Subscriber access provided by University of Florida | Smathers Libraries
Article
Synthesis, Characterization and Evaluation of Triptolide-Cell Penetrating Peptide Derivative for Transdermal Delivery of Triptolide Tian Tian, Yuming Song, Ke Li, Yuming Sun, and Qing Wang Mol. Pharmaceutics, Just Accepted Manuscript • DOI: 10.1021/acs.molpharmaceut.7b00914 • Publication Date (Web): 08 Jan 2018 Downloaded from http://pubs.acs.org on January 10, 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 free 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 accessible to all readers and 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.
Molecular Pharmaceutics 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 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
4
Synthesis, Characterization and Evaluation of Triptolide-Cell Penetrating Peptide Derivative for Transdermal Delivery of Triptolide
5
Tian Tian†, Yuming Song†, Ke Li†, Yuming Sun , Qing Wang†, §, *
1 2 3
‡
6 7
† School of Pharmaceutical Science and Technology, Dalian University of
8
Technology, No. 2 Linggong Road, Dalian 116024, China
9
§ State Key Laboratory of Fine Chemicals, Dalian University of Technology, No. 2
10
Linggong Road, Dalian 116024, China
11
‡ Chemical Analysis and Research Center, Dalian University of Technology, No. 2
12
Linggong Road, Dalian 116024, China
13 14 15 16 17 18 19 20 21 22
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 2 of 31
23
ABSTRACT
24
Triptolide (TP) has been used as one of the most common systemic treatments for
25
various diseases since 1960s. However, TP displays diverse side effects on various
26
organs which limits its clinical application. To overcome this issue, numerous C-14
27
hydroxyl group derivatives of TP have been synthesized. In this research, the C-14
28
hydroxyl group of TP is modified by a cell penetrating peptide poly arginine (R7). The
29
derivative TP-disulfide-CR7 (TP-S-S-CR7) containing a disulfide linkage between TP
30
and R7 possesses less toxicity at various concentrations on immortal human
31
keratinocyte
32
5-diphenyltetrazolium bromide (MTT) assay compared with free TP. Treating HaCaT
33
cells with TP (100 nM) could increase intracellular ROS (reactive oxygen species)
34
and decrease the activity of SOD (Superoxide Dismutase). Meanwhile, treating
35
HaCaT cells with equimolar concentration of TP-S-S-CR7 did not cause both of the
36
above TP-induced alterations. In addition, TP-S-S-CR7 didn’t show significant dermal
37
toxicity on guinea pigs and could efficiently overcome the barrier of corneum then
38
reached epidermis and dermis within 2 h of transdermal administration. In addition,
39
there was a relatively lower concentration of TP in blood indicates a less toxicity on
40
organs. Such results suggest that topical therapy using poly arginine is possible by the
41
transdermal delivery of TP.
42
Key words: Triptolide, Cell penetrating peptide, Toxicity reduction, Drug
43
modification
line
(HaCaT)
cell
line
by
3-(4,
44 45 46 47 48 49
ACS Paragon Plus Environment
5-dimethylthiazol-2-yl)-2,
Page 3 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
50 51
INTRODUCTION
52
Triptolide (TP), an active compound isolated from Tripterygium wilfordii Hook F
53
(TWHF), commonly called lei gong teng or thunder god vine, which displays multiple
54
bioactivities
55
anti-inflammatory, anti-cancer activities1,2. Currently, TP has been used for the
56
treatment of rheumatoid arthritis, psoriasis and leukemia by oral or intravenous route.
57
However, due to its poor water solubility and severe side effects, there are some
58
roadblocks to be applied systemically in clinic. The tissues and organs being inflicted
59
mainly include reproductive system3, liver4, kidney5 as well as heart6. Over a long
60
time, the derivatives aimed at the C-14 hydroxyl group of TP were designed and
61
synthesized to overcome this issue. For example, TRC4, a TP derivative modified
62
with amine ester is water soluble, which remains potent anticancer activity without
63
affecting the growth of normal cells7. Yutaka Aoyagi et al also synthesized a series of
64
C-14-hydroxyl group derivatives, which are more effective than free TP against A549
65
and HT29 cell lines8. Furthermore, TP-LZM, a C14-hydroxyl group derivative, can
66
solute in water, more importantly, this derivative causes less toxicity in Normal Rat
67
Kidney cell line (NRK-52E)9. Moreover, transdermal delivery system can bypass
68
hepatic first pass metabolism and reduce the incidence or severity of gastrointestinal
69
reactions10. Therefore, transdermal delivery system could also reduce the toxicity of
70
TP. Owing to these reasons, there is no doubt that developing the transdermal delivery
71
of TP is imperative. Gui Chen et al. used microneedles to break the barrier of skin and
72
successfully delivered TP through the skin of rat11. However, microneedles could
73
cause micrometer-scale openings on corneum thus causing skin irritation. Based on
74
these results, developing a biological enhancement technology for TP is necessary.
75
Cell-penetrating peptides (CPPs) such as Antp, TAT, PEP-1 and polyarginine12, is a
76
new kind of drug carrier, could enhance the transdermal drug delivery without skin
77
irritation. CPPs are first discovered in 198813,
78
therapeutic molecules (nucleic acids, drugs, imaging agents) into cells and tissues in a
such
as
immunosuppressive,
anti-fertility,
14
anti-cystogenesis,
and have successfully delivered
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 4 of 31
79
nontoxic manner. Recently, a number of interdisciplinary studies proved that CPPs
80
have successfully carried various cargos such as nucleic acids15, polymers16,
81
oligonucleotides17, liposomes18, nanoparticles19, and low molecular drugs20 across
82
cyto-membrane or other bio-membrane21. Moreover, it is reported that a chain of
83
arginines compose one of the most widely used CPPs and more effective than others22.
84
Poly arginine display many structural and functional advantages over other CPPs may
85
because the peptides are positively charged and they interact with critical membrane
86
components needed for penetration. Poly arginine (11R) covalently attaching with
87
hydroquinone successfully cured UV-induced pigmentation by transdermal delivery.
88
Such results demonstrates that topical therapy using the conjugation with poly
89
arginine is thought to be possible for the delivery of small molecular drugs by
90
transdermal route23.
91
The aim of this work is to design and synthesize a TP-poly arginine derivative
92
(TP-S-S-CR7)
93
hydrochloride as linkers. TP-S-S-CR7 showed lower cell toxicity on immortal human
94
keratinocyte (HaCaT) cell line at different concentrations than TP, meanwhile,
95
TP-S-S-CR7 could also cause less dermal toxicity on guinea pigs compare to free TP.
using
succinic
anhydride
and
pyridyl
disulfide
cysteamine
96 97
EXPERIMENTAL SECTION
98
2.1 Animals
99
Male Sprague-Dawley (SD) rats and guinea pigs and Kunming (KM) mice used in all
100
experiments were supplied by Dalian Medical University (Dalian, China). The
101
animals were kept in an animal room with regulated temperature of 20 ± 2 °C and
102
relative humidity of 60 ± 10%. All of them were given free access to food and water.
103
2.2 Synthesis of pyridyl disulfide cysteamine hydrochloride
104
The synthesis procedure of pyridyl disulfide cysteamine hydrochloride is described as
105
follow. An amount of 10 ml methanol containing 1.04 g cysteamine hydrochloride
106
was added to a solution of 2, 2’-dithiodipyridine (4 g) in 16 ml of methanol and 0.6 ml
107
acetic acid dropwisely. The yellow solution was stirred overnight at room temperature. ACS Paragon Plus Environment
Page 5 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
108
Then the solvent was evaporated off. The product was obtained by precipitation in
109
500 ml diethyl ether. After filtration, the residue was dissolved in 15 ml of methanol
110
and again precipitated in 500 ml of diethyl ether. The yellow solid was obtained after
111
drying under reduced pressure. The structure of pyridyl disulfide cysteamine
112
hydrochloride was characterized by 1H-NMR.
113
2.3 Synthesis of TPS
114
An amount of TP 100 mg and succinic anhydride 320 mg were dissolved in 2 ml
115
anhydrous methylene chloride (CH2Cl2) followed by the addition of 1.5 ml CH2Cl2
116
containing 4-dimethylaminopyridine (DMAP) 360 mg and trimethylamine 0.5 ml
117
dropwisely. The reaction stirred at room temperature for 24 hours. The reaction was
118
monitored by TLC with the solvent system of ethyl acetate-petroleum ether (1:1, v/v),
119
and the light purple spot was detected by soaking in 2% 3,5-dinitrobenzene in ethanol
120
and 10% potassium hydroxide in methanol9. After evaporating the solvent away, the
121
crude product was washed by cold carbon tetrachloride (CCl4) three times. The CCl4
122
was removed and then dried under reduced pressure, yellow powder was obtained.
123
The structure of TPS was characterized by 1H-NMR, 13C-NMR and HRMS.
124
2.4 Synthesis of TPSP
125
Amount of TPS 120 mg, pyridyl disulfide cysteamine hydrochloride 61 mg were
126
dissolved in 0.8 ml tetrahydrofuran containing 0.4 ml triethylamine. Then 54 mg
127
4-(4,6-Dimethoxy-1,3,5-triazin-2-yl)-4-methylmorpholinium
128
was added to the solution. At last, 1.8 ml tetrahydrofuran was added. The reaction
129
mixture was stirred at room temperature for 24 hours. After removing the solvent
130
away, the crude product was purified by preparative chromatography (LC-20AP,
131
Shimadzu, Japan) using C18 chromatographic column (ZORBAX SB-C18, 21.2×
132
150 mm,Agilent, America, 10-100% B for 30min, A: 0.1% trifluoroacetic acid in
133
water, B: 0.1% trifluoroacetic acid in acetonitrile, flow rate = 7ml/min). After
134
removing the solvent away light yellow spongy solid was obtained. The structure of
135
TPSP was characterized by 1H-NMR, 13C-NMR and HRMS. The purity of TPSP was
136
determined by HPLC-UV.
ACS Paragon Plus Environment
chloride
(DMTMM)
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
137
2.5 Synthesis of TP-S-S-CR7
138
Amounts of TPSP 60 mg, R7C (adding a cysteine on the N-terminal of R7)·8TFA 203
139
mg were dissolved in 3 ml DMF. Then the reaction solution was stirred for 48 hours at
140
room temperature. After completion of the reaction, the solvent was evaporated off,
141
the crude product was purified by preparative chromatography using SB-C18
142
chromatographic column (10-100% solvent B; 30min, flow rate = 7ml/min; solvent A:
143
0.01% trifluoroacetic acid in water, solvent B: 0.01% trifluoroacetic acid in
144
acetonitrile). After removing the solvent away, white powder was obtained. The
145
structure of TP-S-S-CR7 was characterized by 1H-NMR, HRMS and HRMS2. The
146
proposed fragmentation pathway of TP-S-S-CR7 was simulated using Mass Frontier
147
7.0. The purity of TP-S-S-CR7 was determined by HPLC-UV.
148
2.6 Cell culture
149
HaCaT cell line was kindly offered by Professor Wenli Li (School of Life Science and
150
Biotechnology, Dalian University of Technology) was cultured in Dulbecco modified
151
Eagle medium (DMEM) high glucose containing 10% fetal bovine serum (FBS) and 1%
152
penicillin/streptomycin solution (100 IU/ml penicillin; 100 µg/ml streptomycin) in an
153
incubator at 37 °C with 5% CO2, 95% air, and 90% relative humidity and offered
154
fresh medium every other day.
155
2.7 Cytotoxicity assay of TP and TP-S-S-CR7 on HaCaT cell line
156
The effect of TP-S-S-CR7 and TP on HaCaT cell viability was measured at different
157
concentrations using MTT assay. HaCaT cells (7×103 cells/well) were seeded in
158
96-well tissue culture plates. After 24 h, the medium was replaced with fresh medium
159
containing various concentrations of TP, TP-S-S-CR7 (10, 1, 0.1, 0.01, 0.001 µM) or
160
medium containing 0.5% dimethyl sulfoxide (DMSO). After incubation for 24 h, 110
161
µL of 500 µg/ml MTT was added, and the plates were incubated for another 4 hours.
162
Thereafter, the medium was removed and replaced with 150 µL of DMSO, the optical
163
density (OD) was measured at 490 nm using a microplate reader. The cytotoxic
164
effects of tested agents were expressed as the 50% inhibiting concentration (IC50), and
165
the IC50 values were calculated using SPSS. ACS Paragon Plus Environment
Page 6 of 31
Page 7 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
166
2.8 The stability of TP-S-S-CR7 in HaCaT cell homogenate, rat skin
167
homogenate and rat plasma
168
The HaCaT cells were seeded in the culture plate (diameter was 10 cm), after 80% -
169
90% confluent, cells and cell culture medium were then separated. Cells were then
170
washed with cold PBS 3 times and collected using cell scraper with 1 ml cold PBS in
171
a 10 ml Eppendorf tube. Then 500 µl (1 mg/ml) TP-S-S-CR7 was added to 500 µl of
172
the cell homogenate and mixed then incubated at 37 ℃. After 24 h, an aliquote of the
173
sample (100 µl) was withdrawn from the solution and equal volume acetonitrile was
174
added to stop the reaction. The samples were analyzed by HPLC-HRMS. (n=3)
175
The abdominal skin was obtained soon after the rat was executed. Then the skin
176
samples (200 mg) were cut into small pieces in a 10 ml centrifuge tube, 2.0 ml of PBS
177
was added. The samples were homogenized at 18000 rpm for about 1 min, and the
178
supernatant was obtained by centrifugation at 6×103 g for 5 min. Then 1 ml
179
TP-S-S-CR7 (1 ml) was added to 1ml of the skin homogenate and mixed then
180
incubated at 37℃. Then after 1h, an aliquote of the sample (100 µl) was withdrawn
181
from the solution and equal volume acetonitrile was added to stop the reaction. The
182
amount of TP-S-S-CR7 was detected by HPLC-HRMS. (n=3)
183
Blood was collected from adult male SD rats from orbital plexus to heparinized tubes,
184
then the plasma was separated immediately by centrifugation (6×103 g, 10 minutes).
185
Immediately after the collection of plasma, 1 ml TP-S-S-CR7 (1 mg/ml) was added to
186
1ml plasma. Then an aliquote of the sample (100 µl) was withdrew from the solution
187
and equal volume acetonitrile was added to stop the reaction. The amount of
188
TP-S-S-CR7 was detected by HPLC-HRMS. (n=3)
189
2.9 The stability of TP-S-S-CR7 in glutathione (GSH) solution
190
1 ml TP-S-S-CR7 (1 mg/ml) was mixed with 1 ml (200 µg/ml or 2 µg/ml) GSH in
191
PBS and incubated at 37°C for 24 h. Aliquots were withdrawn from the solution at
192
specified time points (0, 1, 3, 6, 12 and 24 h) and analyzed by HPLC-UV method, and
193
the corresponding degradation products were identified by HPLC-HRMS method.
194
(n=3) ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
195
2.10 Intracellular ROS determination
196
DCFH-DA, a lipophilic dye was used to determine the intracellular accumulation of
197
ROS. HaCaT cells were seeded on 6-well plate until about 80%-90% confluent.
198
Subsequently, the cells were treated with 100 nM TP, TP-S-S-CR7 or fresh medium
199
for 12 h respectively. After that, the medium was removed; the cells were washed
200
twice with PBS. Then, cells were incubated with DCFH-DA (20 µM) for about half an
201
hour at 37 ℃ in dark. Then washed with serum free fresh DMEM 3 times.
202
Fluorescence was detected with flow cytometry. Excitation: λ 488 nm and Emission:
203
λ 520 nm. (n=3)
204
2.11 SOD activity determination
205
HaCaT cells were seeded in a 6-well plate until about 80% - 90% confluent. Then
206
cells were exposed to TP, TP-S-S-CR7 or fresh medium (as negative control) for 12h.
207
After that, the medium was removed, and then the cells were washed twice with cold
208
PBS (pH = 7.4, NaCl: 8 g/L, Na2HPO4: 2.86 g/L, KH2PO4: 0.2 g/L, KCl: 0.19 g/L).
209
Through tissue homogenized on ice, centrifuged at 1×104 g for 15min at 4℃, the
210
total intracellular protein could be obtained. Total protein of each sample was
211
determined by the BCA Protein Assay Kit. SOD activity was determined using SOD
212
Detection Kit. (n=3)
213
2.12 Skin irritation test in guinea pigs
214
6 guinea pigs (200-300 g) were used for this skin irritation test. Three days prior to
215
application, the hair on the dorsal area was removed using clipper and shaver, then the
216
belly skin of each guinea pig was divided into three areas (3 × 3 cm2). Then 100
217
µg/kg/day TP or 722 µg/kg/day TP-S-S-CR7 (equimolar concentration) and equal
218
volume of PBS were smeared on each depilated skin areas for seven days
219
successively24. 24 h after the last administration, collect each area then fixed in 4%
220
paraformaldehyde for at least 24 h. Tissue sections (5 µm) were prepared by mounted
221
on common slides, and then stained with hematoxylin and eosin.
222
2.13 In vivo penetration experiment
ACS Paragon Plus Environment
Page 8 of 31
Page 9 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
223
Six male SD rats (weighing 150-200 g) were randomly divided into two groups. 24
224
hours prior to the experiment, the hair on the dorsal area was removed using clipper
225
and shaver. Then TP at a dose of 3 mg/kg or TP-S-S-CR7 at a dose of 21 mg/kg
226
(equimolar concentration) were smeared onto the dorsed areas of rats (3×3 cm2).
227
Blood samples were collected from the retro-orbital plexus into heparinized tubes at
228
designated time intervals (0.5, 1, 1.5, 2, 3, 4, 6, 8, 12, 24, 36, 48 and 72 hours) post
229
administration then plasma was separated immediately by centrifugation (6×103 g, 10
230
minutes). The plasma samples were stored at -40℃ until analysis. Plasma sample 50
231
µL was mixed with 50 µL of internal standard solution (200 µg/ml triamcinolone
232
acetonide in methanol), then 50 µL of methanol was added and vortexed for 1 minute.
233
After centrifugation at 6×103 g for 5 min, the supernatant was transferred to a new
234
tube. 20 µL of each sample was injected into the LC-MS system for quantitative
235
analysis.
236
Separation was performed on an Agilent Zorbax SB-C18 column (4.6×150 mm, 5
237
µm, Agilent Technologies, USA). The mobile phase was composed of 40-80% B for 6
238
min, then 80-100% B for 1min, then 100% B for 1min (A: water, B: acetonitrile),
239
delivered at a flow rate of 0.5 ml/min.
240
Thermo LTQ orbitrapXL mass spectrometer with high resolution was operated in ESI
241
positive mode. The parameters were as follows: vaperizer temperature 350 ℃; sheath
242
gas flow rate 30 Arb; auxiliary gas flow rate 10 Arb; ion spray voltage 3.5 kV;
243
capillary temperature 350 ℃; capillary voltage 39 V. The resolution was set at 30000
244
with mass error tolerance of ±5 ppm. The retention times of TP and internal standard
245
(triamcinolone acetonide) were 7.01 and 9.69 minutes. Over the range of 1.25-200
246
ng/ml, TP concentrations were linearly proportional to the area ratio of TP/internal
247
standard. Calibration curves were constructed for TP is y=-0.000136353 +
248
0.00144784x (R=0.999).
249
2.14 In vivo skin retention experiment
250
Twenty-four KM male mice weighing 20-25 g were randomly divided into two groups.
251
Sodium sulfide 4 g, Amidulin 3.5 g, sucrose 2 g, Sodium tetraborate 0.5 g, Glycerin
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
252
2.5 g were carefully weighed and dissolved in 50 ml water and stirred to obtain a
253
homogeneous solution. The hairs on the back of the mice were lightly rubbed with a
254
pledget pre-wetted in the above solution. After all the hairs were removed, the
255
remaining solution on the back of the animal was washed away with warm water. The
256
treated animal was kept for at least 48 h to ensure that no visible defect was present25.
257
Then TP at a dose of 4.3 mg/kg or TP-S-S-CR7 at a dose of 30 mg/kg (equimolar
258
concentration) were smeared onto the dorsed areas of mice (2 cm2). After 2, 6, 12
259
hours of administration, the animals were executed and the administration areas of
260
skin were collected. (n=3)
261
The corneum of the skin samples were removed and then the skin samples (100 mg)
262
were cut into small pieces in a 10 ml centrifuge tube, 1.0 ml of methanol was added.
263
The samples were homogenized at 18000 rpm for about 1 min, and the supernatant
264
was obtained by centrifugation at 6×103 g for 5 min26. The supernatant was treated
265
using the protocol used for the plasma samples and analyzed by HPLC-MS method
266
which was the same as that for plasma samples.
267 268
RESULTS
269
3.1 Synthesis and characterization of pyridyl disulfide cysteamine
270
hydrochloride
271
To make the amino acid cysteine at the N-terminal of poly arginine accessible for TP,
272
the corresponding sulfydryl-reactive functionalities had to be introduced. In this
273
research, we choose pyridyl disulfide cysteamine hydrochloride because it is widely
274
used and easy to be synthesized27. The synthesis procedure is described on Scheme 1.
275
The productivity is 74%. 1H NMR (400 MHz D2O): δ = 3.15-3.08 (2H, t), 3.40-3.33
276
(2H, t), 7.37-7.31 (1H, m), 7.77-7.72 (1H, d), 7.87-7.80 (1H, t-d), 8.49-8.44 (1H, m).
277
The spectrum of pyridyl disulfide cysteamine hydrochloride is shown in supporting
278
information.
279
3.2 Synthesis and characterization of TPS
ACS Paragon Plus Environment
Page 10 of 31
Page 11 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
280
TP has an active hydroxyl group at C-14 position, which can be conjugated to poly
281
arginine via spacers. In this research, we choose succinic anhydride as the first spacer.
282
The schematic synthetic route is shown in Scheme 2. The productivity is 94%.
283
1
284
3.54-3.51 (1H, d), 3.47-3.42 (1H, d), 2.84-2.62 (5H, m), 2.34-2.25 (1H, d), 2.21-2.04
285
(2H, m), 1.95-1.82 (2H, m), 1.61-1.47 (1H, dd), 1.30-1.13 (1H, m), 1.05-1.01 (3H, s),
286
0.95-0.90 (3H, d), 0.85-0.79 (3H, d). HRMS m/z 461.1807 [M+H]+. The spectrums of
287
TPS are shown in supporting information.
288
3.3 Synthesis and characterization of TPSP
289
In order to modify TPS with 2-(methyldisulfanyl) pyridine residue, pyridyl disulfide
290
cysteamine hydrochloride is chosen as the second spacer. DMTMM is chosen as
291
catalyst, because it is insoluble in THF, thus easy to remove after the reaction. The
292
reaction procedure was described in scheme 3. The productivity is 50%. 1H-NMR
293
(400 MHz, CDCl3): 8.56-8.48 (1H, s), 7.67-7.58 (1H, s)7.58-7.50 (1H, d), 7.21-7.07
294
(2H, s), 5.11-5.04 (1H, s), 4.71-4.62 (2H, s), 3.88-3.78(1H, s), 3.62-3.43 (4H, t),
295
2.96-2.52 (8H, m), 2.39-2.23 (1H, m), 1.97-1.79 (2H, d), 1.56-1.50 (1H, m), 1.34-1.19
296
(1H, m), 1.06-1.00 (3H, s), 0.96-0.91 (3H, d), 0.84-0.79 (3H, d). HRMS m/z 629.1984
297
[M+H]+. The spectrums of TPSP are shown in supporting information.
298
3.4 Synthesis and characterization of TP-S-S-CR7
299
As previously studied, adding a cysteine to the C-terminal of CPPs could not affect
300
their activities28. So, in this study, we add a cysteine to the N-terminal of poly arginine
301
to offer a sulfydryl in order to react with TPSP. The reaction procedure is described on
302
scheme 4. The productivity is 60% and the purity is 98%. HRMS m/z 289.4912 (z=6),
303
347.1882 (z=5), 433.7333 (z=4), 577.9754 (z=3), 866.4596 (z=2). The structure of
304
TP-S-S-CR7 is shown in Scheme 5. The spectrums of HRMS2, the proposed
305
fragmentation pathways and 1H-NMR of TP-S-S-CR7 are shown in supporting
306
information.
307
3.5 Cytotoxicity of TP and TP-S-S-CR7 on HaCaT cell line
308
HaCaT cell line is frequently used as a paradigm for skin29, so in this study, we
H-NMR (400 MHz, CDCl3): 5.06-5.05 (1H, s), 4.72-4.61 (2H, s), 3.83-3.80 (1H, d),
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
309
choose HaCaT cell line as a cellular model to investigate the dermal toxicity of TP
310
and TP-S-S-CR7 in vitro using MTT assay. The cells were treated with
311
various concentrations of TP and TP-S-S-C-R7 for 24 h. A dose-dependent effect of
312
TP and TP-S-S-CR7 could be observed on HaCaT cell line (Figure 1). The IC50 value
313
of TP on HaCaT cell line for 24 h was 1.00 µM, while the IC50 value of TP-S-S-CR7
314
was 9.06 µM indicating that TP-S-S-CR7 can reduce dermal toxicity in vitro,
315
especially at 10, 1 and 0.1 µM cell viability can be significantly improved. Moreover,
316
poly-arginine has no cell toxicity (data not shown). Moreover, from the result of the
317
stability of TP-S-S-CR7 in HaCaT cell homogenate, TP-S-S-CR7 was found to
318
degrade into TP. But only part of TP-S-S-CR7 was degraded. This perhaps was
319
because the lower esterase concentration in HaCaT cell line and further led to the
320
result of lower cytotoxicity.
321
3.6 Induction of oxidative stress in HaCaT cells by TP and
322
TP-S-S-CR7
323
As previously reported, TP could induce intracellular ROS accumulation and then
324
lead to tissue damage. So in this research, we attempt to investigate whether
325
TP-S-S-CR7 could cause oxidative damage on HaCaT cells as TP does. The cells were
326
treated with 100 nM TP, TP-S-S-CR7 and fresh medium for 12 h. Then, intracellular
327
ROS level as well as SOD activity were measured. As shown in Figure 2, compared
328
with the control group, TP exposure increased the accumulation of intracellular ROS.
329
Meanwhile, the intracellular SOD activity decreased significantly. However,
330
TP-S-S-CR7 treated cells have approximated the same intracellular ROS level and
331
SOD activity as the control group (Figure 2 and 3). Such results indicated that
332
TP-S-S-CR7 didn’t show significant oxidative damage on HaCaT cell line.
333
3.7 Skin irritation test in guinea pigs
334
To further investigate whether TP-S-S-CR7 has less skin irritation, 0.1 mg/kg/day TP,
335
0.722 mg/kg/day TP-S-S-CR7 and equal volume of PBS(pH=7.4 containing 0.1%
336
DMSO)were smeared on the belly skin of guinea pigs for seven days consecutively.
337
After smearing for three days, red and swollen appeared on the area of TP, then after ACS Paragon Plus Environment
Page 12 of 31
Page 13 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
338
six days of TP treatment, scab could be observed. Whereas, no obvious difference can
339
be observed between TP-S-S-CR7 group and control group (Figure 4). TP treatment
340
could induce severe damage around corneum and hair follicle. Never the less, there’s
341
no obvious histopathological changes in TP-S-S-CR7 treated group.
342
3.8 In vivo skin retention and penetration analysis
343
The in vivo skin retention and penetration experiments were performed to explore
344
whether TP-S-S-CR7 is suitable for topical therapy by transdermal route. TP 4.3
345
mg/kg or TP-S-S-CR7 at a dose of 30 mg/kg were transdermal administrated to KM
346
rats. As shown in Figure 8, TP could be detected at 2 h after administration in both TP
347
and TP-S-S-CR7 administration groups, the concentrations of TP in epidermis and
348
dermis in TP administration group and TP-S-S-CR7 administration group are as high
349
as 6352.22 ng/g and 3647.80 ng/g. Such result demonstrates that even through the
350
water solubility of TP has been significantly improved, owing to the presence of poly
351
arginine, the transdermal ability of TP has not been significantly affected. TP-S-S-CR7
352
also can be detected at 2 h, which could probably indicates TP could release in the
353
epidermis and dermis in a slow rate. So the effective concentration of TP may last
354
longer.
355
3mg/kg TP or 21 mg/kg TP-S-S-CR7 were transdermal administration to SD rats to
356
detect the concentration of TP in blood. As shown in Figure 8, TP could not be
357
detected in TP administrated group. In TP-S-S-CR7 administration group, a relatively
358
low concentration of TP could be detected. The highest concentration is 13.52 ng/ml,
359
which could cause less systematic toxicity on organs. Such results proves that
360
TP-S-S-CR7 has the potential for topical therapy by transdermal delivery of TP.
361 362
DISCUSSION
363
4.1 Design and synthesis of TP-S-S-CR7
364
Low molecular weight proteins, which like peptides afford good biodegradability in
365
vivo, have been employed as drug carriers for transdermal delivery of therapeutic
366
drugs. Since the guanidine of arginine is proposed to be crucial for achieving ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Page 14 of 31
367
penetration efficacy30. So in this study, we choose poly arginine to modify the
368
C-14-hydroxyl group of TP to investigate whether TP-CPP conjugate could be
369
designed and synthesized, at the same time with the ability to achieve transdermal
370
delivery, meanwhile, reduce the dermal toxicity of TP. Too much amino acids on the
371
side chains of poly arginine makes it difficult to conjugate TP and R7 directly. So it is
372
imperative to induce linkers. As there is a cysteine on the C-terminal of poly arginine,
373
the most direct linker would be 3-(pyridine-2-yldisulfancyl) propanoic acid31.
374
However, it changes into 1,2-di (pyridine-2-yl) disulfane during the reaction in
375
CH2Cl2 at room temperature at the presence of 1-(3-Dimethylaminopropyl)-
376
3-ethylcarbodiimide
377
3-(pyridine-2-yldisulfancyl) propanoic acid with benzotriazole, the productivity was
378
as low as 20%. So in this research, succinic anhydride and pyridyl disulfide
379
cysteamine hydrochloride are chosen as linkers to conjugate TP and poly arginine.
380
Because the derivative could be obtained by three steps from the natural product,
381
scale-up of this method for clinical use of the prodrug does not pose a problem.
382
4.2 The toxicity evaluation of TP and TP-S-S-CR7 on HaCaT cell line
383
Oxidative stress, which is caused by the over-accumulation of ROS, induces protein
384
damage and aggregation, leading to impaired cellular homeostasis32. For over a
385
decade, an increasing number of works have reported that oxidative stress is a
386
recognized mode of toxic effects of TP exposure which has been observed in vitro and
387
in vivo. TP exposure has been shown to increase the generation of anion superoxide
388
and inhibit the activity of such antioxidant enzymes as SOD and subsequently induced
389
oxidative stress in the liver, kidney and heart tissues33. So in this study, we investigate
390
whether TP-S-S-CR7 could cause less cytotoxicity on HaCaT cells. The IC50 of
391
TP-S-S-CR7 is about nine-fold higher than that of TP, which indicates that
392
TP-S-S-CR7 could decrease dermal caused by TP in vitro (Figure 1). Based on such
393
result, we further investigated whether TP-S-S-CR7 could cause less ROS
394
accumulation and SOD activity decrease on HaCaT cell line. The results indicated
395
that there’s obvious increase of intracellular ROS of HaCaT cells at the presence of
hydrochloride
(EDC)
and
ACS Paragon Plus Environment
DMAP.
Even
modified
Page 15 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
396
TP (Figure 2). Simultaneously, the activity of SOD significantly decreased.
397
TP-S-S-CR7 treated group did not display such obvious negative effect.(Figure 3).
398
Our results prove that the mechanism of dermal toxicity is in accordance with the
399
toxicity mechanism on other organs.
400
4.3 In vivo dermal toxicity evaluation of TP and TP-S-S-CR7
401
The toxicity of TP has been demonstrated in several experimental animal species. The
402
tissues and organs affected by toxicity of TP include gastrointestinal tract, liver,
403
kidney, heart, blood cells, bonemarrow, testes and ovaries. But the toxicity of skin is
404
rarely investigated34. Obvious toxicities of TP were found toward the kidney and
405
testicles of mice, and these have even resulted in the death of mice at intraperitoneal
406
doses of 0.025, 0.05 and 0.1 mg/kg. As previously reported, obvious hepatotoxicity
407
was also observed at an oral dose of 0.2 mg/kg in mice. It has also been described in
408
rats with oral dosages as low as 0.1 mg/kg, in dogs at intravenous dosages of 0.04
409
mg/kg, and in rabbits at an external concentration of 1.11 mmol/L34. Guinea pigs are
410
commonly used as animal models for skin irritation evaluation35, 34. So in this study,
411
we choose guinea pigs to conduct the comprehensive dermal toxicity study of TP and
412
TP-S-S-CR7 at transdermal delivery dosage as low as 0.1 mg/kg/day TP and 722
413
mg/kg/day TP-S-S-CR7 for 7 days. As shown in Figure 4, TP exposure could cause
414
significant dermal toxicity on corneum and hair follicle. However, no obvious toxicity
415
could be observed in TP-S-S-CR7 smeared group, which provide a promising result
416
for the transdermal delivery of TP.
417
4.4 The degradation of TP-S-S-CR7 in HaCaT cell homogenate, rat
418
skin homogenate, rat plasma and GSH solution
419
The results of in vitro and in vivo dermal toxicity experiment prove that TP-S-S-CR7
420
could cause less dermal toxicity compare with free TP. In order to further discuss the
421
mechanism of the toxicity reduction, the metabolism activity of TP-S-S-CR7 in
422
HaCaT cell homogenate, rat skin homogenate and rat plasma was investigated.
423
In HaCaT cell homogenate, the degradation rate is not so fast, 24 h after TP-S-S-CR7
424
exposed to the cell homogenate, both TP and TP-S-S-CR7 could be detected. This may ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
425
because the esterases in HaCaT cell is not active.
426
The existing formation of TP-S-S-CR7 in skin homogenate is very important for us to
427
understand the transportation form of TP-S-S-CR7 across skin. So in this study, the
428
degradation of TP-S-S-CR7 in rat skin homogenate was investigated in vitro. After 1h
429
exposure to rat skin homogenate, about 85% of TP-S-S-CR7 turned into TP, which
430
was identified by HPLC-HRMS with mass tolerance of ±5 ppm. This may because
431
the activity of esterase is relatively high. What’s more, there’s no other TP related
432
compounds were detected. As previously reported, human esterases are highly active
433
and able to hydrolyze substances extensively during permeation through in vitro
434
human skin, such as 3-alkyl esters of naltrexone and ester derivatives of fluroxypyr36.
435
In addition, rat skin also shows relatively high esterase activity, most of the esterases
436
are located in epidemis and dermis, and the major esterase is carboxylesterase (CES:
437
EC.3.1.1.1)36. Therefore, the degradation might took place after TP-S-S-CR7
438
penetrating across the corneum into epidemis and dermis.
439
As soon as TP-S-S-CR7 exposed to rat plasma, TP was released. And there’s no other
440
TP related compounds detected during the experiment. The result of in vivo
441
penetration experiment showed that TP existed in a relatively low concentration,
442
which means a relatively low toxicity. Previous research demonstrated that
443
p-Nitrophenylacetate, a carboxylic ester containing structure was rapidly hydrolyzed
444
by rat plasma, and the hydrolase activity was as high as 2540 ± 240 µM/min, which
445
indicated the esterase activity in rat plasma is relatively high37.
446
To further investigate whether the esterase is the major factor for the degradation of
447
TP-S-S-CR7, bis-para-nitrophenylphosphate (BNPP; 1 mM), a nonspecific esterase
448
inhibitor was added to skin homogenate and rat plasma, then incubated for 30 min at
449
37 ℃ before TP-S-S-CR7 was added38. After adding BNPP into skin homogenate, TP
450
could not be released from TP-S-S-CR7. However, TP still released from TP-S-S-CR7
451
immediately after TP-S-S-CR7 exposed to rat plasma even when BNPP was added.
452
There are probably some other components in plasma which could also degrade ester
453
bond, such as albumin39,40.
454
As previously reported, reduced GSH could break disulfide bonds41. So in this study, ACS Paragon Plus Environment
Page 16 of 31
Page 17 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
455
the stability of TP-S-S-CR7 in GSH solution was investigated in vitro. The
456
degradation product was detected using HPLC-HRMS. The concentrations of GSH
457
used in this experiment are similar to the concentration of GSH in skin homogenate42
458
(200 µg/ml), HaCaT cell homogenate43 and rat plasma44 (2 µg/ml). The degradation
459
curve is shown in Figure 5 (A and B), and the degradation product (TPSP-D) is
460
formed by the cleavage of the disulfide bond (Figure 5C). The result of TP-S-S-CR7
461
stability studies indicated that TP-S-S-CR7 was degraded very quickly in rat plasma,
462
but it was stable in GSH solution within 1 h, with the GSH concentration level at 2
463
µg/ml, which was similar with that in rat plasma. This indicated that GSH is not the
464
main factor for the degradation of TP-S-S-CR7. On the other hand, about 60% of
465
TP-S-S-CR7 was degraded within 1 h when the GSH was presented at 200 µg/ml,
466
which was similar to the concentration level of that in skin homogenate. But the
467
disulfide bond was stable in skin homogenate (as described above). This indicated
468
that the degradation effect of GSH to TP-S-S-CR7 in skin homogenate was inhibited.
469
Therefore, though GSH in PBS solution can degrade TP-S-S-CR7 at the concentration
470
level of 200 µg/ml in vitro and the degradation product was identified by LC-HRMS
471
method as TPSP-D (Figure 5), it’s not the main factor for the degradation of
472
TP-S-S-CR7 in skin homogenate, HaCaT cell homogenate and rat plasma.
473
4.5 In vivo penetration profile
474
Transdermal drug delivery, which was first applied in 1979, has many advantages. For
475
example, compared with the oral route, transdermal drug delivery can avoid the
476
first-pass effect of the liver that can prematurely metabolize drugs. What’s more
477
transdermal drug delivery also has advantages over hypodermic injections, which are
478
painful, easily generate dangerous medical wastes and pose the risk of disease
479
transmission by needle re-use45. So it is imperative to develop a transdermal delivery
480
system for TP. However, there is some difficulties to overcome since TP could cause
481
severe damage to the corneum (Figure 4). Microinjection, electroporation, and
482
liposome and viralbased vectors have been used for transdermal delivery. However,
483
these methods have various drawbacks, such as low efficiency, severe toxicity,
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
484
penurious bioavailability, and poor specificity46. Using cell penetrating peptide poly
485
arginine to modify the C-14 hydroxyl group TP could not only avoid some of the
486
above drawbacks but also can achieve its topical therapy by transdermal route.
487
Both TP and TP-S-S-CR7 could overcome the barrier of corneum and accumulate in
488
epidermis and dermis. Even though TP could not be detected in TP administration
489
group (Figure 8), which means no toxicity on organs, but TP could cause severe
490
dermal toxicity limits its application for transdermal administration. Such results
491
suggest that topical therapy using poly arginine is possible by the transdermal delivery
492
of TP.
493 494
CONCLUSION
495
In this study, we designed and synthesized a water soluble TP-CPP derivative
496
TP-S-S-CR7 which has less dermal toxicity both in vitro and in vivo. Moreover,
497
TP-S-S-CR7 could cross the barrier of corneum and reach epidermis and dermis at 2 h
498
after transdermal administration which indicates TP-S-S-CR7 can realize the topical
499
therapy of TP in a transdermal way. The blood concentration of TP is as low as 13.52
500
ng/ml, which could cause less toxicity on organs such as liver and heart. In a word,
501
TP-S-S-CR7 makes topical therapy possible by the transdermal delivery of TP and
502
poses the potential to cure some disease such as rheumatoid arthritis.
503
NOTES
504
The authors declare no competing financial interest.
505
ACKNOWLEDGEMENT
506
The study has been financed by Liaoning province natural science funds (2014020015)
507
and project of outstanding talent support program in universities of Liaoning province
508
(LR2014002).
509
Associated Content
510
Supporting information
511
Materials; preparation procedures of the solutions of TP and TP-S-S-CR7 used in
ACS Paragon Plus Environment
Page 18 of 31
Page 19 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
512
MTT assay, skin irritation test in vivo penetration and the stability of TP-S-S-CR7 in
513
GSH solution experiment; HPLC analysis methods of TPSP and TP-S-S-CR7; 1H
514
NMR, 13C NMR and HRMS spectrums of pyridyl disulfide cysteamine hydrochloride,
515
TPS, TPSP and TP-S-S-CR7; proposed fragmentation pathway of TP-S-S-CR7 are
516
listed in supporting information.
517
References
518
1. Li, X. J.; Jiang, Z. Z.; Zhang, L. Y., Triptolide: progress on research in
519
pharmacodynamics and toxicology. J Ethnopharmacol.2014, 155 (1), 67-79.
520
2.
521
modifications, structure-activity relationships, bioactivities, clinical development and
522
mechanisms. Nat Prod Rep.
523
Zhou, Z. L.; Yang, Y. X.; Ding, J.; Li, Y. C.; Miao, Z. H., Triptolide: structural
2012, 29 (4), 457-75.
524
3. Hu, J.; Yu, Q.; Zhao, F.; Ji, J.; Jiang, Z.; Chen, X.; Gao, P.; Ren, Y.; Shao, S.; Zhang,
525
L.; Yan, M., Protection of Quercetin against Triptolide-induced apoptosis by
526
suppressing oxidative stress in rat Leydig cells. Chem. Biol. Interact. 2015, 240,
527
38-46.
528
4. Jin, J.; Sun, X.; Zhao, Z.; Wang, W.; Qiu, Y.; Fu, X.; Huang, M.; Huang, Z.,
529
Activation of the farnesoid X receptor attenuates triptolide-induced liver toxicity.
530
Phytomedicine. 2015, 22 (10), 894-901.
531
5. Li, J.; Jin, J.; Li, M.; Guan, C.; Wang, W.; Zhu, S.; Qiu, Y.; Huang, M.; Huang, Z.,
532
Role of Nrf2 in protection against triptolide-induced toxicity in rat kidney cells.
533
Toxicol. Lett. 2012, 213 (2), 194-202.
534
6. Zhou, J.; Xi, C.; Wang, W.; Yang, Y.; Qiu, Y.; Huang, Z., Autophagy plays an
535
important role in triptolide-induced apoptosis in cardiomyocytes. Toxicol. Lett. 2015,
536
236 (3), 168-183.
537
7. Wang, P. Y.; Zeng, W. J.; Liu, J.; Wu, Y. L.; Ma, Y.; Zeng, Z.; Pang, J. Y.; Zhang, X.
538
K.; Yan, X.; Wong, A. S.; Zeng, J. Z., TRC4, an improved triptolide derivative,
539
specifically targets to truncated form of retinoid X receptor-alpha in cancer cells.
540
Biochem. Pharmacol. 2017, 124, 19-28.
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
541
8. Aoyagi, Y.; Hitotsuyanagi, Y.; Hasuda, T.; Matsuyama, S.; Fukaya, H.; Takeya, K.;
542
Aiyama, R.; Matsuzaki, T.; Hashimoto, S., Fluorination of triptolide and its analogues
543
and their cytotoxicity. Bioorg. Med. Chem. Lett. 2008, 18 (7), 2459-63.
544
9. Zheng, Q.; Gong, T.; Sun, X.; Zhang, Z. R., Synthesis, characterization and in vitro
545
evaluation of triptolide-lysozyme conjugate for renal targeting delivery of triptolide.
546
Arch. Pharm. Res. 2006, 29 (12), 1164-1170.
547
10. Sarkar, V.; Yadav, K., Formulation and evaluation of prolonged release
548
transdermal drug delivery system of atenolol for the treatment of hypertension.
549
Pharmatutor. 2014, 2 (2), 134-140.
550
11. Chen, G.; Hao, B.; Ju, D.; Liu, M.; Zhao, H.; Du, Z.; Xia, J., Pharmacokinetic and
551
pharmacodynamic study of triptolide-loaded liposome hydrogel patch under
552
microneedles on rats with collagen-induced arthritis. Acta Pharm Sin B. 2015, 5 (6),
553
569-76.
554
12. Chang, M.; Li, X.; Sun, Y.; Cheng, F.; Wang, Q.; Xie, X.; Zhao, W.; Tian, X.,
555
Effect of cationic cyclopeptides on transdermal and transmembrane delivery of insulin.
556
Mol. Pharm. 2013, 10 (3), 951-7.
557
13. Derossi, D.; Joliot, A. H.; Chassaing, G.; Prochiantz, A., The third helix of the
558
Antennapedia homeodomain translocates through biological membranes. J. Biol.
559
Chem. 1994, 269 (14), 10444-50.
560
14. Frankel, A. D.; Pabo, C. O., Cellular uptake of the tat protein from human
561
immunodeficiency virus. Cell. 1988, 55 (6), 1189-93.
562
15. Chen, L.; Fang, S.; Xiao, X.; Zheng, B.; Zhao, M., Single-Stranded DNA Assisted
563
Cell Penetrating Peptide-DNA Conjugation Strategy for Intracellular Imaging of
564
Nucleases. Anal. Chem. 2016, 88 (23), 11306-11309.
565
16. Wang, D.; Luo, W.; Wen, G.; Yang, L.; Hong, S.; Zhang, S.; Diao, J.; Wang, J.;
566
Wei, H.; Li, Y.; Wang, Y., Synergistic effects of negative-charged nanoparticles
567
assisted by ultrasound on the reversal multidrug resistance phenotype in breast cancer
568
cells. Ultrason Sonochem. 2017, 34, 448-457.
569
17. Said Hassane, F.; Saleh, A. F.; Abes, R.; Gait, M. J.; Lebleu, B., Cell penetrating
570
peptides: overview and applications to the delivery of oligonucleotides. Cell. Mol. ACS Paragon Plus Environment
Page 20 of 31
Page 21 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
571
Life Sci. 2010, 67 (5), 715-26.
572
18. Lin, W.; Xie, X.; Yang, Y.; Fu, X.; Liu, H.; Yang, Y.; Deng, J., Thermosensitive
573
magnetic liposomes with doxorubicin cell-penetrating peptides conjugate for
574
enhanced and targeted cancer therapy. Drug Deliv. 2016, 23 (9), 3436-3443.
575
19. Olson, E. S.; Jiang, T.; Aguilera, T. A.; Nguyen, Q. T.; Ellies, L. G.; Scadeng, M.;
576
Tsien, R. Y., Activatable cell penetrating peptides linked to nanoparticles as dual
577
probes for in vivo fluorescence and MR imaging of proteases. PNAS. 2010, 107 (9),
578
4311-6.
579
20. Aroui, S.; Brahim, S.; Waard, M. D.; Kenani, A., Cytotoxicity, intracellular
580
distribution and uptake of doxorubicin and doxorubicin coupled to cell-penetrating
581
peptides in different cell lines: a comparative study. Biochem. Biophys. Res. Commun.
582
2010, 391 (1), 419-25.
583
21. He, H.; Sun, L.; Ye, J.; Liu, E.; Chen, S.; Liang, Q.; Shin, M. C.; Yang, V. C.,
584
Enzyme-triggered, cell penetrating peptide-mediated delivery of anti-tumor agents. J.
585
Controlled Release. 2016, 240, 67-76.
586
22. Mitchell, D. J.; Kim, D. T.; Steinman, L.; Fathman, C. G.; Rothbard, J. B.,
587
Polyarginine enters cells more efficiently than other polycationic homopolymers. J.
588
Pept. Res. 2000, 56 (5), 318-325.
589
23. Candan, G.; Michiue, H.; Ishikawa, S.; Fujimura, A.; Hayashi, K.; Uneda, A.;
590
Mori, A.; Ohmori, I.; Nishiki, T.; Matsui, H.; Tomizawa, K., Combining poly-arginine
591
with the hydrophobic counter-anion 4-(1-pyrenyl)-butyric acid for protein
592
transduction in transdermal delivery. Biomaterials. 2012, 33 (27), 6468-75.
593
24. Chen, H.; Chang, X.; Weng, T.; Zhao, X.; Gao, Z.; Yang, Y.; Xu, H.; Yang, X., A
594
study of microemulsion systems for transdermal delivery of triptolide. J Control
595
Release 2004, 98 (3), 427-36.
596
25. Hao, J.; Sun, Y.; Wang, Q.; Tong, X.; Zhang, H.; Zhang, Q., Effect and mechanism
597
of penetration enhancement of organic base and alcohol on glycyrrhetinic acid in vitro.
598
Int J Pharm 2010, 399 (1-2), 102-8.
599
26. Sun, Y.; Yang, Y.; Li, D.; Pu, T.; Xie, X.; Wang, Q., Effect of paeoniflorin on the
600
pharmacokinetics of glycyrrhizic acid and glycyrrhetinic acid in rats. Chinese Pharm. ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
601
J. 2012, 47, 599–603.
602
27. Lelle, M.; Peneva, K., An amino acid-based heterofunctional cross-linking reagent.
603
Amino acids 2014, 46 (5), 1243-51.
604
28. Åmand, H. L.; Nordén, B.; Fant, K., Functionalization with C-terminal cysteine
605
enhances transfection efficiency of cell-penetrating peptides through dimer formation.
606
Biochem. Biophys. Res. Commun. 2012, 418 (3), 469-474.
607
29. Schoop, V. M.; Mirancea, N.; Fusenig, N. E., Epidermal organization and
608
differentiation of HaCaT keratinocytes in organotypic coculture with human dermal
609
fibroblasts. J. Invest. Dermatol. 1999, 112 (3), 343-53.
610
30. Liou, J. T.; Chen, Z. Y.; Ho, L. J.; Yang, S. P.; Chang, D. M.; Liang, C. C.; Lai, J.
611
H., Differential effects of triptolide and tetrandrine on activation of COX-2,
612
NF-kappaB, and AP-1 and virus production in dengue virus-infected human lung cells.
613
Eur. J. Pharmacol. 2008, 589 (1-3), 288-98.
614
31. Chuan, X.; Song, Q.; Lin, J.; Chen, X.; Zhang, H.; Dai, W.; He, B.; Wang, X.;
615
Zhang, Q., Novel free-paclitaxel-loaded redox-responsive nanoparticles based on a
616
disulfide-linked poly(ethylene glycol)-drug conjugate for intracellular drug delivery:
617
synthesis, characterization, and antitumor activity in vitro and in vivo. Mol. Pharm.
618
2014, 11 (10), 3656-70.
619
32. Prakash, C.; Soni, M.; Kumar, V., Mitochondrial oxidative stress and dysfunction
620
in arsenic neurotoxicity: A review. J Appl Toxicol. 2016, 36 (2), 179-88.
621
33. Xi, C.; Peng, S.; Wu, Z.; Zhou, Q.; Zhou, J., Toxicity of triptolide and the
622
molecular mechanisms involved. Biomed. Pharmacother. 2017, 90, 531-541.
623
34. Xu, L.; Pan, J.; Chen, Q.; Yu, Q.; Chen, H.; Xu, H.; Qiu, Y.; Yang, X., In vivo
624
evaluation of the safety of triptolide-loaded hydrogel-thickened microemulsion. Food
625
Chem. Toxicol. 2008, 46 (12), 3792-9.
626
35. Frosch, P. J.; Schulzedirks, A.; Hoffmann, M.; Axthelm, I., Efficacy of skin barrier
627
creams (II). Ineffectiveness of a popular "skin protector" against various irritants in
628
the repetitive irritation test in the guinea pig. Contact Derm. 1993, 29 (2), 74-77.
629
36.Imai, T.; Ariyoshi, S.; Ohura, K.; Sawada, T.; Nakada, Y., Expression of
630
Carboxylesterase Isozymes and Their Role in the Behavior of a Fexofenadine Prodrug ACS Paragon Plus Environment
Page 22 of 31
Page 23 of 31 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
Molecular Pharmaceutics
631
in Rat Skin. J Pharm Sci. 2016, 105 (2), 714-721.
632
37. Bahar, F. G.; Ohura, K.; Ogihara, T.; Imai, T., Species difference of esterase
633
expression and hydrolase activity in plasma. J Pharm Sci. 2012, 101 (10), 3979-88.
634
38. Fu, J.; Sadgrove, M.; Marson, L.; Jay, M., Biotransformation Capacity of
635
Carboxylesterase in Skin and Keratinocytes for the Penta-Ethyl Ester Prodrug of
636
DTPA. Drug Metab Dispos. 2016, 44 (8), 1313-1318.
637
39. Jin, Q.; Feng, L.; Zhang, S. J.; Wang, D. D.; Wang, F. J.; Zhang, Y.; Cui, J. N.;
638
Guo, W. Z.; Ge, G. B.; Yang, L., Real-Time Tracking the Synthesis and Degradation
639
of Albumin in Complex Biological Systems with a near-Infrared Fluorescent Probe.
640
Anal Chem. 2017, 89 (18), 9884-9891.
641
40. Ge, G. B.; Feng, L.; Jin, Q.; Wang, Y. R.; Liu, Z. M.; Zhu, X. Y.; Wang, P.; Hou, J.;
642
Cui, J. N.; Yang, L., A novel substrate-inspired fluorescent probe to monitor native
643
albumin in human plasma and living cells. Anal Chim Acta. 2017, 989, 71-79.
644
41. Pillow, T. H.; Sadowsky, J. D.; Zhang, D.; Yu, S. F.; Del Rosario, G.; Xu, K.; He,
645
J.; Bhakta, S.; Ohri, R.; Kozak, K. R.; Ha, E.; Junutula, J. R.; Flygare, J. A.,
646
Decoupling stability and release in disulfide bonds with antibody-small molecule
647
conjugates. Chem Sci. 2017, 8 (1), 366-370.
648
42. Tunali-Akbay, T.; Alev, B.; Tunali, S.; Oktay, S.; Emekli-Alturfan, E.; Ipekci, H.;
649
Ozturk, L. K.; Yanardag, R.; Yarat, A., Protective role of edaravone against valproic
650
acid induced changes in skin. Indian J Exp Biol. 2017, 55 (5), 286-291
651
43. Inbaraj, J. J.; Chignell, C. F.; Cytotoxic action of juglone and plumbagin: a
652
mechanistic study using HaCaT keratinocytes. Chem Res Toxicol, 2004, 17(1): 55-62.
653
44. Semercioz, A.; Baltaci, A. K.; Mogulkoc, R.; Avunduk, M. C., Effect of Zinc and
654
Melatonin on Oxidative Stress and Serum Inhibin-B Levels in a Rat Testicular
655
Torsion-Detorsion Model. Biochem genet. 2017, 55 (5-6), 395-409.
656
45. Prausnitz, M. R.; Langer, R., Transdermal drug delivery. Nat. Biotechnol. 2008, 26
657
(11), 1261-8.
658
46. Copolovici, D. M.; Langel, K.; Eriste, E.; Langel, Ü., Cell-penetrating peptides:
659
design, synthesis, and applications. Acs Nano. 2014, 8 (3), 1972-94.
ACS Paragon Plus Environment
Molecular Pharmaceutics 1 2 3 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 38 39 40 41 42 43 44 45 46 47 48 49 50 51 52 53 54 55 56 57 58 59 60
660 661
Figure 1 Cell viability of HaCaT cell line at the presentence of different
662
concentrations of TP, TP-S-S-C-R7 for 24 hours (n=3) (*P