38

Top I was driven mainly by hydrophobic interactions. The molecular docking results

39

revealed isoliquiritigenin actually interacted with the primary amino acid residues on

40

the active site of Top I, and the detection results of fluorescence staining and the

41

inhibitory effect on the growth of HCC SUN475 showed that isoliquiritigenin

MTT

method

of

inhibition

rate of HCC

tumor cell SNU475

AC C

EP

TE D

the

M AN U

27

2

ACCEPTED MANUSCRIPT induced the apoptosis cells increased gradually. The interaction of ISO with DNA

43

can cause the denaturation temperature to be increased, which indicated that the

44

stabilization of the DNA helix was increased in the presence of ISO, which indicated

45

that the results provide strong evidence for intercalative binding of ISO with DNA.

46

Keywords: Isoliquiritigenin; DNA topoisomerase I; Inhibition effect; Fluorescence

47

quenching; Melting measurements; Molecular simulation; Apoptosis cells

48

1.Introduction

SC M AN U

49

RI PT

42

The topoisomerase I (Top I) family of eukaryotic enzymes is required to relax DNA

supercoiling

generated

by

replication,

transcription,

and

chromatin

51

remodeling[1-4]. Human Top I acts through a nucleophilic tyrosine residue (Tyr723),

52

which nicks the phosphodiester backbone of double-stranded, supercoiled DNA and

53

forms a transient cleavage complex in which the 3′end of the broken DNA strand is

54

covalently linked to the enzyme[5-8]. Camptothecin is a natural product for which

55

Top I is its only cellular target[9]. Two camptothecin derivatives, irinotecan and

56

topotecan, are the only current Top I inhibitors approved by the U.S. Food and Drug

57

Administration for the treatment of cancer[10,11]. However, these camptothecin

58

derivatives have several major drawbacks. First, camptothecins are compromised by

59

the reversibility of the Top1-DNA cleavage complex, which necessitates long

60

infusion times for maximum activity[12-14]. Second, the lactone ring is inherently

61

unstable and hydrolyzes to form an inactive hydroxy acid[15,16]. In addition, the

62

anticancer activities of the camptothecins are compromised by R364H[17] and

AC C

EP

TE D

50

3

ACCEPTED MANUSCRIPT N722S[18] mutations as well as by induction of the ABCG2[19-21] and MXR[21]

64

ATP-binding cassette drug efflux transporters. Myelosuppression is dose-limiting

65

with topotecan[22], whereas the major dose-limiting toxicities of irinotecan are

66

neutropenia and diarrhea[23]. These limitations have stimulated the search for

67

non-camptothecin Top I inhibitors as anticancer agents.

RI PT

63

Isoliquiritigenin(Fig. 1A) was found to be a Top I inhibitor with anticancer

69

activity in the National Cancer Institutes 60 (NCI60)-cell screen indicated a high

70

degree of correlation with camptothecin. Subsequent studies confirmed that the

71

mechanism of action of the isoliquiritigenin is identical to that of the camptothecins.

72

Specifically, they stabilize the ternary cleavage complex by intercalation between the

73

DNA base pairs at the cleavage site after single-strand cleavage by Top I, thus

74

preventing religation of the broken phosphodiester backbone, which are therefore

75

classified as Top I poisons as opposed to Top I suppressors, which inhibit the DNA

76

cleavage reaction.

77

2. Materials and methods

78

2.1. Fluorescence Spectra Measurements

M AN U

TE D

EP

AC C

79

SC

68

Fluorescence (F-7000, Hitachi) measurements were measured at different

80

temperatures (298, 304, and 310 K) in range of 300–500 nm with an excitation

81

wavelength at 280 nm, and the excitation and emission bandwidths were both set at

82

2.5 nm. Appropriate blanks corresponding to the buffer are subtracted to correct

83

background fluorescence. 4

ACCEPTED MANUSCRIPT In the present work, the following relationship was used to correct the

84 85

inner-filter effect [24]:

86

Fc = Fme ( A1 +A 2 )/2

87

where Fc and Fm are the corrected and measured fluorescence, respectively. A1 and A2

88

are the absorbance of ISO at excitation and emission wavelengths, respectively.

RI PT

(1)

SC

The fluorescence quenching data were analyzed by the Stern–Volmer equation

89

[25]:

91

F0 = 1 + K SV [Q] = 1 + K qt 0 [Q] F

92

where F0 and F are the fluorescence intensities (344 nm) of Top I in the absence and

93

presence of quencher, respectively. KSV is the Stern–Volmer quenching constant (KSV

94

= Kqτ0), Kq is the quenching rate constant of biomolecule, [Q] is the concentration of

95

ISO. τ0 is the average lifetime of the fluorophore without quencher, and the value of

96

τ0 of the biopolymer is 10−8 s [26].

(2)

EP

TE D

M AN U

90

AC C

The binding constant (K) and the number of bound ISO to TOP I (n) were

97 98

determined by the following equation [27]:

99

log

F0 - F F

= log K + n log[Q]

(3)

100

If the temperature does not vary significantly, the enthalpy change (∆H°) can be

101

regarded as a constant, then its value and entropy change (∆S°) value can be

102

evaluated from the van’t Hoff equation as follows: 5

ACCEPTED MANUSCRIPT DH ° DS ° + 2.303RT 2.303R

103

log K a = -

104

where R is the gas constant, and the temperatures used were 298, 304, and 310 K.

105

The value of free energy change (∆G°) can be obtained from the following equation:

106

DG ° = DH ° - T DS °

107

2.2 In-vitro enzyme inhibition study

RI PT

(4)

SC

(5)

In nature, usually plasmids exist as negatively supercoiled forms. Supercoiled

109

pUC 19 was used as the substrate for relaxation assays. IU of enzyme was defined as

110

the least quantity of the TOP I enzyme required for complete relaxation of 0.5 µg of

111

negatively supercoiled plasmid pUC 19 DNA in 30 min at 37℃ [28]. The DNA

112

relaxation assay was performed in a standard reaction volume of 30 µL assay buffer

113

with 10 mM Tris–HCl (pH 7.0), 50 mM NaCl, 6 mM MgCl2, 0.1 mg/mL gelatin and

114

0.5 µg of supercoiled pUC 19 plasmid DNA (HiPurA plasmid DNA maxiprep

115

purification spin kit, HiMedia). Reactions were initiated by adding 800 ng TOP I

116

protein. Later, after incubation at 37℃ for 30 min, the reactions were quenched by

117

adding 5 µL of Stop buffer (50 mM EDTA (pH 7.0); 50% glycerol and 0.5% (v/v)

118

bromophenol blue). Subsequently, the DNA was electrophoresed in a 1% (w/v)

119

agarose gel with TAE buffer. The gels were stained after the electrophoresis to

120

prevent any differences in mobility of DNA due to chelation;gel was stained with

121

ethidiumbromide and photographed over UV light using GelDoc imager (Bio-Rad).

122

The supercoiled plasmid was considered as blank; the supercoiled DNA with the

AC C

EP

TE D

M AN U

108

6

ACCEPTED MANUSCRIPT TOP I enzyme was used as control with 100% relaxation. Different concentrations of

124

the ISO tested were assayed to know the percentage inhibitions on the enzyme.

125

2.3 In-vitro cytotoxicity screening

RI PT

123

ISO was further screened for cytotoxic activity using Human HCC tumor cell

127

line (SUN 475) at 25 µM. The cells were grown in RPMI medium with 10% FBS.

128

The activity was carried out using MTT assay. The enzymes in the viable cell have

129

the capability of reducing the tetrazolium dye MTT to its reduced form formazon,

130

which has purple color. The cells were grown in RPMI-1640 medium supplemented

131

with 10% FBS (Fetal bovine serum), 100 IU/mL penicillin, 100 mg/mL streptomycin

132

and 2 mM glutamine. Cell line was maintained at 37℃ in a humidified 5% CO2

133

incubator. Cells were scrapped and seeded into wells approx 5000 cells per well in

134

poly-L-lysine coated plates. The plates were incubated at 37℃in humidified CO2

135

incubator and then treated with the synthesized compounds at 25 µM concentration

136

and incubated for 48 h. 10 µL of 0.5 mg/mL concentration of MTT was added and

137

incubated for 3 h at 37℃ and the final product formazon crystals were dissolved in

138

100 µL of DMSO and the viability was measured at 595 nm.

139

2.4 DNA melting studies

AC C

EP

TE D

M AN U

SC

126

140

The melting temperatures of DNA and ISO–DNA complex were measured by

141

monitoring the absorption intensities at 260 nm of samples at various temperatures.

142

The absorbance intensities were then plotted as a function of temperature ranging

143

from 20 to 100℃. The melting temperatures (Tm) of DNA and ISO–DNA complex 7

ACCEPTED MANUSCRIPT 144

were determined as the transition midpoint.

145

2.5. Molecular Docking Studies

The 3D structure of ISO was generated in Sybyl × 1.1 (Tripos Inc., St. Louis),

147

its energy-minimized conformation was obtained with the help of the MMFF94 force

148

field using MMFF94 charges, and the TOP I(PDB: 1T8I), DNA(PDB: 453D)[29]

149

model were used as the receptor model in the virtual screening with docking

150

simulations. All the water molecules were removed, the polar hydrogen atoms and

151

kollman charges were added to the TOP, DNA model, the grid maps for energy

152

scoring were calculated using AutoGrid. Docking calculations were performed using

153

the default parameters (runs 100) and the rotatable bonds in the ligand were assigned

154

with AutoDock Tools, and the ligand docking was performed with the AutoDock 4.2

155

[30] Lamarckian Genetic Algorithm (LGA), and output from AutoDock was rendered

156

with PyMol,

157

2.6. Statistical Analysis

EP

TE D

M AN U

SC

RI PT

146

The experimental results was expressed as mean value ± standard deviation (n =

159

3), and data were analyzed using the SAS statistical package (version 8.1, SAS

160

Institute, Cary, NC, USA).

161

3.Results and discussion

162

3.1. Fluorescence quenching studies of TOP I by lSO

163

AC C

158

Fig1. A shows that the fluorescence intensity of TOP I at 344 nm decreased 8

ACCEPTED MANUSCRIPT 164

markedly With increasing amounts of ISO to TOP I solution, while ISO did not show

165

intrinsic fluorescence under the same experimental conditions, indicating that ISO

166

could interact with TOP I.

The Stern–Volmer plots for the quenching of TOP I by ISO at three different

168

temperatures are presented in Fig1. A. Based on equation 2, table 1 shows that the

169

KSV values for the interaction of ISO with TOP I decreased with increasing

170

temperature, and the corresponding Kq values were in order of 1012 L mol−1 s−1,

171

which were greater than the limiting diffusion rate constant of the biomolecule (2 ×

172

1010 L mol−1 s−1). It was mainly controlled by a static quenching mechanism

173

resulting from the formation of ISO–TOP I complex [31].

M AN U

SC

RI PT

167

Basis of equation 3, the increasing trend of binding constant (K) with rising

175

temperature was observed (Table 1), indicating that the capacity of ISO binding to

176

TOP I was enhanced and the binding was an endothermic and entropy-driven

177

reaction [32]. The number of binding sites (n) were approximately equal to 1,

178

suggesting the existence of just a single binding site on TOP I for ISO which

179

convincingly supported the conclusion obtained from Lineweaver–Burk Plot

180

Analysis.

181

3.2. Thermodynamic analysis and the nature of the binding forces

AC C

EP

TE D

174

182

Table 1 lists the thermodynamic parameters for the interaction of ISO with

183

TOP I. The negative signal for ∆G° indicated the ISO–TOP I interaction was

184

spontaneous. The values of ∆H° and ∆S° were 17.61 ± 0.1 kJ mol−1 and 140.65 ± 9

ACCEPTED MANUSCRIPT 0.22 J mol−1 K−1, respectively, suggesting that hydrophobic interactions played

186

predominant roles in the interaction process [33].

187

3.3 In-vitro enzymatic assay of ISO

RI PT

185

TOP I assay performed by gel based assays, with a range of ISO concentrations

189

from 0.01 to 3.0 mM. A dose dependent inhibitory profile of ISO is shown in Fig1 B.

190

Fig1. B showed a better activity for ISO against the enzyme, when tested at lower

191

concentrations of 0.01 mM, 0.05 mM, and 0.1 mM, ISO was not emerged as the

192

good inhibitory effect on TOP I, with increasing concentration of ISO, the enzymatic

193

activity decreased remarkably. The concentrations of ISO resulting in 50% activity

194

lost (IC50) were estimated to be 0.178 ± 0.12 mM.

195

3.4. Computational docking of ISO on TOP I

TE D

M AN U

SC

188

As shown in Fig. 2A, cluster analysis was performed using a rmsd tolerance of

197

2.0 Ǻ. A total of 12 multimember conformational clusters were obtained from 20

198

docking runs. The most energetically favorable cluster possessed lowest energy and

199

the most frequent locus (red histogram) was used for binding orientation analysis.

200

Table 2 lists the binding free energy of this position, it can be seen that the calculated

201

binding energy (–4.71 kcal mol–1) was same to the predicted ∆Gbinding (–4.83 kcal

202

mol–1), indicating that the docking result was credible. The binding energy was a little

203

larger than ∆G° (–5.72 kcal mol–1) obtained from the thermodynamic analysis at 298

204

K. As the molecular docking was conducted under simulation of vacuum

205

environment, the difference of the binding energy between docking results and

AC C

EP

196

10

ACCEPTED MANUSCRIPT 206

experiment results could be attributed to the lack of desolvation energy.

In view of the above, the docking result (Fig. 2A) showed that the binding site

208

on TOP I for ISO was binding to the active site. Indeed, it was also observed that the

209

binding site for ISO was close to the active site pocket (Fig. 2B), and ISO was

210

surrounded by the residues, namely, Tyr723, Thr718, Asn352, and Arg364. Moreover,

211

one hydroxyl group on A ring could form hydrogen bonds (green line) with residue

212

Tyr723, indicating hydrogen bond was another main role in the binding of ISO with

213

TOP I. The docking results indicated ISO actually interacted with the amino acid

214

residues located in the region of TOP I, which confirmed ISO as a competitive

215

inhibitor with a single high-affinity binding site on TOP I.

216

3.5 In-vitro cytotoxicity screening

TE D

M AN U

SC

RI PT

207

Fig 3 exhibits the inhibition effect of ISO on SUN475, a concentration

218

dependent-manner was observed in the SUN475 inhibitory activity. With increasing

219

concentration of ISO, the SUN475 inhibitory activity decreased remarkably. The

220

concentrations of ISO and camptothecin resulting in 50% activity lost (IC50) were

221

estimated to be 0.243 ± 0.21 and 0.238 ± 0.41 mM, respectively. The results

222

indicated that ISO has significant SUN475 inhibitory activity compared with

223

camptothecin.

224

3.6. DNA melting studies

225

AC C

EP

217

Under the condition that DNA solutions are exposed to extremes of pH or heat, 11

ACCEPTED MANUSCRIPT the double helical structure of DNA undergoes a transition into a randomly

227

single-stranded form at the melting temperature (Tm). In addition, the melting

228

temperature can be altered by the interaction of DNA with small molecules [34].

229

Generally, classical intercalation of small molecules into the double helix causes

230

stabilization of base stacking and leads to a significant increase of Tm (about 5–8℃),

231

while non-intercalation (e.g. groove or electrostatic) binding causes no obvious

232

increase in Tm [35]. As shown in Fig. 4A, upon the binding of ISO, the Tm value of

233

DNA was increased from 80.6℃ to 83.2℃ and increment was smaller than 5–8℃.

234

Therefore, it can be deduced that ISO can interact with DNA via non-classical rather

235

than classical intercalation.

236

3.7. Computational docking of ISO on DNA

TE D

M AN U

SC

RI PT

226

Cluster analysis was performed using a rmsd tolerance of 2.0 Ǻ. A total of 15

238

multimember conformational clusters were obtained from 20 docking runs. For the

239

reason that the most energetically favorable cluster possessed lowest energy, the

240

predicted binding model with the lowest binding energy was then used for binding

241

orientation analysis. Fig. 4B showed the docked conformation of the ligand ISO with

242

DNA. ISO was surrounded by DA5, DA6, DT7, DT8, DT19, and DT20. It can be

243

observed that the aromatic ring of ISO can intercalate into the intermediate space of

244

the two base pairs parallelly and the planar double bond vertically interact with the

245

base pair of DNA. The docking studies gave a prediction of the interaction between

246

ISO and DNA based on lowest energy principle. According to the results, the

AC C

EP

237

12

ACCEPTED MANUSCRIPT probable binding mode was intercalating binding and the formation of one hydrogen

248

bonds suggested that A-T base pair may be the active binding site. It should be noted

249

that such extracted information is unobtainable with the use of conventional methods

250

of data interpretation. We hope that this work can benefit further understanding of

251

the

252

pharmacological effects as well as the design of the structure of new and efficient

253

drug molecules.

254

Conflict of interest

255

None declared.

of

with

DNA and

comprehension

ISO’s

TE D

256

257

ISO

SC

mechanism

M AN U

binding

RI PT

247

References

259

[1] Y. Pommier, DNA Topoisomerase I Inhibitors: Chemistry, Biology, and

260

Interfacial Inhibition, Chem. Rev. 109 (2009) 2894−2902.

261

[2] Y. Pommier, Topoisomerase I Inhibitors: Camptothecins and Beyond, Nat. Rev.

262

Cancer 6 (2006) 789 −802.

263

[3] J.J. Champoux, DNA Topoisomerases: Structure, Function, and Mechanism,

264

Annu, Rev. Biochem. 70 (2001) 369−413.

265

[4] L.Stewart, M.R. Redinbo, X. Qiu, W.G.J. Hol, J.J. Champoux, A Model for the

AC C

EP

258

13

ACCEPTED MANUSCRIPT Mechanism of Human Topoisomerase I, Science 279 (1998) 1534 −1541.

267

[5] Y. Pommier, J.A. Barcelo, V.A. Rao, O. Sordet, A.G. Jobson, L. Thibaut, Z.H.

268

Miao, J.A. Seiler, H. Zhang, C. Marchand, K. Agama, C. Redon, Repair of

269

Topoisomerase I-Mediated DNA Damage, Prog. Nucleic Acid Res. Mol. Biol. 81

270

(2006) 179 −229.

271

[6] O. Sordet, Q.A. Khan, K.W. Kohn, Y. Pommier, Apoptosis Induced by

272

Topoisomerase Inhibitors, Curr. Med. Chem.: Anti-Cancer Agents 3 (2003) 271

273

−290.

274

[7] B.L. Staker, K. Hjerrild, M.D. Feese, C.A. Behnke, A.B. Burgin, L. Stewart,

275

The Mechanism of Topoisomerase I Poisoning by a Camptothecin Analogue, Proc.

276

Natl. Acad. Sci. U.S.A. 99 (2002) 15387−15392.

277

[8] J.C. Wang, Cellular Roles of DNA Topoisomerases: A Molecular Perspective,

278

Nat. Rev. Mol. Cell Biol. 3 (2002) 430 −440.

279

[9] M.E. Wall, M.C. Wani, C.E. Cook, K.H. Palmer, A.T. McPhail,. G.A. Sim,

280

Plant Antitumor Agents. I. The Isolation and Structure of Camptothecin, a Novel

281

Alkaloidal Leukemia and Tumor Inhibitor from Camptotheca acuminata, J. Am.

282

Chem. Soc. 88 (1966) 3888−3890.

283

[10] B. Teicher, Next Generation of Topoisomerase I Inhibitors: Rationale and

284

Biomarker Strategies, Biochem. Pharmacol. 75 (2008) 1262−1271.

285

[11] C.J. Thomas, N.J. Rahier, S.M. Hecht, Camptothecin: Current Perspectives.

AC C

EP

TE D

M AN U

SC

RI PT

266

14

ACCEPTED MANUSCRIPT Bioorg. Med. Chem. 12 (2004) 1585−1604.

287

[12] C.J. Li, L. Averboukh, A.B. Pardee, Beta-Lapachone, a Novel DNA

288

Topoisomerase I Inhibitor with a Mode of Action Different from Camptothecin, J.

289

Biol. Chem. 268 (1993) 22463−22468.

290

[13] C .Jaxel, K.W. Kohn, M.C. Wani, Y. Pommier, Structure−Activity Study of the

291

Actions of Camptothecin Derivatives on Mammalian Topoisomerase I: Evidence for

292

a Specific Receptor Site and a Relation to Antitumor Activity. Cancer Res. 49 (1989)

293

1465−1469.

294

[14] H. Minami, J.H. Beijen, J. Verwij, M.J. Ratain, Limited Sampling Model for

295

Area under the Concentration Time Curve of Total Topotecan, Clin. Cancer Res. 2

296

(1996) 43−46.

297

[15] M.J. Luzzio, J.M. Besterman, D.L. Emerson, M.G. Evans, K. Lackey, P.L.

298

Leitner, G. McIntyre, B. Morton, P.L. Myers, et al, Synthesis and Antitumor-Activity

299

of Novel Water-Soluble Derivatives of Camptothecin as Specific Inhibitors of

300

Topoisomerase-I, J. Med. Chem. 38 (1995) 395−401.

301

[16] Z. Mi, T.G. Burke, Differential Interactions of Camptothecin Lactone and

302

Carboxylate Forms with Human Blood Components, Biochemistry 33 (1994)

303

10325−10336.

304

[17] Y. Urasaki, G.S. Laco, P. Pourquier, Y. Takebayashi, G. Kohlhagen, C. Gioffre,

305

H.L. Zhang, D. Chatterjee, P. Pantazis, Y. Pommier, Characterization of a Novel

AC C

EP

TE D

M AN U

SC

RI PT

286

15

ACCEPTED MANUSCRIPT Topoisomerase I Mutation from a Camptothecin-Resistant Human Prostate Cancer

307

Cell Line, Cancer Res. 61 (2001) 1964−1969.

308

[18] A. Fujimori, W.G. Harker, G. Kohlhagen, Y. Hoki, Y. Pommier, Mutation at the

309

Catalytic Site of Topoisomerase I in CEM/C2, a Human Leukemia Cell Resistant to

310

Camptothecin, Cancer Res. 55 (1995) 1339−1346.

311

[19] S.E. Bates, W.Y. Medina-Perez, G. Kohlhagen, S. Antony, T. Nadjem, R.W.

312

Robey, Y. Pommier, ABCG2 Mediates Differential Resistance to SN-38

313

(7-Ethyl-10-hydroxycamptothecin) and Homocamptothecins, J. Pharmacol. Exp.

314

Ther. 310 (2004) 836 −842.

315

[20] Y. Hoki, A. Fujimori, Y. Pommier, Differential Cytotoxicity of Clinically

316

Important Camptothecin Derivatives in P-Glycoprotein Overexpressing Cell Lines.

317

Cancer Chemother. Pharmacol, 40 (1997) 433−438.

318

[21] M. Brangi, T. Litman, M. Ciotti, K. Nishiyama, G. Kohlhagen, C. Takimoto, R.

319

Robey, Y. Pommier, T. Fojo, Bates, S. E. Camptothecin Resistance: Role of the

320

ATP-Binding Cassette (ABC), Mitoxantrone-Resistance Half-Transporter (MXR),

321

and Potential for Glucuronidation in MXR-Expressing Cells, Cancer Res. 59 (1999)

322

5938 −5946.

323

[22] D.K. Armstrong, Topotecan Dosing Guidelines in Ovarian Cancer: Reduction

324

and Management of Hematologic Toxicity, Oncologist 9 (2004) 33 −42.

325

[23] L.B. Saltz, Clinical Use of Irinotecan: Current Status and Future Considerations,

AC C

EP

TE D

M AN U

SC

RI PT

306

16

ACCEPTED MANUSCRIPT Oncologist 2 (1997) 402 −409.

327

[24] S.Y. Bi, Y.T. Sun, C.Y. Qiao, H.Q. Zhang, C.M. Liu, Binding of several

328

anti-tumor drugs to bovine serum albumin: Fluorescence study, J. Lumin. 129 (2009)

329

541–547.

330

[25] Y.Q. Li, F.C. Zhou, F. Gao, J.S. Bian, F. Shan, Comparative Evaluation of

331

Quercetin, Isoquercetin and Rutin as Inhibitors of α-Glucosidase, J. Agric. Food

332

Chem. 57 (2009) 11463−11468.

333

[26] G.W. Zhang, L. Wang, J.H. Pan, Probing the Binding of the Flavonoid

334

Diosmetin to Human Serum Albumin by Multispectroscopic Techniques, J. Agric.

335

Food Chem. 60 (2012) 2721−2729.

336

[27] M. Liu, W. Zhang, L. Qiu, X.K. Lin, Synthesis of butyl-isobutyl-phthalate and

337

its interaction with α-glucosidase in vitro, J. Biochem. 149 (2011) 27–33.

338

[28] A.A. Godbole, M.N. Leelaram, A.G. Bhat, P. Jain, V. Nagaraja, Characterization

339

of DNA topoisomerase I from Mycobacterium tuberculosis: DNA cleavage and

340

relegation

341

Biochem. Biophys. 528 (2012) 197–203.

342

[29] H.M. Berman, J. Westbrook, Z.K. Feng, G. Gilliland, T.N. Bhat, H. Weissig,

343

L.N. Shindyalov, P.E. Bourne, The protein data bank, Nucl. Acid Res. 28(2000)

344

235–242.

345

[30] C.G. Ricci, P.A. Netz, Docking Studies on DNA-Ligand Interactions: Building

EP

TE D

M AN U

SC

RI PT

326

AC C

properties

and

inhibition

of

its

activity,

Arch.

17

ACCEPTED MANUSCRIPT and Application of a Protocol To Identify the Binding Mode, J. Chem. Inf. Model. 49

347

(2009) 1925–1935.

348

[31] H. Zhang, R.T. Liu, Z.X. Chi, C.Z. Gao, Toxic effects of different charged metal

349

ions on the target-Bovine serum albumin, Spectrochim. Acta, Part A. 78 (2011)

350

523–527.

351

[32] G.W. Zhang, Y.D. Ma, L. Wang, Y.P. Zhang, J. Zhou, Multispectroscopic studies

352

on the interaction of maltol, a food additive, with bovine serum albumin, Food Chem.

353

133 (2012) 264–270.

354

[33] P.D. Ross, S. Subramanian, Thermodynamics of protein association reactions:

355

Forces contributing to stability, Biochemistry. 20 (1981) 3096−3102.

356

[34] Y.D. Ma, G.W. Zhang, J.H. Pan, Spectroscopic studies of DNA interaction with

357

food colorant indigo carmine with the use of ethidium bromide as a fluorescence

358

probe, J. Agric. Food Chem. 60(2012) 10867–10875.

359

[35] Y.T. Sun, H.Q. Zhang, S.Y. Bi, X.F. Zhou, L.Wang, Y.S. Yan, Studies on the

360

arctilin and its interaction with DNA by spectral methods, J. Lumin. 131(2011)

361

2299–2306.

SC

M AN U

TE D

EP

AC C

362

RI PT

346

363

364

18

ACCEPTED MANUSCRIPT 365

366

RI PT

367

Figure Legends

369

Fig 1. (A) Fluorescence spectra of TOP I in the presence of ISO at different

370

concentrations (pH 7.0, T = 298 K, λex = 280 nm, λem = 341 nm). c(TOP I) = 2.0 ×

371

10−6 M, and c(ISO) = 0, 0.3, 0.6, 0.9, 1.2, 1.5, 1.8, 2.1, and 2.4 mM for curves a→i,

372

respectively; The Stern–Volmer plots for the fluorescence quenching at different

373

temperatures. (B) Dose dependent inhibitory profile of ISO on TOP I; DNA-blank

374

(supercoiled DNA); TOP I-(supercoiled DNA + TOP I + DMSO); ISO- (supercoiled

375

DNA + TOP I + ISO at varied concentrations in DMSO), and the predicted binding

376

mode of ISO docked with TOP I.

377

Fig 2. (A) Cluster analysis of the AutoDock docking runs of ISO with TOP I, and the

378

interaction between ISO and TOP I. The residues of TOP I and ISO structure were

379

represented

380

hydrogen-bonding interactions. (B) Predicted binding mode of ISO docked with TOP

381

I on molecular surface.

382

Fig. 3 Human HCC tumor cell line (SUN 475) were treated with the ISO and

383

camptothecin for 7 days at 37℃. The apoptosis of both treated and untreated cells

384

was tested by MTT assay.

EP

TE D

M AN U

SC

368

stick

AC C

using

structures;

The

dashed

lines

(green)

represent

19

ACCEPTED MANUSCRIPT Fig. 4 (A) Melting curves of DNA in the absence and presence of ISO at pH 7.0. cISO

386

= 0.25 mM, and cDNA = 0.06 mM. (B) Cluster analyses of the AutoDock docking

387

runs of ISO with DNA; Docking pose of the energy-minimized structure of

388

ISO–TOP I complex.

RI PT

385

AC C

EP

TE D

M AN U

SC

389

20

ACCEPTED MANUSCRIPT Table 1. The quenching constants (KSV), binding constants (K), number of binding sites (n) and relative thermodynamic parameters for the interaction of ISO with TOP I at different temperatures. KSV 4

−1

(×10 L mol )

Ra

K 4

−1

(×10 L mol )

n

Rb

−1

(kJ mol )

2.69 ± 0.02

0.9985

1.55 ± 0.02

1.00 ± 0.02

0.9998

304

1.98 ± 0.02

0.9988

1.78 ± 0.01

1.07 ± 0.01

0.9992

310

1.61 ± 0.01

0.9991

2.04 ± 0.01

1.08 ± 0.01

0.9992

R is the correlation coefficient for the KSV values.

17.61 ± 0.1

TE D EP AC C

(kJ mol )

(J mol−1 K−1)

–24.77 ± 0.02

140.65 ± 0.22

–25.61 ± 0.02

R is the correlation coefficient for the K

M AN U

values.

b

∆S° −1

–23.93 ± 0.02

SC

298

a

∆G°

∆H°

RI PT

T (K)

ACCEPTED MANUSCRIPT Table 2. The binding free energies of ISO and TOP I according to docking result.

Complex ISO–TOP I

Intermol energy

Internal energy

Torsional energy

Binding energy

Estimated ∆Gbinding

(kcal/mol)

(kcal/mol)

(kcal/mol)

(kcal/mol)

(kcal/mol)

–5.20

–0.32

0.37

–4.71

–4.83

AC C

EP

TE D

M AN U

SC

RI PT

Estimated ∆Gbinding is the sum of intermolecular energy and torsional free energy.

ACCEPTED MANUSCRIPT Fig. 1 (A)

RI PT

1

SC

Isoliquiritigenin

M AN U

2 3

(B)

AC C

EP

TE D

4

5 6 1

ACCEPTED MANUSCRIPT Fig. 2 (A)

SC

RI PT

7

8

(B)

11

AC C

EP

TE D

10

M AN U

9

12 13 14 15 2

ACCEPTED MANUSCRIPT 16

Fig.3

17

Camptothecin ISO

60 40 20 0 0.5

1.0 [I]/(mM)

18 19

23 24 25 26 27

EP

22

AC C

21

2.0

TE D

20

1.5

M AN U

0.0

RI PT

80

SC

Cell apoptosis rate /%

100

28 29 30 31 3

ACCEPTED MANUSCRIPT Fig. 4 (A)

SC

RI PT

32

33

M AN U

34 35 (B)

AC C

EP

TE D

36

37 38 39

4

ACCEPTED MANUSCRIPT

Highlights

> ISO reversibly inhibits TOP I activity in a A dose dependent manner.

RI PT

> Hydrophobic interactions play a major role in ISO–TOP I interaction. > ISO has a high affinity close to the active site pocket of TOP I.

AC C

EP

TE D

M AN U

SC

> The binding of ISO to DNA induces the stability of the structure of DNA.

Novel insights into the apoptosis mechanism of DNA topoisomerase I inhibitor isoliquiritigenin on HCC tumor cell.

The inhibitory effect of DNA topoisomerase (Top I) by isoliquiritigenin(ISO) were investigated and their interaction mechanism was evaluated using met...
924KB Sizes 0 Downloads 7 Views