Toxicokinetics, Tissue Distribution, and Excretion of Dufulin Racemate

May 21, 2018 - ABSTRACT: Dufulin is a plant antiviral agent with a novel molecular structure and has been used widely to prevent and control tobacco a...
0 downloads 0 Views 1MB Size
Subscriber access provided by Kaohsiung Medical University

Agricultural and Environmental Chemistry

Toxicokinetics, tissue distribution and excretion of dufulin racemate and its R (S)- enantiomers in rats Huaguo Chen, XIN ZHOU, and Baoan Song J. Agric. Food Chem., Just Accepted Manuscript • DOI: 10.1021/acs.jafc.8b01101 • Publication Date (Web): 21 May 2018 Downloaded from http://pubs.acs.org on May 21, 2018

Just Accepted “Just Accepted” manuscripts have been peer-reviewed and accepted for publication. They are posted online prior to technical editing, formatting for publication and author proofing. The American Chemical Society provides “Just Accepted” as a service to the research community to expedite the dissemination of scientific material as soon as possible after acceptance. “Just Accepted” manuscripts appear in full in PDF format accompanied by an HTML abstract. “Just Accepted” manuscripts have been fully peer reviewed, but should not be considered the official version of record. They are citable by the Digital Object Identifier (DOI®). “Just Accepted” is an optional service offered to authors. Therefore, the “Just Accepted” Web site may not include all articles that will be published in the journal. After a manuscript is technically edited and formatted, it will be removed from the “Just Accepted” Web site and published as an ASAP article. Note that technical editing may introduce minor changes to the manuscript text and/or graphics which could affect content, and all legal disclaimers and ethical guidelines that apply to the journal pertain. ACS cannot be held responsible for errors or consequences arising from the use of information contained in these “Just Accepted” manuscripts.

is published by the American Chemical Society. 1155 Sixteenth Street N.W., Washington, DC 20036 Published by American Chemical Society. Copyright © American Chemical Society. However, no copyright claim is made to original U.S. Government works, or works produced by employees of any Commonwealth realm Crown government in the course of their duties.

Page 1 of 35

Journal of Agricultural and Food Chemistry

Toxicokinetics, tissue distribution and excretion of dufulin racemate and its R (S)- enantiomers in rats Huaguo Chen #, §, Xin Zhou §, Baoan Song #, * #

State Key Laboratory Breeding Base of Green Pesticide and Agricultural

Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Guizhou University, Huaxi District, Guiyang 550025, China §

Guizhou Engineering Laboratory for Quality Control & Evaluation Technology of

Medicine, Guizhou Normal University, 116 Baoshan North Rd., Guiyang, 550001, China Corresponding author * Tel.: +86-851-8362-0521; Fax: +86-851-8362-2211 E-mail address: [email protected]

1

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

1

ABSTRACT: Dufulin is a plant antiviral agent with a novel molecular structure and

2

has been used widely to prevent and control tobacco and rice viral diseases. In this

3

study, an UHPLC-MS/MS method was developed for rapid determination of dufulin

4

racemate (rac-DFL) and its R (S)-enantiomers in rat plasma, tissues, urine and feces.

5

A MALDI-MSI method was further used to visual research on tissue distribution after

6

intragastric administration of the three analytes. Toxicokinetic study showed that both

7

(R)-enantiomers of dufulin ((R)-DFL) and (S)-enantiomers of dufulin ((S)-DFL) had a

8

faster ability to reach Cmax than that of rac-DFL. (R)-DFL and (S)-DFL had a similar

9

T1/2, though both were significantly lower than rac-DFL. Cmax of rac-DFL was

10

obviously higher than (R)-DFL or (S)-DFL. Meanwhile Cmax of (S)-DFL was only

11

about 60 % of (R)-DFL. Rac-DFL and its R (S)-enantiomer had a dose-dependent

12

toxicokinetic profile. Tissue distribution results revealed rac-DFL, (R)-DFL and

13

(S)-DFL mainly distributed in liver and kidney, but the maximum concentration was

14

only ng/g grade and could significant degradation within 3 hours. This indicates that

15

dufulin does not cause liver and kidney toxicity in animals. In addition, rac-DFL and

16

its R (S)-enantiomers were not been detected in brain tissue. Cumulative excretion of

17

rac-DFL and its R (S)-enantiomers within 24 h in urine and feces were less than 20 %,

18

indicating that they mainly excreted as metabolites. These results could provide

19

evidence for the in-depth toxicity evaluation of dufulin pesticide. In addition, its

20

metabolic selectivity information in vivo was also been obtained.

21

KEY WORDS: Dufulin; toxicokinetics; tissue distribution; excretions

22 2

ACS Paragon Plus Environment

Page 2 of 35

Page 3 of 35

Journal of Agricultural and Food Chemistry

23

INTRODUCTION

24

Dufulin is a new type of plant antiviral agent with a novel amino phosphate chemical

25

structure invented by Guizhou university1, and granted registration as a new chemical

26

entity by the Ministry of Agriculture of China. It has high activities on tobacco,

27

cucumber and tomato virus and has been used widely to prevent and control tobacco2,

28

3

29

agent to meet China's environmental friendly standards.

30

As a kind of highly active antiretroviral pesticide, dufulin has gained the researchers'

31

more and more interests, in the recent years. Its effectiveness, functional mechanism,

32

environmental biological toxicity and safety evaluation have been widely studied. For

33

example, Xiangyang Li et al.

34

infection of southern rice black-streaked dwarf virus. Chen Zhuo et al.2 reported the

35

protein target and mechanism of dufulin on prevent and control tobacco virus. Wang

36

Hua Zi et al6 reported the biotic and abiotic degradation of dufulin in soils.

37

Enantioselective degradation of dufulin in four types of soil was reported by Zhang

38

Kankan et al7. Fan Huitao et al8 reported the acute toxicity of dufulin and safety

39

evaluation to environmental biology. As a whole, the antiviral effect of dufulin has

40

confirmed by various kinds of studies and its environmental biological toxicity or

41

safety extensively surveyed. However, there are limited research publications

42

reporting the metabolic processes and changes of dufulin in vivo. In fact, with the

43

continuous progress and development of science and technology, the metabolic

44

processes and changes of pesticide in vivo should become one of the important

and rice viral diseases4, et al. In addition, dufulin is also the first anti-plant viral

5

reported the inhibitory effect of dufulin on the

3

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

45

indexes for pesticide’s toxicity evaluation. To evaluate the safety of a pesticide, its

46

metabolic processes and changes in vivo should not been ignored. As for dufulin, it is

47

a kind of racemic mixture and has two chemical configurations of (R)-enantiomers or

48

(S)-enantiomers. Except for metabolic processes and changes in vivo, metabolic

49

selectivity of dufulin racemate and its R (S)-enantiomers should also been taken into

50

the value.

51

Toxicokinetics is mainly concerning the absorption, distribution, metabolism,

52

excretion process and characteristics of toxic substances in animal9-11. The

53

pharmacokinetic parameters can describe the systemic exposure of poisons and the

54

relationship between dose and time, to evaluate the toxicity of a drug in different

55

species, sex, age, and physical state, such as disease or pregnancy12-15. Thus, in order

56

to clarify the metabolism process, changes and metabolic selectivity of dufulin in vivo,

57

we examined and compared the pharmacokinetics, tissue distribution and excretion of

58

dufulin racemate and its R (S)-enantiomers after oral administration in rats, by using a

59

validated UHPLC-MS/MS assay and a matrix assisted laser desorption/ionization

60

mass spectrometry imaging (MALDI-MSI) method.

61

MATERIALS AND METHODS

62

Materials Dufulin racemate (rac-DFL), (R)-enantiomers of dufulin ((R)-DFL) and

63

(S)-enantiomers of dufulin ((S)-DFL) were obtained from Guizhou University, and

64

their purity were more than 99 %. The internal standard substance (IS) of Bergenin

65

(Batch No. 20150322, purity > 99 %) was purchased from the National Institute for

66

Food and Drug Control of China. HPLC grade methanol and acetonitrile were 4

ACS Paragon Plus Environment

Page 4 of 35

Page 5 of 35

Journal of Agricultural and Food Chemistry

67

obtained from TEDIA company (USA). Formic acid of MS grade purchased from Roe

68

Scientific Inc (USA). Ultra-pure water prepared by a Millipore Milli-Q purification

69

system (Bedford, USA). All other chemicals were analytical grade.

70

Animal and experimental design

71

(weighting 250 ± 20 g ) were obtained from Changsha Tianqin Bio-technology Co.,

72

Ltd (Changsha, China, Certificate No. SCXK2016-0015). All rats need to adapt to the

73

environment (25 ± 1 °C, 12/12 h circadian cycle, free feeding and water) for at least

74

one week. The rats were required to fast over night before the experiments but could

75

supply with water. All experimental programs were been conducted according to the

76

Guide of the Care and Use of Laboratory Animal, Eighth Edition (2011) and approved

77

by the Guizhou Normal University Animal Care and Use Committee.

78

Different concentration of rac-DFL, (R)-DFL and (S)-DFL solutions for toxicokinetics,

79

tissue distribution and excretion studies were prepared by dissolving the appropriate

80

amount of control in 0.2 % DMSD aqueous solution, respectively.

81

For the toxicokinetic studies16, 108 male rats were randomly divided into 9 groups (n

82

= 12). Group 1 to 3 were intragastric administration of rac-DFL (2.5, 5.0 and 10.0

83

mg·kg-1), group 4 to 6 were orally administration of (R)-DFL (2.5, 5.0 and 10.0

84

mg·kg-1), group 7 to 9 were orally administration of (S)-DFL (2.5, 5.0 and 10.0

85

mg·kg-1), respectively. The blood samples (500 µL) were collected from the

86

suborbital vein at 0, 10, 20, 30, 40, 60, 80, 100, 120, 180, 300 and 480 min

87

respectively, and were been placed into the EP tube containing heparin sodium. After

88

blood collecting at each collection point, the equal volume of physiological saline

Adult male pathogen-free Sprague-Dawley rats

5

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 6 of 35

89

supplied in time. The plasma was been immediately separated by high-speed

90

centrifugation (2000 rpm, 10 min) for UHPLC-MS/MS analysis. Toxicokinetics

91

parameters including the elimination half-life time (T½), area under the time

92

concentration curve (AUC), apparent volume of distribution (Vd), the number of Vd

93

removed from the body within a unit time (CL) and mean retention time (MRT) were

94

calculated by non-compartmental analysis mode of Phoenix WinNonlin 6.4 (Pharsight,

95

Missouri, USA). Drug peak time (Tmax) and drug peak concentration (Cmax) acquired

96

directly from the concentration versus time curve.

97

Tissue distribution studies17 were conducted in 120 rats randomly divided into 10

98

groups (n = 12). Animal grouping and administration of Group 1 to Group 9 were the

99

same as the toxicokinetic study section. The tenth group given a physiological saline

100

orally. Tissues including liver, heart, lung, spleen, kidney and brain harvested at 60

101

min and 3 h and thoroughly eliminate blood and other interfering substances by

102

ice-cold saline rinsing, then dried with filter paper. Half of tissue samples in the

103

middle dose group were placed at -80 ºC for MALDI-MSI analysis18-21, and the other

104

tissue samples were accurately weighed and homogenized using 3 times 50 %

105

acetonitrile (v/v) solution, then kept in the freezer (-80 °C) until UHPLC-MS/MS

106

analysis.

107

Excretion studies were performed in 54 rats which were divided into 9 groups (n = 6),

108

with a same mode of administration as toxicokinetics study section

109

rat was been reared in an independent metabolic cage after orally administration of

110

rac-DFL, (R)-DFL and (S)-DFL, respectively. The sampling time of urine samples 6

ACS Paragon Plus Environment

22, 23

. Then each

Page 7 of 35

Journal of Agricultural and Food Chemistry

111

was set to 0-3, 3-6, 6-12 and 12-24 h after oral administration of the analyte, while

112

feces sample collecting was set at intervals of 0-6, 6-12 and 12-24 h. During the

113

experiment, all rats allowed to take both food and water freely. After the feces sample

114

was accurately weighed, the homogenate was treated with 3 times 50 %

115

acetonitrile(v/v) solution, then the supernatant liquor was immediately separated by

116

high-speed centrifugation (5000 rpm, 10 min) and stored at -80 °C until analysis.

117

Sample preparation for UHPLC-MS/MS analysis

118

precipitation method

119

biological samples including plasma, tissue homogenates, urine and feces supernatant

120

liquor. An aliquot of 100 µL plasma samples were transferred into the 1.5 ml EP tubes,

121

then 20 µL bergenin solution (IS, 12 µg·mL-1) was added into each tube except the

122

blank. The mixtures were vortex-mixed for 50 s and then centrifuged (15000 rpm) 8

123

min at 4 °C. After centrifugation, the supernatant was separated and a 5 µL aliquot

124

was used for UHPLC-MS/MS analysis. Other biological samples (tissue homogenates,

125

urine and feces supernatant liquor) followed with the same procedure.

126

Sample preparation for MALDI-MSI analysis

127

at -18 °C using a Cryostar NX70 freezing microtome (Thermo Fisher Scientific Ltd.,).

128

The heart, brain, and kidney tissues were been sliced in sagittal directions, and the

129

lung, spleen and liver tissues were sliced in lateral directions. Eight microns thick

130

sections were been cut and mounted onto conductive Indium Tin Oxide (ITO) glasses.

131

Matrix deposition for MALDI-MSI analysis was performed by spraying 2, 5-

132

Dihydroxybenzoic acid (30 g/L in 50 % MeOH within 0.2 % trifluoroacetic acid)

24

In this study, a simple protein

was used to remove impurities and extract the analytes from

All rat tissue sections were sliced

7

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 8 of 35

133

using the ImagePrep automated spraying device (Bruker Daltonics, Bremen,

134

Germany).

135

Preparation of stock, standard and quality control samples

136

rac-DFL (503 µg·mL-1), (R)-DFL (505 µg·mL-1) and (S)-DFL (508 µg·mL-1) were

137

prepared in methanol, and then diluted into 0.0503-5.0300 µg·mL-1, 0.0505-5.0500

138

µg·mL-1 and 0.0508-5.0800 µg·mL-1 for calibration curves, respectively. All the

139

solutions were stored at 4 °C and reverted to room temperature before analysis. The

140

final calibration standard solutions prepared by transferring appropriate amount of the

141

stock solution into 100 µL of blank biological matrices to get a series of rac-DFL

142

(concentration range from 5.03 to 5030.00 ng·mL-1), (R)-DFL (concentration range

143

from 5.05 to 5050.00 ng·mL-1) and (S)-DFL solutions (concentration range from 5.08

144

to 5080.00 ng·mL-1) in plasma, respectively. The same procedure was followed to

145

gain rac-DFL (15.09-5030.00 ng·mL-1), (R)-DFL (15.15-5050.00 ng·mL-1) and

146

(S)-DFL solutions (15.24-5080.00 ng·mL-1) in tissue homogenates, feces and urine,

147

respectively. Quality control samples (QCs) were prepared by the similar procedure,

148

to achieve three different concentrations of 15, 420, 4200 ng·mL-1 in plasma and 35,

149

400, 4000 ng·mL-1 in tissue homogenates, feces and urine.

150

UHPLC-MS/MS analysis

151

were performed on an UHPLC-MS/MS system, consisting of an Accela1250 UHPLC

152

system and a TSQ quantum ultra-triple-quadrupole mass spectrometer detector

153

(Thermo Fisher Scientific Inc., USA). Chromatographic separation was performed on

154

an Agilent XB-C18 column (2.1×150 mm, 1.7 µm) maintaining temperature at 40 °C

Stock solutions of

The quantification of rac-DFL, (R)-DFL and (S)-DFL

8

ACS Paragon Plus Environment

Page 9 of 35

Journal of Agricultural and Food Chemistry

155

and flow rate of 0.2 mL·min-1. The mobile phase system consisted of 0.1 % formic

156

acid in acetonitrile (A) and 0.1 % formic acid in water (B). The gradient elution

157

program was as follows: 0-3 min, linear change from A to B (3:97, v/v) to A-B (5:95,

158

v/v); 3-10 min, linear change from A to B (5:95, v/v) to A-B (12:88, v/v); 10-17 min,

159

linear change from A to B (12:88, v/v) to A-B (20:80, v/v). The injection volume was

160

5 µL. Mass spectrometric analyses were performed on a TSQ quantum ultra-triple-

161

quadrupole mass spectrometer (Thermo Fisher Scientific Inc., USA) equipped with an

162

electrospray ionization interface in negative mode. The three analytes and the IS were

163

detected by negative ion mode and quantified in multiple reactions monitoring (MRM)

164

mode with transitions of m/z 407.432-125.268 for rac-DFL, 407.427-125.261 for

165

(R)-DFL, 407.429-125.263 for (S)-DFL and m/z 326.978-192.152 for IS. Sheath gas

166

flow rate, auxiliary gas flow rate, spray voltage, vaporizer temperature and capillary

167

temperature were set to 40 arbitrary units, 10 arbitrary units, 2500 V, 340 °C and

168

340 °C, respectively.

169

Statistical analysis carried out by SPSS 18.0 software (SPSS, Inc., Chicago, USA).

170

Data were presented as mean ± S.D. and P value < 0.05 suggested a statistically

171

significance.

172

UHPLC-MS/MS method validation

173

biological samples (plasma, tissues, urine and feces ), according to the guide of

174

European Medical Agency (EMA) Guideline for Bioanalytical Method Validation and

175

FDA Guidance for Industry, Bioanalytical Method Validation (US-FDA, 2001). The

176

validation content including selectivity, matrix effects, calibration curves, accuracy,

The analytical method was verified on all

9

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

Page 10 of 35

177

precision and stability of the method.

178

MALDI-MSI analysis

179

MALDI Tissuetyper (Bruker, Germany) in negative-ion mode, using 600 laser shots

180

per spot and 50 × 50 µm pixel size. Data were acquired in the m/z range from 380-420.

181

Peptide Calibration Standard II (Bruker product no. #8217498) was been used for

182

external instrument calibration. The average mass spectra was been normalized to

183

their total-ion-count (TIC). Data acquisition and visualizations performed using the

184

flex software-package (flexControl 3.4; flexImaging 4.1, Bruker, Germany).

185

RESULTS AND DISCUSSION

186

Optimization of chromatography and mass spectrometry conditions

187

(R)-DFL, (S)-DFL and IS were monitored under negative ion mode and quantified in

188

multiple reactions monitoring (MRM) conditions. Under the UHPLC-MS/MS

189

conditions, there were no interfering peaks at the elution times of rac-DFL, (R)-DFL,

190

(S)-DFL and IS. Figure 1 shows the representative chromatograms of blank plasma,

191

spiked plasma containing rac-DFL, (R)-DFL, (S)-DFL and IS at LLOQ level and

192

plasma collected at 1 h after oral administration of the three analytes (5.0 mg·kg-1).

193

Retention times of three compounds and internal standard were in vicinity of 7.11 min,

194

9.66 min, 9.55 min and 12.17 min, respectively. The chromatographic condition was

195

also optimized for separation of rac-DFL, (R)-DFL, (S)-DFL and IS with higher

196

resolution. Different types of chromatographic columns were also screened, and

197

Agilent XB-C18 column (2.1×150 mm, 1.7 µm) was been considered to be the most

198

appropriate one. Different mobile phase systems, such as water-acetonitrile,

MALDI-MSI analyses were performed using a rapiflex

10

ACS Paragon Plus Environment

Rac-DFL,

Page 11 of 35

Journal of Agricultural and Food Chemistry

199

water-methanol, water (0.1 % formic acid)-methanol, water (0.1 % formic

200

acid)-methanol (0.1 % formic acid), water (0.1 % formic acid)-acetonitrile and water

201

(0.1 % formic acid)-acetonitrile (0.1 % formic acid) were evaluated for

202

chromatographic behaviors. As a result, acetonitrile (0.1 % formic acid)-water (0.1 %

203

formic acid) system was found to has a satisfactory resolution value, sharp and

204

symmetrical peaks.

205

Figure 1

206

Method validation

207

Assay linearity and sensitivity To evaluate linearity, calibration solutions of

208

rac-DFL, (R)-DFL and (S)-DFL were prepared and detected. The peak area ratios of

209

rac-DFL, (R)-DFL and (S)-DFL to IS in rat plasma and other biological samples

210

changed linearly over the concentration ranges. The calibration curves showed

211

excellent linearity with correlation coefficients (γ) ≥ 0.99 in all matrices. The

212

weighing factor 1/X2 was been selected for back calculation of nominal value because

213

it produced best linear fit with minimum bias. The regression equations for rac-DFL,

214

(R)-DFL and (S)-DFL were listed in Table 1, respectively, where Y refers to the peak

215

area ratios (analyte/IS) and X is the concentration of rac-DFL or (R)-DFL or (S)-DFL.

216

The developed method offered LLOQ of 10 ng·mL-1 in plasma, urine, feces and

217

tissues and was quantified with acceptable accuracy and precision (≤ 15 %).

218

Table 1

219

Precision and accuracy

220

rac-DFL, (R)-DFL and (S)-DFL in biological samples were assessed, according to the

The intra-day and inter-day precision and accuracy of

11

ACS Paragon Plus Environment

Journal of Agricultural and Food Chemistry

221

guidance of the US Food and Drug Administration. The intra-day and inter-day

222

precision (RSD, %) values were ≤ 10.56 % and ≤ 11.27 %, respectively. While the

223

intra-day and inter-day accuracy (%) were in the range of 92.5-107.4 % and

224

93.1-108.2 %, respectively. All the results are within the technical scope stipulated in

225

the management guidelines, indicating that the analytical method established was

226

suitable for quantitative determination of rac-DFL, (R)-DFL and (S)-DFL in rat

227

biological samples.

228

Recovery and matrix effects

229

effects for rac-DFL, (R)-DFL and (S)-DFL in biological samples. The recoveries of

230

rac-DFL, (R)-DFL and (S)-DFL in all biological samples were reproducible,

231

concentration independent and consistent. The recovery for rac-DFL, (R)-DFL and

232

(S)-DFL was between 77.65 % and 87.59 %. Meanwhile, the recovery for IS was

233

ranged from 76.95 % to 88.55 %. They were both according with the guideline for

234

validation of bioanalytical method, which issued by the U. S. Food and drug

235

administration, drug evaluation and Research Center. The matrix effects for rac-DFL,

236

(R)-DFL and (S)-DFL in all biological matrices were within ± 11.00 % with RSD %

237

of ≤ 7.78 % which was considered as negligible or insignificant matrix effects.

238

Table 2 shows the extraction recovery and matrix

Table 2

239

Stability

240

change under different storage conditions. As shown in Table 3, the stability of

241

rac-DFL, (R)-DFL and (S)-DFL in different conditions (short-term, freeze/thaw and

242

long-term stability) were within the acceptable levels (RSD % of precision values