Accepted Manuscript Chidamide, a histone deacetylase inhibitor-based anticancer drug, effectively reactivates latent HIV-1 provirus Wenqian Yang, Zhiwu Sun, Chen Hua, Qian Wang, Wei Xu, Qiwen Deng, Yanbin Pan, Lu Lu, Shibo Jiang PII:
S1286-4579(17)30178-8
DOI:
10.1016/j.micinf.2017.10.003
Reference:
MICINF 4516
To appear in:
Microbes and Infection
Received Date: 19 September 2017 Revised Date:
18 October 2017
Accepted Date: 20 October 2017
Please cite this article as: W. Yang, Z. Sun, C. Hua, Q. Wang, W. Xu, Q. Deng, Y. Pan, L. Lu, S. Jiang, Chidamide, a histone deacetylase inhibitor-based anticancer drug, effectively reactivates latent HIV-1 provirus, Microbes and Infection (2017), doi: 10.1016/j.micinf.2017.10.003. 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.
1
ACCEPTED MANUSCRIPT
Chidamide, a histone deacetylase inhibitor-based anticancer drug,
1
effectively reactivates latent HIV-1 provirus
2 3
Wenqian Yang a,#, Zhiwu Sun a,#, Chen Hua a, Qian Wang a, Wei Xu a, Qiwen Deng b,
5
Yanbin Pan c, Lu Lu a,*, Shibo Jiang a,b,d,*
RI PT
4
6 a
Key Lab of Medical Molecular Virology of Ministries of Education and Health, School of
SC
7
Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University,
9
Shanghai, China b
10
Shenzhen Nanshan People's Hospital of Shenzhen University, Shenzhen 518052, China c
11 d
Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
#
These authors have equal contribution.
13
EP
14 15
Aris Pharmaceuticals Inc., Bristol, PA19007, USA
TE D
12
M AN U
8
AC C
16 17
*Address for correspondence: Shibo Jiang:
[email protected]; Lu Lu:
18
[email protected].
19 20 21 22
2
ACCEPTED MANUSCRIPT 23
Abstract Although combination antiretroviral therapy (cART) is highly effective in suppressing
25
human immunodeficiency virus type 1 (HIV-1) replication, it fails to eradicate the virus from
26
HIV-1-infected individuals because HIV-1 integrates into the resting CD4+ T cells,
27
establishing latently infected reservoirs. Histone deacetylation is a key element in regulating
28
HIV-1 latent infection. Chidamide, a new anticancer drug, is a novel type of selective histone
29
deacetylase inhibitor. Here we showed that chidamide effectively reactivated HIV-1 latent
30
provirus in different latently infected cell lines in a dose- and time-dependent manner.
31
Chidamide had relatively low cytotoxicity to peripheral blood mononuclear cells (PBMCs)
32
and other latent cell lines. We have demonstrated that chidamide reactivates HIV-1 latent
33
provirus through the NF-κB signaling pathway. The replication of the newly reactivated
34
HIV-1 could then be effectively inhibited by the anti-HIV drugs Zidovudine, Nevirapine, and
35
Indinavir. Therefore, chidamide might be used in combination with cART for functional
36
HIV-1 cure.
37
Key Words: Histone deacetylase inhibitor; Chidamide; HIV-1 latent infection
SC
M AN U
TE D
EP AC C
38
RI PT
24
3
ACCEPTED MANUSCRIPT 39
1. Introduction Combination antiretroviral therapy (cART) has succeeded in reducing human
41
immunodeficiency virus type 1 (HIV-1) load to undetectable levels in most patients [1], but
42
the therapy must be maintained throughout life. Once cART is withdrawn, viral loads could
43
return to even pretreatment levels within weeks. One of the principal obstacles to a thorough
44
cleaning of the virus is the integration of HIV-1 genome into that of the resting CD4+ T cells
45
during acute infection, thereby establishing the latently infected reservoir that harbors
46
transcriptionally silent proviruses refractory to current cART [2-4]. Therefore, the
47
development of HIV-1 therapeutic strategies to eliminate this latent reservoir is urgently
48
needed.
M AN U
SC
RI PT
40
Some host cell-mediated molecular mechanisms maintain the quiescence of HIV-1 gene
50
expression, including epigenetic silencing of the proviral promoter, transcription complex
51
formation, transcript initiation, and transcription complex processivity, all of which might act
52
as therapeutic targets to disrupt HIV-1 latency [5-8]. Among these mechanisms, histone
53
deacetylase (HDAC)-mediated chromosomal suppression of HIV-1 long terminal repeat (LTR)
54
is a key regulator leading to the establishment of HIV-1 latency [7, 9]. It was reported the host
55
transcription factors comprised of NF-κB family members are involved in HDAC1
56
recruitment [10]. HDAC inhibitors have been shown to increase cell-associated HIV-1 RNA
57
production and/or plasma viremia in vivo [6, 11, 12], suggesting that they could play a role in
58
reactivating latent HIV-1 provirus.
AC C
EP
TE D
49
59
A strategy known as ‘shock and kill’, or ‘kick and kill’, has been developed for HIV-1
60
functional cure [13, 14]. To explain, the latent HIV-1 provirus was first reactivated with some
61
small-molecule HIV-1 latency-reversing agents (LRAs) [15] and then killed by the patient’s
62
immune system or suppressed by cART. Clinical trials have demonstrated that administration
63
of HDAC inhibitors, such as vorinostat [16, 17] or panobinostat [18], to the patients in whom
4
ACCEPTED MANUSCRIPT
viremia was fully suppressed by cART, resulted in a significant increase in HIV-1 RNA
65
production in resting CD4+ cells. Another clinical trial had shown that the HDAC inhibitor
66
romidepsin was effective in disrupting HIV-1 latency in vivo [19]. However, some LRAs may
67
be toxic and mutagenic or harmful to host brain during prolonged treatment [17, 20, 21].
68
Therefore, identification of new HDAC inhibitors with improved efficacy and safety is fully
69
essential.
RI PT
64
Chidamide is a novel benzamide chemical class of HDAC inhibitor that selectively inhibits
71
the histone deacetylation activities of HDAC1, 2, 3, as well as 10 [22]. It has been approved
72
by the Chinese Food and Drug Administration for the treatment of patients with recurrent or
73
refractory peripheral T-cell lymphoma (PTCL) [23]. Recently, a study screening active LRAs
74
in a primary cell model indicated that the HDAC inhibitors containing a benzamide functional
75
group and a pyridyl cap are the most effective, as exemplified by chidamide [24]. However,
76
unlike vorinostat, which belongs to the class of hydroxamic acids [6], little is known about
77
chidamide’s potential use and mechanism to reactivate latent HIV-1 provirus in latently
78
infected cells. Therefore, in this study, we dissected the effect of chidamide on reactivation of
79
HIV-1 provirus in latently infected cell models, as well as its possible mechanism of action.
80
We found chidamide to be much safer with more long-lasting effect than vorinostat. Our
81
results suggest that chidamide has the potential to be further developed as an effective and
82
safe LRA for HIV-1 functional cure.
M AN U
TE D
EP
AC C
83
SC
70
84
2. Materials and Methods
85
2.1. Cell Culture and Chemical Treatment
86
J-Lat A2 cells [25] (obtained from NIH AIDS Reagent Program), J-Lat C11 cells [26],
87
PBMCs (Allcells), ACH-2 cells [27] and U1 cells [28] were cultured in RPMI 1640 medium
88
with 10% fetal bovine serum (FBS) and 1% Pen/strep in a 37 °C incubator containing 5%
5
ACCEPTED MANUSCRIPT 89
CO2. HEK 293 cells and TZM-Bl cells [29] were grown in Dulbecco’s modified Eagle’s
90
medium (DMEM) with 10% fetal bovine serum and 1% Pen/strep at 37 °C under 5% CO2.
91
Chidamide
92
Pyrrolidinedithiocarbamic acid (PDTC) (Sigma), vorinostat (Sigma) and chidamide were
93
dissolved in anhydrous dimethyl sulfoxide (DMSO) and were stored at -20 °C. Recombinant
94
Human TNF-alpha Protein (R&D Systems) was dissolved in sterile PBS.
95
2.2. Flow Cytometry
provided
by
Aris
Pharmaceuticals
Inc.
Aspirin
(Sigma),
RI PT
was
C11 and A2 cells were incubated with the indicated concentrations of chidamide or
97
vorinostat at different time points, or pretreated with various concentrations of Aspirin for 3 h
98
and subsequently treated with chidamide, TNF-α or control DMSO for indicated hours. Cells
99
were washed and resuspended in PBS 3 times. GFP expression was measured by a BD Accuri
100
C6 Flow Cytometer (BD Biosciences) as previously described [30, 31]. All experiments were
101
performed independently at least three times in triplicate per experimental point.
102
2.3. HIV-1 antigen p24 assay
TE D
M AN U
SC
96
ACH-2 and U1 cells (4×104 cells) were seeded into a 96-well plate and then incubated with
104
various concentrations of chidamide and vorinostat at different time points. Viral release in
105
the supernatant was quantified by p24 ELISA assay as previously described [32]. All
106
experiments were performed independently at least three times in triplicate per experimental
107
point.
108
2.4. Cytotoxicity Assay
AC C
EP
103
109
Cell viability was measured following the instructions in the protocol provided in the cell
110
counting kit-8 (CCK-8; Dojindo Molecular Technologies, Gaithersburg, MD, USA) as
111
described [33]. J-Lat cells, HEK 293 T cells and PBMCs were seeded into a 96-well plate,
112
approximately 4×104 cells per well; 20 µL of CCK-8 solution were added to each well of the
113
plate, and cells were treated with or without chidamide for 48 h. After 2 to 3 h of incubation
6
ACCEPTED MANUSCRIPT
at 37 °C, the absorbance at 450 nm was taken by using a microplate reader (Infinite F200
115
PRO, Tecan, Switzerland). In this assay, the 50% cytotoxic concentration (CC50) was
116
calculated by use of the CalcuSyn software program (Biosoft, Ferguson, MO) [34] as we
117
previously described [35].
118
2.5. Western Blot
RI PT
114
Western blot analysis was performed as described [36]. Cells were harvested, washed once
120
with ice-cold PBS, and lysed in ice-cold RIPA buffer [50 mM Tris (pH 8.0), 150 mM NaCl, 1%
121
NP-40, and 2 mM EDTA)] containing PMSF and phosphatase inhibitor cocktails
122
(Thermofisher Scientific, Omaha, NE) for 15 minutes. Protein samples were separated by
123
SDS-PAGE and transferred onto nitrocellulose membrane (Pall Corporation, Port Washington,
124
NY). After blocking with 5% non-fat dry milk in PBS containing 0.1% Tween 20, proteins of
125
interest were probed with the corresponding primary antibodies [pp65 (Ser536) and IκBα
126
antibodies obtained from Cell Signaling (Danvers, MA)], followed by appropriate
127
infrared-conjugated secondary antibodies, Alexa Fluor 800 or 680 (Carlsbad, CA). The
128
immunoblotting was carried out using the LiCOR (Lincoln, NE) Odyssey scanner.
129
2.6. Luciferase Assay
TE D
M AN U
SC
119
1×104 ACH-2 cells per well were treated with chidamide or vorinostat and incubated with
131
the anti-HIV-1 drugs for 4 days at 37 °C, followed by collection of 100 µl culture
132
supernatants from each well on the fourth day. Then all supernatants were added into the
133
96-well polystyrene plate coated with TZM-Bl cells. After 48 h, TZM-Bl cells were lysed in
134
200µl of luciferase reporter lysis buffer. Luciferase activity was measured using luciferase
135
assay regents (Promega, Madison, WI, USA) and a luminescence counter (Infinite M200 Pro)
136
according to the manufacturer’s instructions as previously described [37].
137
2.7. Statistical Analysis
138
AC C
EP
130
Means and standard errors (SE) were calculated for all data points from at least 3
7
ACCEPTED MANUSCRIPT 139
independent experiments in triplicate. Statistical significance was determined using the
140
Student’s unpaired two-tailed t-test. All statistical analyses were carried out using GraphPad
141
Prism 6.0 (GraphPad Software). P < 0.05 was considered statistically significant.
RI PT
142
3. Results
144
3.1. Activation of HIV-1 expression in latently infected cells by chidamide in a
145
dose-dependent manner
SC
143
To investigate if chidamide has the potential to induce latent HIV-1 expression, we
147
performed an experiment on the J-Lat C11 cell line, which is a latently infected Jurkat T cell
148
line with a single provirus integrated into intron of RNPS1 and an EGFP gene under control
149
of the HIV-1 LTR [26]. We treated C11 cells with increasing concentrations of chidamide,
150
while using vorinostat as a positive control, for 48 h and measured the percentage of
151
GFP-positive cells by flow cytometry. We found a 4- to 25-fold increase in the percentage of
152
GFP-positive cells from the C11 cells treated with chidamide subjected to background levels
153
(Fig. 1A).
154
GFP-expressing cells reached approximately 82.6%. Similarly, the percentage of
155
GFP-positive cells rose from 3.2% to 87.7% after treatment of 0 to 2 µM vorinostat on C11
156
cells, but these percentages soon decreased after higher concentration of vorinostat was
157
introduced (Fig. 1A). The A2 cell line is another well-established HIV-1 J-Lat clone [25].
158
Therefore, as a check, we detected the effect of chidamide on the J-Lat A2 cell line to test
159
whether the same result could be achieved. Consistently, as shown in Figure 1B, the
160
percentage of GFP-positive cells was positively associated with the 0 to 4 µM concentration
161
of chidamide after 2 days of incubation in the culture medium, and the maximal percentage
162
increased to about 80.6% at 4 µM of chidamide over mock treatment. In addition, we found
TE D
M AN U
146
AC C
EP
When the concentration of chidamide was increased to 4 µM, the percentage of
8
ACCEPTED MANUSCRIPT 163
that vorinostat was toxic to C11 cells and A2 cells when its concentration exceeded 3 µM
164
based on the obvious reduction of the percentage of GFP-positive cells. Activity of chidamide as an HIV-1 reactivation agent was further confirmed in latent
166
HIV-1 chronically infected ACH-2 cells [27] and U1 cells [28], according to the production
167
of HIV-1 antigen p24 in the supernatant of cells cultured in the presence of LRAs. As shown
168
in Figure 1C and D, when the concentration of chidamide increased from 0 µM to 4 µM, p24
169
antigen production gradually rose. In ACH-2 cells, HIV-1 reactivation induced by chidamide
170
was positively correlated with 0 to 4 µM concentration, and its effect was frequently higher
171
than that of vorinostat (Fig. 1C). We also observed that the decreased activation of HIV-1
172
with vorinostat at 4 µM, as measured by p24 antigen production, was likely caused by
173
excessive cytotoxicity of vorinostat during continual 48 hours incubation in U1 cells (Fig. 1D).
174
These results demonstrated that chidamide potently induced HIV-1 LTR reactivation,
175
indicating its effect on HIV-1 production in a dose-dependent manner.
176
3.2. Chidamide activates HIV-1 expression in latent HIV-1 infected cells in a time-dependent
177
manner
TE D
M AN U
SC
RI PT
165
To assess the kinetics of HIV-1 LTR expression induced by chidamide, we performed a
179
kinetics experiment in which J-Lat A2 cells and ACH-2 cells were grown for 1 to 4 days with
180
or without chidamide for different concentrations. As shown in Figure 2A, after A2 cells were
181
treated with 1 µM chidamide, the percentage of GFP-positive cells increased over time and
182
rose to maximum (approximately 55%) on the fourth day. The kinetics of HIV-1 LTR
183
expression induction by vorinostat showed an apparent rise for the first 2 days, but then
184
tended toward constancy, or even reduction, by day 4 (Fig. 2A). We then tested the effect on
185
ACH-2 cells (Fig. 2B). From the first 3 days, we found that the effect of activation, as
186
measured by p24 antigen production, was almost the same between the treatment of
187
chidamide and vorinostat (Fig. 2B). In particular, p24 antigen production of the chidamide
AC C
EP
178
9
ACCEPTED MANUSCRIPT
group continuously increased after 3 days, while treatment with vorinostat resulted in only
189
negligible difference in variation. On the fourth day, we noticed that the effect of chidamide
190
on activation was significantly much higher than that of vorinostat in both cell lines (Fig. 2C
191
and D). These results indicated a time-dependent effect of chidamide on HIV-1 expression,
192
and as an HDAC inhibitor, chidamide could sustain reactivation over a period of time longer
193
than that shown by vorinostat.
194
3.3. Chidamide has minimal cytotoxicity in vitro
RI PT
188
To test the cytotoxicity of chidamide on different cells, we measured cell viability in A2,
196
C11, HEK 293 and PBMC cells treated with different concentrations of chidamide. After
197
treatment for 48h, cells were analyzed by CCK-8 assay. In the PBMC cells derived from
198
healthy HIV-negative donors, we did not find a large reduction in cell viability when the
199
concentration increased from 1 µM to 100 µM, while cell viability was clearly reduced at
200
concentrations > 25 µM, following treatment with vorinostat (Fig. 3A). In HEK293 T cells,
201
the CC50 was 37.3 µM (chidamide) and 12.5 µM (vorinostat) (Fig. 3B), respectively. This
202
result indicated that chidamide has relatively low toxicity compared to vorinostat on normal
203
human cells. The lower toxicity of chidamide compared with vorinostat was also observed in
204
J-Lat cells, including A2 cells and C11 cells, following incubation with chidamide at the same
205
concentrations as vorinostat based on the curve shown in Figure 3C and 3D, respectively.
206
CC50 of chidamide in J-Lat A2 cells and C11 cells was 22.3 µM and 18.6 µM, respectively.
207
CC50 of vorinostat in J-Lat A2 and C11 cells was 14 µM and 7.38 µM, respectively. Because
208
chidamide is an anticancer drug, it could be somewhat toxic to A2 and C11 cells from acute T
209
cell leukemia Jurkat cells. However, considering the lower toxicity in normal cells, such as
210
PBMCs and 293 T cells, our results illustrated that chidamide was safe at its active
211
concentration.
212
3.4. Chidamide reactivates HIV-1 latent provirus through NF-κB signaling
AC C
EP
TE D
M AN U
SC
195
10
ACCEPTED MANUSCRIPT
Binding sites for several inducible transcription factors, such as NF-κB, AP-1, and Sp1, can
214
be found in the HIV-1 LTR region [5, 10, 38]. Among these, the host transcription factor
215
NF-κB is critical for HIV-1 replication and has been most studied by LRAs [20, 39-44]. To
216
determine whether chidamide-mediated activation of HIV-1 is related to the NF-κB pathway,
217
we first examined protein expression of two components of the NF-κB pathway in J-Lat A2
218
cells by western blot analysis using antibodies specific for two NF-κB families, including
219
phospho-p65 (pp65) and IκBα. Our data showed a significant increase in the expression of
220
pp65 following treatment with chidamide as early as 2.5h (Fig. 4A and B), indicating that
221
NF-κB p65 was induced to be phosphorylated at Ser536 and transformed to pp65. The
222
activation of NF-κB usually involves degradation of the IκBα subunit bound to the NF-κB
223
dimer. Therefore, we next examined IκBα protein levels in J-Lat A2 cells stimulated with
224
chidamide (Fig. 4A). We observed an increased rate of IκB degradation following 2 hours of
225
treatment with chidamide (Fig. 4C). To dissect the involvement of the NF-κB pathway in viral
226
reactivation mediated by chidamide, the effect of some agents inhibiting the NF-κB pathway,
227
such as Aspirin and PDTC, was determined when used prior to chidamide treatment. J-Lat A2
228
cells were pretreated with different concentrations of aspirin, which could inhibit
229
TNF-α-induced activation of NF-κB [44, 45], and subsequently treated with chidamide or
230
TNF-α. We observed that the pretreatment of aspirin not only significantly inhibited
231
TNF-α-induced GFP expression in a dose-dependent manner, but also inhibited GFP
232
expression induced by chidamide at the concentrations tested (Fig. 4D). PDTC, a NF-κB
233
selective inhibitor [46], was also evaluated for HIV-1 p24 production in ACH-2 cells. The
234
increasing addition of PDTC to ACH-2 cells contributed to the significant inhibition of
235
chidamide-mediated reactivation from latency (Fig. 4E). Our findings collectively indicated
236
that chidamide-induced reactivation of latent HIV-1 might be mediated through the NF-κB
237
pathway.
AC C
EP
TE D
M AN U
SC
RI PT
213
11
ACCEPTED MANUSCRIPT 238
3.5. Combination of chidamide and anti-HIV-1 drugs inhibits viral infection To investigate whether chidamide could be combined with several anti-HIV-1 drugs to
240
inhibit active HIV-1 infection, we used TZM-Bl cells that contain a stably-integrated HIV-1
241
LTR linked to the luciferase reporter gene in their genome to examine the effect. ACH-2 cells
242
were treated with chidamide or anti-HIV-1 drugs, including AZT (Zidovudine), NVP
243
(Nevirapine), and IDV (Indinavir), for 4 days. Then we collected the supernatant of ACH-2
244
cells and added it to TZM-Bl cells to detect HIV-1 infection through luciferase assay. As
245
shown in Figure 5, in the absence of stimulation by chidamide, TZM-Bl cells did not appear
246
to express HIV-1 LTR with the treatment of anti-HIV drugs. When we performed chidamide
247
treatment alone, a 10-fold increase of HIV-1 expression could be evaluated by detecting the
248
expression of luciferase reporter gene, which demonstrated that 1) the latently infected HIV-1
249
was activated by chidamide, and 2) the reactivated HIV-1 could effectively infect and
250
replicate in the TZM-Bl cells that express CD4, CCR5 and CXCR4. By combining chidamide
251
with anti-HIV-1 drugs, the expression of HIV-1 LTR showed a rapid decrease, illustrating
252
that replication of the chidamide-reactivated HIV-1 in TZM-Bl cells was significantly
253
suppressed by the antivirus drugs tested. These results suggest that chidamide has a potential
254
to be used for HIV-1 functional cure based on the “shock and kill” strategy.
EP
TE D
M AN U
SC
RI PT
239
256
AC C
255
4. Discussion
257
Since HIV-1 remains integrated in the DNA of memory CD4+ T cells, cART for HIV-1
258
infection limits HIV-1 replication, but it still does not eliminate the virus [3]. The emergence
259
of the ‘‘shock and kill’’ strategy seems to be a harbinger of some new phase of HIV-1
260
therapeutics. This promising strategy means that the latent virus can be forced out of its
261
sanctuary, resulting in the eradication of the HIV-1 reservoir with targeted immunotherapy or
262
cART [5, 14, 47]. Therefore, identification of a highly effective LRA is critical for
12
ACCEPTED MANUSCRIPT
reactivation of most, if not all, of the latently infected provirus, in order to eliminate the latent
264
HIV-1 reservoir [48]. It was reported that some reactivators have been applied in clinical trials,
265
such as the HDAC inhibitors romidepsin [19], panobinostat [49], and vorinostat [16]. It is
266
well known that histone deacetylases (HDACs) belong to a family of enzymes equipped to
267
remove the acetyl group from histone lysine residues, inducing transcriptional repression
268
through chromatin condensation [50]. Thus, it can be seen that HDAC blocking is an
269
attractive means of inducing broad reactivation of latent HIV-1.
RI PT
263
Chidamide was a newly identified orally active benzamide class of HDAC inhibitor. In
271
recent years, researchers have made great effort in studying the HIV latency reversing effect
272
of the HDAC inhibitors belonging to the class of hydroxamic acids (e.g., vorinostat,
273
panobinostat and belinostat) or the class of cyclic peptide (e.g., apicidin and romidepsin).
274
However, few of the benzamide class of HDAC inhibitors have ever been reported to
275
reactivate the latent HIV provirus. Therefore, it is necessary to determine the ability of
276
chidamide to reverse HIV-1 latency. In this study, we explored chidamide’s potency in
277
reactivating latent HIV-1 expression on different latently infected cell models, including J-Lat
278
A2 cells, C11 cells, ACH-2 cells and U1 cells [28, 51]. Among these latent models, both C11
279
and J-Lat clone A2 cells are latently infected Jurkat T cells encoding GFP gene under the
280
control of HIV-1 LTR. The expression of GFP is used as a marker of latent HIV-1 expression,
281
and it can be detected by fluorescence microscopy and flow cytometry.
282
Jurkat T cells infected with pNL4-3-EGFP, which was reconstructed from pNL4-3 through the
283
introduction of gene-encoded enhancement of green fluorescent protein and mutation in Vpr
284
and Env [26]. Consequently, the same stimulation by chidamide or vorinostat could cause a
285
slight difference between these two cell lines. Moreover, as a marker of reactivation efficacy,
286
HIV-1 p24 antigen production levels were measured with chidamide treatment in ACH-2 and
287
U1 cells. These cell lines exhibit low levels of basal transcription, despite mutations in Tat
AC C
EP
TE D
M AN U
SC
270
C11 cells were
13
ACCEPTED MANUSCRIPT
(U1 cell) and TAR (ACH-2 cell) [27, 51]. In spite of the different integration, or mutation, site
289
in the cell lines from various origins, our results powerfully demonstrated that chidamide
290
could broadly and effectively activate HIV-1 production in a manner that was both
291
concentration- and time-dependent and do so in vitro at micromolar levels in these cells. It
292
was worth noting that chidamide induces HIV-1 LTR with lower toxicity in various cell lines
293
than vorinostat. Consistently, the benzamide class of HDAC inhibitors showed the least
294
pronounced toxicity in primary resting CD4+ T cells among most of the HDAC inhibitors [24].
295
Furthermore, in contrast to vorinostat, chidamide could retain its ability to reactivate HIV-1
296
much longer. Therefore, when carrying out the “shock and kill” strategy in clinical trials, we
297
recommend reducing the frequency of chidamide dosing in order to lessen its toxicity.
M AN U
SC
RI PT
288
After integration of HIV-1 into the host chromatin, proviral expression is largely controlled
299
by host cellular factors. Host transcription factors, including nuclear factor-κB (NF-κB),
300
nuclear factor of activated T cells (NFAT), AP1 and SP1, are sequestered in the cytoplasm in
301
resting cells and thus do not promote HIV-1 transcription until an appropriate cellular
302
activation signal is transmitted [38]. Among these transcription factors, NF-κB plays a central
303
role in the activation pathway of the HIV-1 provirus [10]. Therefore, the identification of
304
NF-κB activities has evoked a new direction in strategies to combat the effects of HIV-1
305
latency [31]. Chidamide could induce increasing levels of phospho-p65 expression and lead to
306
the degradation of IκBα which is an inhibited subunit in latently infected cells. It has been
307
reported that Aspirin can inhibit NF-κB activation induced by TNF-α by preventing the
308
phosphorylation and degradation of IκBα and nuclear translocation of NF-κB [45]. Therefore,
309
we examined the effect through Aspirin-inhibition experiments, following chidamide
310
treatment with TNF-α as a positive control, in J-Lat A2 cells. PDTC has been studied as a
311
NF-κB inhibitor, as well as an antioxidant agent, and we also showed that pretreatment of
312
ACH-2 cells with PDTC could remarkably prevent chidamide from inducing HIV-1
AC C
EP
TE D
298
14
ACCEPTED MANUSCRIPT
reactivation through expression of p24. The main activity of NF-κB on chidamide effects was
314
confirmed by using both inhibitors in viral reactivation experiments, which showed that
315
chidamide-mediated HIV-1 reactivation was abrogated. An increasing number of LRAs, such
316
as Oxaliplatin [30], M344 [44], Bryostatin [52], and PEP005 [40], have been demonstrated to
317
be involved in the NF-κB signaling pathway. However, besides M344, it has not be
318
demonstrated that other HDAC inhibitor, which contains a benzamide functional group, such
319
as entinostat and pimelic diphenylamide 106 [24], is involved in this pathway. Therefore, we
320
wanted to know if chidamide and other HDACs containing a benzamide functional group use
321
the NF-κB signal pathway or other pathways to activate latent HIV-1.
SC
RI PT
313
To achieve the “shock and kill” strategy for an HIV-1 cure, the role of anti-HIV-1 drugs on
323
viral clearance cannot be ignored. Therefore, we combined chidamide with anti-HIV-1 drugs
324
in order to determine if the active virus could be inhibited by antivirus drugs after activation
325
of HIV-1 from latently infected cells by chidamide. Among a variety of anti-HIV drugs, we
326
chose some drugs appearing to have optimal effect on eliminating HIV-1, such as nucleoside
327
reverse transcriptase inhibitors (NRTI), AZT , nonnucleoside reverse transcriptase inhibitors
328
(NNRTI), NVP, or the protease inhibitor IDV [53]. Our results showed that infection of
329
TZM-Bl cells by HIV-1 was significantly lower after treatment of AZT, NVP or IDV
330
cotreatment with chidamide, suggesting that treatments of such highly active antiretroviral
331
therapy could protect uninfected cells from becoming reinfected when using a combination of
332
HIV activators. Moreover, when combined with chidamide, we found that such anti-HIV-1
333
drugs as AZT, NVP and IDV did not lose their anti-HIV-1 potency, suggesting that these
334
anti-HIV-1 drugs can be used in combination therapy aimed at eliminating latent HIV-1
335
reservoirs.
AC C
EP
TE D
M AN U
322
336
To sum up, our results have demonstrated that chidamide plays an important role in histone
337
modification via the NF-κB pathway to regulate HIV-1 LTR gene expression and to reactivate
15
ACCEPTED MANUSCRIPT 338
latent HIV-1, thus having good potential to be used in combination with anti-HIV drugs for
339
HIV-1 functional cure.
340
Conflict of Interests
342
The authors declare no conflict of interests regarding the publication of this paper.
RI PT
341
343
Acknowledgments
SC
344
This work was supported by grants from the National Natural Science Foundation of China
346
(81630090 to S.J.; 81373456, 81672019 and 81661128041 to L.L.), the Sanming Project of
347
Medicine in Shenzhen, the National 863 Program of China (2015AA020930 to LL) and the
348
Shanghai Rising-Star Program (16QA1400300).
M AN U
345
EP AC C
350
TE D
349
16
ACCEPTED MANUSCRIPT
References
352
[1] Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al.
353
Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy.
354
Science 1997;278:1295-300.
355 356
RI PT
351
[2] Siliciano RF, Greene WC. HIV latency. Cold Spring Harb. Perspect. Med. 2011;1:a007096.
[3] Ruelas DS, Greene WC. An integrated overview of HIV-1 latency. Cell 2013;155:519-29.
358
[4] Siliciano JM, Siliciano RF. The remarkable stability of the latent reservoir for HIV-1 in
363 364 365 366 367
M AN U
362
the development of new therapeutic strategies. Retrovirology 2009;6:111. [6] Xing S, Siliciano RF. Targeting HIV latency: pharmacologic strategies toward eradication.
TE D
361
[5] Colin L, Van Lint C. Molecular control of HIV-1 postintegration latency: implications for
Drug Discov Today 2013;18:541-51.
[7] Margolis DM, Garcia JV, Hazuda DJ, Haynes BF. Latency reversal and viral clearance to
EP
360
resting memory CD4+ T cells. J. Infect. Dis. 2015;212:1345-7.
cure HIV-1. Science 2016;353:aaf6517. [8] Martin AR, Siliciano RF. Progress toward HIV eradication: case reports, current efforts,
AC C
359
SC
357
and the challenges associated with cure. Annu. Rev. Med. 2016;67:215-28.
368
[9] Coiras M, Lopez-Huertas MR, Perez-Olmeda M, Alcami J. Understanding HIV-1 latency
369
provides clues for the eradication of long-term reservoirs. Nat Rev Microbiol
370
2009;7:798-812.
371
[10] Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, Greene WC. NF-kappaB p50
372
promotes HIV latency through HDAC recruitment and repression of transcriptional
17
ACCEPTED MANUSCRIPT 373
initiation. EMBO J. 2006;25:139-49. [11] Lucera MB, Tilton CA, Mao H, Dobrowolski C, Tabler CO, Haqqani AA, et al. The
375
histone deacetylase inhibitor vorinostat (SAHA) increases the susceptibility of uninfected
376
CD4+ T cells to HIV by increasing the kinetics and efficiency of postentry viral events. J.
377
Virol. 2014;88:10803-12.
RI PT
374
[12] Banga R, Procopio FA, Cavassini M, Perreau M. In vitro reactivation of
379
replication-competent and infectious HIV-1 by histone deacetylase inhibitors. J. Virol.
380
2015;90:1858-71.
M AN U
SC
378
381
[13] Deeks SG. HIV: Shock and kill. Nature 2012;487:439-40.
382
[14] Xu W, Li H, Wang Q, Hua C, Zhang H, Li W, et al. Advancements in developing strategies
383
for sterilizing and functional HIV cures. Biomed Res Int 2017;2017:6096134. [15] Laird GM, Bullen CK, Rosenbloom DI, Martin AR, Hill AL, Durand CM, et al. Ex vivo
385
analysis identifies effective HIV-1 latency-reversing drug combinations. J. Clin. Invest.
386
2015;125:1901-12.
TE D
384
[16] Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, et al.
388
Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy.
389
Nature 2012;487:482-5.
AC C
EP
387
390
[17] Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, et al. Activation
391
of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive
392
antiretroviral therapy. PLoS Pathog. 2014;10:e1004473.
393
[18] Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A, et al.
394
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected
18
ACCEPTED MANUSCRIPT 395
patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. The
396
lancet. HIV 2014;1:e13-21. [19] Sogaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, Nissen SK, et al. The
398
depsipeptide
romidepsin
399
2015;11:e1005142.
reverses
HIV-1
latency
in
vivo.
PLoS
Pathog.
RI PT
397
[20] Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1
401
infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 2014;12:750-64.
402
[21] Gama L, Abreu CM, Shirk EN, Price SL, Li M, Laird GM, et al. Reactivation of simian
403
immunodeficiency virus reservoirs in the brain of virally suppressed macaques. AIDS
404
2017;31:5-14.
M AN U
SC
400
[22] Gu R, Liu T, Zhu X, Gan H, Wu Z, Li J, et al. Development and validation of a sensitive
406
HPLC-MS/MS method for determination of chidamide (epidaza), a new benzamide class
407
of selective histone deacetylase inhibitor, in human plasma and its clinical application. J.
408
Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2015;1000:181-6.
TE D
405
[23] Lu X, Ning Z, Li Z, Cao H, Wang X. Development of chidamide for peripheral T-cell
410
lymphoma, the first orphan drug approved in China. Intractable Rare Dis Res
411
2016;5:185-91.
AC C
EP
409
412
[24] Kobayashi Y, Gelinas C, Dougherty JP. Histone deacetylase inhibitors containing a
413
benzamide functional group and a pyridyl cap are preferentially effective human
414
immunodeficiency virus-1 latency-reversing agents in primary resting CD4+ T cells. J.
415
Gen. Virol. 2017;98:799-809.
416
[25] Jordan A, Bisgrove D, Verdin E. HIV reproducibly establishes a latent infection after
19
ACCEPTED MANUSCRIPT 417
acute infection of T cells in vitro. EMBO J. 2003;22:1868-77. [26] Ding D, Qu X, Li L, Zhou X, Liu S, Lin S, et al. Involvement of histone methyltransferase
419
GLP in HIV-1 latency through catalysis of H3K9 dimethylation. Virology
420
2013;440:182-9.
RI PT
418
[27] Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, et al. Tumor necrosis
422
factor alpha induces expression of human immunodeficiency virus in a chronically
423
infected T-cell clone. Proc. Natl. Acad. Sci. U. S. A. 1989;86:2365-8.
SC
421
[28] Clouse KA, Powell D, Washington I, Poli G, Strebel K, Farrar W, et al. Monokine
425
regulation of human immunodeficiency virus-1 expression in a chronically infected
426
human T cell clone. J. Immunol. 1989;142:431-8.
M AN U
424
[29] Contreras X, Schweneker M, Chen CS, McCune JM, Deeks SG, Martin J, et al.
428
Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J. Biol.
429
Chem. 2009;284:6782-9.
TE D
427
[30] Zhu X, Liu S, Wang P, Qu X, Wang X, Zeng H, et al. Oxaliplatin antagonizes HIV-1
431
latency by activating NF-kappaB without causing global T cell activation. Biochem.
432
Biophys. Res. Commun. 2014;450:202-7.
434
AC C
433
EP
430
[31] Lu P, Qu X, Shen Y, Jiang Z, Wang P, Zeng H, et al. The BET inhibitor OTX015 reactivates latent HIV-1 through P-TEFb. Sci. Rep. 2016;6:24100.
435
[32] Li W, Yu F, Wang Q, Qi Q, Su S, Xie L, et al. Co-delivery of HIV-1 entry inhibitor and
436
nonnucleoside reverse transcriptase inhibitor shuttled by nanoparticles: cocktail
437
therapeutic strategy for antiviral therapy. AIDS 2016;30:827-38.
438
[33] Yu Y, Deng YQ, Zou P, Wang Q, Dai Y, Yu F, et al. A peptide-based viral inactivator
20
ACCEPTED MANUSCRIPT 439
inhibits Zika virus infection in pregnant mice and fetuses. Nat Commun 2017;8:15672. [34] Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined
441
effects of multiple drugs or enzyme inhibitors. Adv. Enzyme Regul. 1984;22:27-55.
442
[35] Jiang S, Lu H, Liu S, Zhao Q, He Y, Debnath AK. N-substituted pyrrole derivatives as
443
novel human immunodeficiency virus type 1 entry inhibitors that interfere with the gp41
444
six-helix bundle formation and block virus fusion. Antimicrob. Agents Chemother.
445
2004;48:4349-59.
447
SC
[36] Gan J, Wang C, Jin Y, Guo Y, Xu F, Zhu Q, et al. Proteomic profiling identifies the
M AN U
446
RI PT
440
SIM-associated complex of KSHV-encoded LANA. Proteomics 2015;15:2023-37. [37] Sun Z, Zhu Y, Wang Q, Ye L, Dai Y, Su S, et al. An immunogen containing four tandem
449
10E8 epitope repeats with exposed key residues induces antibodies that neutralize HIV-1
450
and activates an ADCC reporter gene. Emerg Microbes Infect 2016;5:e65.
TE D
448
[38] Victoriano AF, Imai K, Togami H, Ueno T, Asamitsu K, Suzuki T, et al. Novel histone
452
deacetylase inhibitor NCH-51 activates latent HIV-1 gene expression. FEBS Lett.
453
2011;585:1103-11.
EP
451
[39] Jiang G, Mendes EA, Kaiser P, Sankaran-Walters S, Tang Y, Weber MG, et al.
455
Reactivation of HIV latency by a newly modified Ingenol derivative via protein kinase
456
Cdelta-NF-kappaB signaling. AIDS 2014;28:1555-66.
AC C
454
457
[40] Jiang G, Mendes EA, Kaiser P, Wong DP, Tang Y, Cai I, et al. Synergistic reactivation of
458
latent HIV expression by ingenol-3-angelate, PEP005, targeted NF-κB signaling in
459
combination with JQ1 induced p-TEFb activation. PLoS Pathog. 2015;11:e1005066.
460
[41] Stroud JC, Oltman A, Han A, Bates DL, Chen L. Structural basis of HIV-1 activation by
21
ACCEPTED MANUSCRIPT 461
NF-kappaB--a higher-order complex of p50:RelA bound to the HIV-1 LTR. J. Mol. Biol.
462
2009;393:98-112.
464 465 466
[42] Wang P, Qu X, Wang X, Liu L, Zhu X, Zeng H, et al. As2O3 synergistically reactivate latent HIV-1 by induction of NF-kappaB. Antiviral Res. 2013.
RI PT
463
[43] Williams SA, Chen LF, Kwon H, Fenard D, Bisgrove D, Verdin E, et al. Prostratin antagonizes HIV latency by activating NF-kappaB. J. Biol. Chem. 2004;279:42008-17. [44] Ying H, Zhang Y, Zhou X, Qu X, Wang P, Liu S, et al. Selective histonedeacetylase
468
inhibitor M344 intervenes in HIV-1 latency through increasing histone acetylation and
469
activation of NF-kappaB. PLoS One 2012;7:e48832.
471
M AN U
470
SC
467
[45] Kutuk O, Basaga H. Aspirin inhibits TNFalpha- and IL-1-induced NF-kappaB activation and sensitizes HeLa cells to apoptosis. Cytokine 2004;25:229-37. [46] Alvarez S, Blanco A, Kern F, Fresno M, Munoz-Fernandez MA. HIV-2 induces
473
NF-kappaB activation and cyclooxygenase-2 expression in human astroglial cells.
474
Virology 2008;380:144-51.
EP
476
[47] Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science 2009;323:1304-7.
AC C
475
TE D
472
477
[48] Gallo RC. Shock and kill with caution. Science 2016;354:177-8.
478
[49] Clutton G, Xu Y, Baldoni PL, Mollan KR, Kirchherr J, Newhard W, et al. The differential
479
short- and long-term effects of HIV-1 latency-reversing agents on T cell function. Sci. Rep.
480
2016;6:30749.
481 482
[50] Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015;20:3898-941.
22
ACCEPTED MANUSCRIPT 483 484
[51] Folks TM, Justement J, Kinter A, Dinarello CA, Fauci AS. Cytokine-induced expression of HIV-1 in a chronically infected promonocyte cell line. Science 1987;238:800-2. [52] Mehla R, Bivalkar-Mehla S, Zhang R, Handy I, Albrecht H, Giri S, et al. Bryostatin
486
modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute
487
infection in a receptor independent manner. PLoS Onke 2010;5:e11160.
489
[53] Staszewski S. Update on study 006--EFV + AZT + 3TC versus the current 'standard of care' IDV + AZT + 3TC. Int. J. Clin. Pract. Suppl. 1999;103:10-5.
SC
488
RI PT
485
M AN U
490
AC C
EP
TE D
491
23
ACCEPTED MANUSCRIPT
Figure legends
493
Fig. 1. Reactivation of latent HIV-1 in different latently infected cells by chidamide. (A) J-Lat
494
clone C11 cells were treated with chidamide and vorinostat for 48 h at the indicated
495
concentrations. Results are expressed as a percentage of GFP-positive cells within the entire
496
population. (B) J-Lat clone A2 cells treated with chidamide and vorinostat for 48 h at the
497
indicated concentrations and analyzed as in (A). ACH-2 cells (C) or U1 (D) cells were
498
stimulated with increasing concentrations of chidamide and vorinostat for 48 h. Viral
499
replication was measured in the supernatant of the cells using p24 ELISA. Data show
500
dose-dependent effects of chidamide on HIV-1 production in different latently infected cells
501
and represent the means ± standard deviations of three independent experiments.
M AN U
SC
RI PT
492
502
Fig. 2. Time-dependent effects of chidamide on HIV-1 production. J-Lat A2 cells (A) or
504
ACH-2 cells (B) were treated with DMSO or with 1µM chidamide or vorinostat for 0, 24, 48,
505
72, 96 h. A time-dependent curve on HIV-1 transcription is represented by two latent cell
506
models. (C) and (D) represented the comparison of the effect on activating latently infected
507
cells between chidamide and vorinostat at the third day and the fourth day. Data show
508
time-dependent effects of chidamide on activating latently infected cell lines and represent the
509
means ± standard deviations of three independent experiments. (**P < 0.01 and***P < 0.001).
AC C
EP
TE D
503
510
511
Fig. 3. Viability of cells treated with chidamide or vorinostat. PBMCs from HIV-negative
512
donors (A), HEK293 T cells (B), J-Lat A2 cells (C) and C11 cells (D) were treated with
24
ACCEPTED MANUSCRIPT
chidamide and vorinostat from 0 to 100 µM for 48 h, and cytotoxicity was measured by
514
CCK-8 kit. The absorbance at 450 nm (OD450) of each well was determined as the readout of
515
cell viability. Data represent the means ± standard deviations of three independent
516
experiments.
RI PT
513
517
Fig. 4. Reactivation of HIV-1 latent provirus by chidamide through NF-κB pathway. (A) J-Lat
519
A2 cells were treated with 2 µM of chidamide for up to 3.5 h hours. Western blot analysis was
520
performed to detect the expression of p-p65 and IκBα. (B) Quantitation of phosphorylation of
521
p65 in J-Lat A2 cells after 3.5 hours treatment with chidamide in panel (A). Relative band
522
intensities from three independent experiments in J-Lat A2 cells, as determined using Image J
523
(NIH), are shown in the bar graph. (C) Quantitation of IκBα in J-Lat A2 cells after 3.5 hours
524
treatment with chidamide in panel (A). Relative band intensities from three independent
525
experiments in J-Lat A2 cells, as determined using ImageJ (NIH), are shown in the bar graph.
526
(D) J-Lat A2 cells were pretreated with various concentrations of Aspirin for 3 h and
527
subsequently treated with chidamide (1, 2 and 5 µM), TNF-a (10 ng/mL) or DMSO as control
528
for 48 h. The percentage of GFP-positive cells in drug-treated cells in the absence or presence
529
of the inhibitor Aspirin was measured by flow cytometry. (E) ACH-2 cells were pretreated
530
with increasing concentrations of PDTC for 2 h and subsequently treated with chidamide (2
531
µM); p24 production from latently infected cells was measured by p24 ELISA. Data represent
532
the means ± standard deviations of three independent experiments. (*P < 0.05, P < **0.01 and
533
***P < 0.001).
534
AC C
EP
TE D
M AN U
SC
518
25
ACCEPTED MANUSCRIPT
Fig. 5. Combination of chidamide with anti-HIV-1 drugs to suppress infection of reactivated
536
HIV-1. (a) 1×104 ACH-2 cells were treated with chidamide, vorinostat or PBS in the presence
537
or absence of the anti-HIV drugs AZT, NVP, or IDV for 4 days. The supernatants of ACH-2
538
cells containing the residual reactivated HIV-1 were cultured with TZM-Bl cells for 48 hours
539
before testing residual HIV-1 infectivity using the luciferase assay. Data represent the means ±
540
standard deviations of three independent experiments (***P < 0.001).
AC C
EP
TE D
M AN U
SC
RI PT
535
ACCEPTED MANUSCRIPT
Fig. 1. C11 cells
A2 cells
A
AC C
C
EP
ACH-2 cells
TE D
M AN U
SC
RI PT
B
U1 cells D
Fig. 2.
ACCEPTED MANUSCRIPT
A2 cells A A
ACH-2 cells
EP AC C
C C
TE D
M AN U
SC
RI PT
B
D
ACCEPTED MANUSCRIPT
Fig. 3. PBMCs
HEK 293 T cells
A
AC C
C
EP
A2 cells
TE D
M AN U
SC
RI PT
B
C11 cells
D
Fig. 4.
ACCEPTED MANUSCRIPT
B
Chidamide 0h
1h
2h
2.5 h 3 h
3.5 h
p-p65 (S536) β-actin
SC
C
M AN U
IκBα
AC C
EP
TE D
β-actin
D
RI PT
A
E
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
Fig. 5.
ACCEPTED MANUSCRIPT Elsevier Editorial System(tm) for Microbes and Infection Manuscript Draft Manuscript Number: MICINF-D-17-00218R1
Article Type: Special Issue
RI PT
Title: Chidamide, a histone deacetylase inhibitor-based anticancer drug, effectively reactivates latent HIV-1 provirus
Keywords: Key Words: Histone deacetylase inhibitor; Chidamide; HIV-1 latent infection Corresponding Author: Professor Shibo Jiang,
SC
Corresponding Author's Institution: Fudan University First Author: Wenqian Yang
M AN U
Order of Authors: Wenqian Yang; Zhiwu Sun; Chen Hua; Qian Wang; Wei Xu; Qiwen Deng; Yanbin Pan; Lu Lu; Shibo Jiang
AC C
EP
TE D
Abstract: Although combination antiretroviral therapy (cART) is highly effective in suppressing human immunodeficiency virus type 1 (HIV-1) replication, it fails to eradicate the virus from HIV-1-infected individuals because HIV-1 integrates into the resting CD4+ T cells, establishing latently infected reservoirs. Histone deacetylation is a key element in regulating HIV-1 latent infection. Chidamide, a new anticancer drug, is a novel type of selective histone deacetylase inhibitor. Here we showed that chidamide effectively reactivated HIV-1 latent provirus in different latently infected cell lines in a dose- and time-dependent manner. Chidamide had relatively low cytotoxicity to peripheral blood mononuclear cells (PBMCs) and other latent cell lines. We have demonstrated that chidamide reactivates HIV-1 latent provirus through the NF-κB signaling pathway. The replication of the newly reactivated HIV-1 could then be effectively inhibited by the anti-HIV drugs Zidovudine, Nevirapine, and Indinavir. Therefore, chidamide might be used in combination with cART for functional HIV-1 cure.
Cover Letter
ACCEPTED MANUSCRIPT Dear Dr. Ojcius: Thank you very much for your further consideration of our manuscript (MICINF-D-17-00218) entitled “Chidamide, a histone deacetylase inhibitor-based anticancer drug, effectively reactivates latent HIV-1 provirus” for publication in the special issue of Microbes and Infection.
SC
RI PT
We would like to express our sincere appreciation for your time and efforts in handling the above referenced manuscript and to the reviewers for their constructive and thoughtful comments. We have now revised our manuscript according to these suggestions and tried our best to address all the concerns. The added or modified sentences were highlighted in red for your convenience to review. The point-by-point responses to reviewers’ comments were submitted separately.
M AN U
We hope that we have satisfactorily addressed the reviewers’ concerns and that the revised manuscript will now be found acceptable for publication in Microbes and Infection. Sincerely yours,
AC C
EP
TE D
Dr. Shibo Jiang Professor and Director, Institute of Medical Microbiology Key Laboratory of Medical Molecular Virology of Ministries of Education and Health School of Basic Medical Sciences, Fudan University 130 Dong An Road, Building #13, Rm 611 Xuhui District, Shanghai 200032, China Phone: +86 21-54237673 Fax: +86 21-54237465 E-mail:
[email protected]
*Manuscript Click here to view linked References
1
ACCEPTED MANUSCRIPT
Chidamide, a histone deacetylase inhibitor-based anticancer drug,
1
effectively reactivates latent HIV-1 provirus
2 3
Wenqian Yang a,#, Zhiwu Sun a,#, Chen Hua a, Qian Wang a, Wei Xu a, Qiwen Deng b,
5
Yanbin Pan c, Lu Lu a,*, Shibo Jiang a,b,d,*
RI PT
4
6 a
Key Lab of Medical Molecular Virology of Ministries of Education and Health, School of
SC
7
Basic Medical Sciences & Shanghai Public Health Clinical Center, Fudan University,
9
Shanghai, China b
10
Shenzhen Nanshan People's Hospital of Shenzhen University, Shenzhen 518052, China c
11 d
Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA
#
These authors have equal contribution.
13
EP
14 15
Aris Pharmaceuticals Inc., Bristol, PA19007, USA
TE D
12
M AN U
8
AC C
16 17
*Address for correspondence: Shibo Jiang:
[email protected]; Lu Lu:
18
[email protected].
19 20 21 22
2
ACCEPTED MANUSCRIPT 23
Abstract Although combination antiretroviral therapy (cART) is highly effective in suppressing
25
human immunodeficiency virus type 1 (HIV-1) replication, it fails to eradicate the virus from
26
HIV-1-infected individuals because HIV-1 integrates into the resting CD4+ T cells,
27
establishing latently infected reservoirs. Histone deacetylation is a key element in regulating
28
HIV-1 latent infection. Chidamide, a new anticancer drug, is a novel type of selective histone
29
deacetylase inhibitor. Here we showed that chidamide effectively reactivated HIV-1 latent
30
provirus in different latently infected cell lines in a dose- and time-dependent manner.
31
Chidamide had relatively low cytotoxicity to peripheral blood mononuclear cells (PBMCs)
32
and other latent cell lines. We have demonstrated that chidamide reactivates HIV-1 latent
33
provirus through the NF-κB signaling pathway. The replication of the newly reactivated
34
HIV-1 could then be effectively inhibited by the anti-HIV drugs Zidovudine, Nevirapine, and
35
Indinavir. Therefore, chidamide might be used in combination with cART for functional
36
HIV-1 cure.
37
Key Words: Histone deacetylase inhibitor; Chidamide; HIV-1 latent infection
SC
M AN U
TE D
EP AC C
38
RI PT
24
3
ACCEPTED MANUSCRIPT 39
1. Introduction Combination antiretroviral therapy (cART) has succeeded in reducing human
41
immunodeficiency virus type 1 (HIV-1) load to undetectable levels in most patients [1], but
42
the therapy must be maintained throughout life. Once cART is withdrawn, viral loads could
43
return to even pretreatment levels within weeks. One of the principal obstacles to a thorough
44
cleaning of the virus is the integration of HIV-1 genome into that of the resting CD4+ T cells
45
during acute infection, thereby establishing the latently infected reservoir that harbors
46
transcriptionally silent proviruses refractory to current cART [2-4]. Therefore, the
47
development of HIV-1 therapeutic strategies to eliminate this latent reservoir is urgently
48
needed.
M AN U
SC
RI PT
40
Some host cell-mediated molecular mechanisms maintain the quiescence of HIV-1 gene
50
expression, including epigenetic silencing of the proviral promoter, transcription complex
51
formation, transcript initiation, and transcription complex processivity, all of which might act
52
as therapeutic targets to disrupt HIV-1 latency [5-8]. Among these mechanisms, histone
53
deacetylase (HDAC)-mediated chromosomal suppression of HIV-1 long terminal repeat (LTR)
54
is a key regulator leading to the establishment of HIV-1 latency [7, 9]. It was reported the host
55
transcription factors comprised of NF-κB family members are involved in HDAC1
56
recruitment [10]. HDAC inhibitors have been shown to increase cell-associated HIV-1 RNA
57
production and/or plasma viremia in vivo [6, 11, 12], suggesting that they could play a role in
58
reactivating latent HIV-1 provirus.
AC C
EP
TE D
49
59
A strategy known as ‘shock and kill’, or ‘kick and kill’, has been developed for HIV-1
60
functional cure [13, 14]. To explain, the latent HIV-1 provirus was first reactivated with some
61
small-molecule HIV-1 latency-reversing agents (LRAs) [15] and then killed by the patient’s
62
immune system or suppressed by cART. Clinical trials have demonstrated that administration
63
of HDAC inhibitors, such as vorinostat [16, 17] or panobinostat [18], to the patients in whom
4
ACCEPTED MANUSCRIPT
viremia was fully suppressed by cART, resulted in a significant increase in HIV-1 RNA
65
production in resting CD4+ cells. Another clinical trial had shown that the HDAC inhibitor
66
romidepsin was effective in disrupting HIV-1 latency in vivo [19]. However, some LRAs may
67
be toxic and mutagenic or harmful to host brain during prolonged treatment [17, 20, 21].
68
Therefore, identification of new HDAC inhibitors with improved efficacy and safety is fully
69
essential.
RI PT
64
Chidamide is a novel benzamide chemical class of HDAC inhibitor that selectively inhibits
71
the histone deacetylation activities of HDAC1, 2, 3, as well as 10 [22]. It has been approved
72
by the Chinese Food and Drug Administration for the treatment of patients with recurrent or
73
refractory peripheral T-cell lymphoma (PTCL) [23]. Recently, a study screening active LRAs
74
in a primary cell model indicated that the HDAC inhibitors containing a benzamide functional
75
group and a pyridyl cap are the most effective, as exemplified by chidamide [24]. However,
76
unlike vorinostat, which belongs to the class of hydroxamic acids [6], little is known about
77
chidamide’s potential use and mechanism to reactivate latent HIV-1 provirus in latently
78
infected cells. Therefore, in this study, we dissected the effect of chidamide on reactivation of
79
HIV-1 provirus in latently infected cell models, as well as its possible mechanism of action.
80
We found chidamide to be much safer with more long-lasting effect than vorinostat. Our
81
results suggest that chidamide has the potential to be further developed as an effective and
82
safe LRA for HIV-1 functional cure.
M AN U
TE D
EP
AC C
83
SC
70
84
2. Materials and Methods
85
2.1. Cell Culture and Chemical Treatment
86
J-Lat A2 cells [25] (obtained from NIH AIDS Reagent Program), J-Lat C11 cells [26],
87
PBMCs (Allcells), ACH-2 cells [27] and U1 cells [28] were cultured in RPMI 1640 medium
88
with 10% fetal bovine serum (FBS) and 1% Pen/strep in a 37 °C incubator containing 5%
5
ACCEPTED MANUSCRIPT 89
CO2. HEK 293 cells and TZM-Bl cells [29] were grown in Dulbecco’s modified Eagle’s
90
medium (DMEM) with 10% fetal bovine serum and 1% Pen/strep at 37 °C under 5% CO2.
91
Chidamide
92
Pyrrolidinedithiocarbamic acid (PDTC) (Sigma), vorinostat (Sigma) and chidamide were
93
dissolved in anhydrous dimethyl sulfoxide (DMSO) and were stored at -20 °C. Recombinant
94
Human TNF-alpha Protein (R&D Systems) was dissolved in sterile PBS.
95
2.2. Flow Cytometry
provided
by
Aris
Pharmaceuticals
Inc.
Aspirin
(Sigma),
RI PT
was
C11 and A2 cells were incubated with the indicated concentrations of chidamide or
97
vorinostat at different time points, or pretreated with various concentrations of Aspirin for 3 h
98
and subsequently treated with chidamide, TNF-α or control DMSO for indicated hours. Cells
99
were washed and resuspended in PBS 3 times. GFP expression was measured by a BD Accuri
100
C6 Flow Cytometer (BD Biosciences) as previously described [30, 31]. All experiments were
101
performed independently at least three times in triplicate per experimental point.
102
2.3. HIV-1 antigen p24 assay
TE D
M AN U
SC
96
ACH-2 and U1 cells (4×104 cells) were seeded into a 96-well plate and then incubated with
104
various concentrations of chidamide and vorinostat at different time points. Viral release in
105
the supernatant was quantified by p24 ELISA assay as previously described [32]. All
106
experiments were performed independently at least three times in triplicate per experimental
107
point.
108
2.4. Cytotoxicity Assay
AC C
EP
103
109
Cell viability was measured following the instructions in the protocol provided in the cell
110
counting kit-8 (CCK-8; Dojindo Molecular Technologies, Gaithersburg, MD, USA) as
111
described [33]. J-Lat cells, HEK 293 T cells and PBMCs were seeded into a 96-well plate,
112
approximately 4×104 cells per well; 20 μL of CCK-8 solution were added to each well of the
113
plate, and cells were treated with or without chidamide for 48 h. After 2 to 3 h of incubation
6
ACCEPTED MANUSCRIPT
at 37 °C, the absorbance at 450 nm was taken by using a microplate reader (Infinite F200
115
PRO, Tecan, Switzerland). In this assay, the 50% cytotoxic concentration (CC50) was
116
calculated by use of the CalcuSyn software program (Biosoft, Ferguson, MO) [34] as we
117
previously described [35].
118
2.5. Western Blot
RI PT
114
Western blot analysis was performed as described [36]. Cells were harvested, washed once
120
with ice-cold PBS, and lysed in ice-cold RIPA buffer [50 mM Tris (pH 8.0), 150 mM NaCl, 1%
121
NP-40, and 2 mM EDTA)] containing PMSF and phosphatase inhibitor cocktails
122
(Thermofisher Scientific, Omaha, NE) for 15 minutes. Protein samples were separated by
123
SDS-PAGE and transferred onto nitrocellulose membrane (Pall Corporation, Port Washington,
124
NY). After blocking with 5% non-fat dry milk in PBS containing 0.1% Tween 20, proteins of
125
interest were probed with the corresponding primary antibodies [pp65 (Ser536) and IκBα
126
antibodies obtained from Cell Signaling (Danvers, MA)], followed by appropriate
127
infrared-conjugated secondary antibodies, Alexa Fluor 800 or 680 (Carlsbad, CA). The
128
immunoblotting was carried out using the LiCOR (Lincoln, NE) Odyssey scanner.
129
2.6. Luciferase Assay
TE D
M AN U
SC
119
1×104 ACH-2 cells per well were treated with chidamide or vorinostat and incubated with
131
the anti-HIV-1 drugs for 4 days at 37 °C, followed by collection of 100 μl culture
132
supernatants from each well on the fourth day. Then all supernatants were added into the
133
96-well polystyrene plate coated with TZM-Bl cells. After 48 h, TZM-Bl cells were lysed in
134
200μl of luciferase reporter lysis buffer. Luciferase activity was measured using luciferase
135
assay regents (Promega, Madison, WI, USA) and a luminescence counter (Infinite M200 Pro)
136
according to the manufacturer’s instructions as previously described [37].
137
2.7. Statistical Analysis
138
AC C
EP
130
Means and standard errors (SE) were calculated for all data points from at least 3
7
ACCEPTED MANUSCRIPT 139
independent experiments in triplicate. Statistical significance was determined using the
140
Student’s unpaired two-tailed t-test. All statistical analyses were carried out using GraphPad
141
Prism 6.0 (GraphPad Software). P < 0.05 was considered statistically significant.
RI PT
142
3. Results
144
3.1. Activation of HIV-1 expression in latently infected cells by chidamide in a
145
dose-dependent manner
SC
143
To investigate if chidamide has the potential to induce latent HIV-1 expression, we
147
performed an experiment on the J-Lat C11 cell line, which is a latently infected Jurkat T cell
148
line with a single provirus integrated into intron of RNPS1 and an EGFP gene under control
149
of the HIV-1 LTR [26]. We treated C11 cells with increasing concentrations of chidamide,
150
while using vorinostat as a positive control, for 48 h and measured the percentage of
151
GFP-positive cells by flow cytometry. We found a 4- to 25-fold increase in the percentage of
152
GFP-positive cells from the C11 cells treated with chidamide subjected to background levels
153
(Fig. 1A). When the concentration of chidamide was increased to 4 μM, the percentage of
154
GFP-expressing cells reached approximately 82.6%. Similarly, the percentage of
155
GFP-positive cells rose from 3.2% to 87.7% after treatment of 0 to 2 μM vorinostat on C11
156
cells, but these percentages soon decreased after higher concentration of vorinostat was
157
introduced (Fig. 1A). The A2 cell line is another well-established HIV-1 J-Lat clone [25].
158
Therefore, as a check, we detected the effect of chidamide on the J-Lat A2 cell line to test
159
whether the same result could be achieved. Consistently, as shown in Figure 1B, the
160
percentage of GFP-positive cells was positively associated with the 0 to 4 μM concentration
161
of chidamide after 2 days of incubation in the culture medium, and the maximal percentage
162
increased to about 80.6% at 4 μM of chidamide over mock treatment. In addition, we found
AC C
EP
TE D
M AN U
146
8
ACCEPTED MANUSCRIPT 163
that vorinostat was toxic to C11 cells and A2 cells when its concentration exceeded 3 μM
164
based on the obvious reduction of the percentage of GFP-positive cells. Activity of chidamide as an HIV-1 reactivation agent was further confirmed in latent
166
HIV-1 chronically infected ACH-2 cells [27] and U1 cells [28], according to the production
167
of HIV-1 antigen p24 in the supernatant of cells cultured in the presence of LRAs. As shown
168
in Figure 1C and D, when the concentration of chidamide increased from 0 μM to 4 μM, p24
169
antigen production gradually rose. In ACH-2 cells, HIV-1 reactivation induced by chidamide
170
was positively correlated with 0 to 4 μM concentration, and its effect was frequently higher
171
than that of vorinostat (Fig. 1C). We also observed that the decreased activation of HIV-1
172
with vorinostat at 4 μM, as measured by p24 antigen production, was likely caused by
173
excessive cytotoxicity of vorinostat during continual 48 hours incubation in U1 cells (Fig. 1D).
174
These results demonstrated that chidamide potently induced HIV-1 LTR reactivation,
175
indicating its effect on HIV-1 production in a dose-dependent manner.
176
3.2. Chidamide activates HIV-1 expression in latent HIV-1 infected cells in a time-dependent
177
manner
TE D
M AN U
SC
RI PT
165
To assess the kinetics of HIV-1 LTR expression induced by chidamide, we performed a
179
kinetics experiment in which J-Lat A2 cells and ACH-2 cells were grown for 1 to 4 days with
180
or without chidamide for different concentrations. As shown in Figure 2A, after A2 cells were
181
treated with 1 μM chidamide, the percentage of GFP-positive cells increased over time and
182
rose to maximum (approximately 55%) on the fourth day. The kinetics of HIV-1 LTR
183
expression induction by vorinostat showed an apparent rise for the first 2 days, but then
184
tended toward constancy, or even reduction, by day 4 (Fig. 2A). We then tested the effect on
185
ACH-2 cells (Fig. 2B). From the first 3 days, we found that the effect of activation, as
186
measured by p24 antigen production, was almost the same between the treatment of
187
chidamide and vorinostat (Fig. 2B). In particular, p24 antigen production of the chidamide
AC C
EP
178
9
ACCEPTED MANUSCRIPT
group continuously increased after 3 days, while treatment with vorinostat resulted in only
189
negligible difference in variation. On the fourth day, we noticed that the effect of chidamide
190
on activation was significantly much higher than that of vorinostat in both cell lines (Fig. 2C
191
and D). These results indicated a time-dependent effect of chidamide on HIV-1 expression,
192
and as an HDAC inhibitor, chidamide could sustain reactivation over a period of time longer
193
than that shown by vorinostat.
194
3.3. Chidamide has minimal cytotoxicity in vitro
RI PT
188
To test the cytotoxicity of chidamide on different cells, we measured cell viability in A2,
196
C11, HEK 293 and PBMC cells treated with different concentrations of chidamide. After
197
treatment for 48h, cells were analyzed by CCK-8 assay. In the PBMC cells derived from
198
healthy HIV-negative donors, we did not find a large reduction in cell viability when the
199
concentration increased from 1 μM to 100 μM, while cell viability was clearly reduced at
200
concentrations > 25 μM, following treatment with vorinostat (Fig. 3A). In HEK293 T cells,
201
the CC50 was 37.3 μM (chidamide) and 12.5 μM (vorinostat) (Fig. 3B), respectively. This
202
result indicated that chidamide has relatively low toxicity compared to vorinostat on normal
203
human cells. The lower toxicity of chidamide compared with vorinostat was also observed in
204
J-Lat cells, including A2 cells and C11 cells, following incubation with chidamide at the same
205
concentrations as vorinostat based on the curve shown in Figure 3C and 3D, respectively.
206
CC50 of chidamide in J-Lat A2 cells and C11 cells was 22.3 μM and 18.6 μM, respectively.
207
CC50 of vorinostat in J-Lat A2 and C11 cells was 14 μM and 7.38 μM, respectively. Because
208
chidamide is an anticancer drug, it could be somewhat toxic to A2 and C11 cells from acute T
209
cell leukemia Jurkat cells. However, considering the lower toxicity in normal cells, such as
210
PBMCs and 293 T cells, our results illustrated that chidamide was safe at its active
211
concentration.
212
3.4. Chidamide reactivates HIV-1 latent provirus through NF-κB signaling
AC C
EP
TE D
M AN U
SC
195
10
ACCEPTED MANUSCRIPT
Binding sites for several inducible transcription factors, such as NF-κB, AP-1, and Sp1, can
214
be found in the HIV-1 LTR region [5, 10, 38]. Among these, the host transcription factor
215
NF-κB is critical for HIV-1 replication and has been most studied by LRAs [20, 39-44]. To
216
determine whether chidamide-mediated activation of HIV-1 is related to the NF-κB pathway,
217
we first examined protein expression of two components of the NF-κB pathway in J-Lat A2
218
cells by western blot analysis using antibodies specific for two NF-κB families, including
219
phospho-p65 (pp65) and IκBα. Our data showed a significant increase in the expression of
220
pp65 following treatment with chidamide as early as 2.5h (Fig. 4A and B), indicating that
221
NF-κB p65 was induced to be phosphorylated at Ser536 and transformed to pp65. The
222
activation of NF-κB usually involves degradation of the IκBα subunit bound to the NF-κB
223
dimer. Therefore, we next examined IκBα protein levels in J-Lat A2 cells stimulated with
224
chidamide (Fig. 4A). We observed an increased rate of IκB degradation following 2 hours of
225
treatment with chidamide (Fig. 4C). To dissect the involvement of the NF-κB pathway in viral
226
reactivation mediated by chidamide, the effect of some agents inhibiting the NF-κB pathway,
227
such as Aspirin and PDTC, was determined when used prior to chidamide treatment. J-Lat A2
228
cells were pretreated with different concentrations of aspirin, which could inhibit
229
TNF-α-induced activation of NF-κB [44, 45], and subsequently treated with chidamide or
230
TNF-α. We observed that the pretreatment of aspirin not only significantly inhibited
231
TNF-α-induced GFP expression in a dose-dependent manner, but also inhibited GFP
232
expression induced by chidamide at the concentrations tested (Fig. 4D). PDTC, a NF-κB
233
selective inhibitor [46], was also evaluated for HIV-1 p24 production in ACH-2 cells. The
234
increasing addition of PDTC to ACH-2 cells contributed to the significant inhibition of
235
chidamide-mediated reactivation from latency (Fig. 4E). Our findings collectively indicated
236
that chidamide-induced reactivation of latent HIV-1 might be mediated through the NF-κB
237
pathway.
AC C
EP
TE D
M AN U
SC
RI PT
213
11
ACCEPTED MANUSCRIPT 238
3.5. Combination of chidamide and anti-HIV-1 drugs inhibits viral infection To investigate whether chidamide could be combined with several anti-HIV-1 drugs to
240
inhibit active HIV-1 infection, we used TZM-Bl cells that contain a stably-integrated HIV-1
241
LTR linked to the luciferase reporter gene in their genome to examine the effect. ACH-2 cells
242
were treated with chidamide or anti-HIV-1 drugs, including AZT (Zidovudine), NVP
243
(Nevirapine), and IDV (Indinavir), for 4 days. Then we collected the supernatant of ACH-2
244
cells and added it to TZM-Bl cells to detect HIV-1 infection through luciferase assay. As
245
shown in Figure 5, in the absence of stimulation by chidamide, TZM-Bl cells did not appear
246
to express HIV-1 LTR with the treatment of anti-HIV drugs. When we performed chidamide
247
treatment alone, a 10-fold increase of HIV-1 expression could be evaluated by detecting the
248
expression of luciferase reporter gene, which demonstrated that 1) the latently infected HIV-1
249
was activated by chidamide, and 2) the reactivated HIV-1 could effectively infect and
250
replicate in the TZM-Bl cells that express CD4, CCR5 and CXCR4. By combining chidamide
251
with anti-HIV-1 drugs, the expression of HIV-1 LTR showed a rapid decrease, illustrating
252
that replication of the chidamide-reactivated HIV-1 in TZM-Bl cells was significantly
253
suppressed by the antivirus drugs tested. These results suggest that chidamide has a potential
254
to be used for HIV-1 functional cure based on the “shock and kill” strategy.
EP
TE D
M AN U
SC
RI PT
239
256
AC C
255
4. Discussion
257
Since HIV-1 remains integrated in the DNA of memory CD4+ T cells, cART for HIV-1
258
infection limits HIV-1 replication, but it still does not eliminate the virus [3]. The emergence
259
of the ‘‘shock and kill’’ strategy seems to be a harbinger of some new phase of HIV-1
260
therapeutics. This promising strategy means that the latent virus can be forced out of its
261
sanctuary, resulting in the eradication of the HIV-1 reservoir with targeted immunotherapy or
262
cART [5, 14, 47]. Therefore, identification of a highly effective LRA is critical for
12
ACCEPTED MANUSCRIPT
reactivation of most, if not all, of the latently infected provirus, in order to eliminate the latent
264
HIV-1 reservoir [48]. It was reported that some reactivators have been applied in clinical trials,
265
such as the HDAC inhibitors romidepsin [19], panobinostat [49], and vorinostat [16]. It is
266
well known that histone deacetylases (HDACs) belong to a family of enzymes equipped to
267
remove the acetyl group from histone lysine residues, inducing transcriptional repression
268
through chromatin condensation [50]. Thus, it can be seen that HDAC blocking is an
269
attractive means of inducing broad reactivation of latent HIV-1.
RI PT
263
Chidamide was a newly identified orally active benzamide class of HDAC inhibitor. In
271
recent years, researchers have made great effort in studying the HIV latency reversing effect
272
of the HDAC inhibitors belonging to the class of hydroxamic acids (e.g., vorinostat,
273
panobinostat and belinostat) or the class of cyclic peptide (e.g., apicidin and romidepsin).
274
However, few of the benzamide class of HDAC inhibitors have ever been reported to
275
reactivate the latent HIV provirus. Therefore, it is necessary to determine the ability of
276
chidamide to reverse HIV-1 latency. In this study, we explored chidamide’s potency in
277
reactivating latent HIV-1 expression on different latently infected cell models, including J-Lat
278
A2 cells, C11 cells, ACH-2 cells and U1 cells [28, 51]. Among these latent models, both C11
279
and J-Lat clone A2 cells are latently infected Jurkat T cells encoding GFP gene under the
280
control of HIV-1 LTR. The expression of GFP is used as a marker of latent HIV-1 expression,
281
and it can be detected by fluorescence microscopy and flow cytometry.
282
Jurkat T cells infected with pNL4-3-EGFP, which was reconstructed from pNL4-3 through the
283
introduction of gene-encoded enhancement of green fluorescent protein and mutation in Vpr
284
and Env [26]. Consequently, the same stimulation by chidamide or vorinostat could cause a
285
slight difference between these two cell lines. Moreover, as a marker of reactivation efficacy,
286
HIV-1 p24 antigen production levels were measured with chidamide treatment in ACH-2 and
287
U1 cells. These cell lines exhibit low levels of basal transcription, despite mutations in Tat
AC C
EP
TE D
M AN U
SC
270
C11 cells were
13
ACCEPTED MANUSCRIPT
(U1 cell) and TAR (ACH-2 cell) [27, 51]. In spite of the different integration, or mutation, site
289
in the cell lines from various origins, our results powerfully demonstrated that chidamide
290
could broadly and effectively activate HIV-1 production in a manner that was both
291
concentration- and time-dependent and do so in vitro at micromolar levels in these cells. It
292
was worth noting that chidamide induces HIV-1 LTR with lower toxicity in various cell lines
293
than vorinostat. Consistently, the benzamide class of HDAC inhibitors showed the least
294
pronounced toxicity in primary resting CD4+ T cells among most of the HDAC inhibitors [24].
295
Furthermore, in contrast to vorinostat, chidamide could retain its ability to reactivate HIV-1
296
much longer. Therefore, when carrying out the “shock and kill” strategy in clinical trials, we
297
recommend reducing the frequency of chidamide dosing in order to lessen its toxicity.
M AN U
SC
RI PT
288
After integration of HIV-1 into the host chromatin, proviral expression is largely controlled
299
by host cellular factors. Host transcription factors, including nuclear factor-κB (NF-κB),
300
nuclear factor of activated T cells (NFAT), AP1 and SP1, are sequestered in the cytoplasm in
301
resting cells and thus do not promote HIV-1 transcription until an appropriate cellular
302
activation signal is transmitted [38]. Among these transcription factors, NF-κB plays a central
303
role in the activation pathway of the HIV-1 provirus [10]. Therefore, the identification of
304
NF-κB activities has evoked a new direction in strategies to combat the effects of HIV-1
305
latency [31]. Chidamide could induce increasing levels of phospho-p65 expression and lead to
306
the degradation of IκBα which is an inhibited subunit in latently infected cells. It has been
307
reported that Aspirin can inhibit NF-κB activation induced by TNF-α by preventing the
308
phosphorylation and degradation of IκBα and nuclear translocation of NF-κB [45]. Therefore,
309
we examined the effect through Aspirin-inhibition experiments, following chidamide
310
treatment with TNF-α as a positive control, in J-Lat A2 cells. PDTC has been studied as a
311
NF-κB inhibitor, as well as an antioxidant agent, and we also showed that pretreatment of
312
ACH-2 cells with PDTC could remarkably prevent chidamide from inducing HIV-1
AC C
EP
TE D
298
14
ACCEPTED MANUSCRIPT
reactivation through expression of p24. The main activity of NF-κB on chidamide effects was
314
confirmed by using both inhibitors in viral reactivation experiments, which showed that
315
chidamide-mediated HIV-1 reactivation was abrogated. An increasing number of LRAs, such
316
as Oxaliplatin [30], M344 [44], Bryostatin [52], and PEP005 [40], have been demonstrated to
317
be involved in the NF-κB signaling pathway. However, besides M344, it has not be
318
demonstrated that other HDAC inhibitor, which contains a benzamide functional group, such
319
as entinostat and pimelic diphenylamide 106 [24], is involved in this pathway. Therefore, we
320
wanted to know if chidamide and other HDACs containing a benzamide functional group use
321
the NF-κB signal pathway or other pathways to activate latent HIV-1.
SC
RI PT
313
To achieve the “shock and kill” strategy for an HIV-1 cure, the role of anti-HIV-1 drugs on
323
viral clearance cannot be ignored. Therefore, we combined chidamide with anti-HIV-1 drugs
324
in order to determine if the active virus could be inhibited by antivirus drugs after activation
325
of HIV-1 from latently infected cells by chidamide. Among a variety of anti-HIV drugs, we
326
chose some drugs appearing to have optimal effect on eliminating HIV-1, such as nucleoside
327
reverse transcriptase inhibitors (NRTI), AZT , nonnucleoside reverse transcriptase inhibitors
328
(NNRTI), NVP, or the protease inhibitor IDV [53]. Our results showed that infection of
329
TZM-Bl cells by HIV-1 was significantly lower after treatment of AZT, NVP or IDV
330
cotreatment with chidamide, suggesting that treatments of such highly active antiretroviral
331
therapy could protect uninfected cells from becoming reinfected when using a combination of
332
HIV activators. Moreover, when combined with chidamide, we found that such anti-HIV-1
333
drugs as AZT, NVP and IDV did not lose their anti-HIV-1 potency, suggesting that these
334
anti-HIV-1 drugs can be used in combination therapy aimed at eliminating latent HIV-1
335
reservoirs.
AC C
EP
TE D
M AN U
322
336
To sum up, our results have demonstrated that chidamide plays an important role in histone
337
modification via the NF-κB pathway to regulate HIV-1 LTR gene expression and to reactivate
15
ACCEPTED MANUSCRIPT 338
latent HIV-1, thus having good potential to be used in combination with anti-HIV drugs for
339
HIV-1 functional cure.
340
Conflict of Interests
342
The authors declare no conflict of interests regarding the publication of this paper.
RI PT
341
343
Acknowledgments
SC
344
This work was supported by grants from the National Natural Science Foundation of China
346
(81630090 to S.J.; 81373456, 81672019 and 81661128041 to L.L.), the Sanming Project of
347
Medicine in Shenzhen, the National 863 Program of China (2015AA020930 to LL) and the
348
Shanghai Rising-Star Program (16QA1400300).
M AN U
345
EP AC C
350
TE D
349
16
ACCEPTED MANUSCRIPT
References
352
[1] Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al.
353
Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy.
354
Science 1997;278:1295-300.
355 356
RI PT
351
[2] Siliciano RF, Greene WC. HIV latency. Cold Spring Harb. Perspect. Med. 2011;1:a007096.
[3] Ruelas DS, Greene WC. An integrated overview of HIV-1 latency. Cell 2013;155:519-29.
358
[4] Siliciano JM, Siliciano RF. The remarkable stability of the latent reservoir for HIV-1 in
363 364 365 366 367
M AN U
362
the development of new therapeutic strategies. Retrovirology 2009;6:111. [6] Xing S, Siliciano RF. Targeting HIV latency: pharmacologic strategies toward eradication.
TE D
361
[5] Colin L, Van Lint C. Molecular control of HIV-1 postintegration latency: implications for
Drug Discov Today 2013;18:541-51.
[7] Margolis DM, Garcia JV, Hazuda DJ, Haynes BF. Latency reversal and viral clearance to
EP
360
resting memory CD4+ T cells. J. Infect. Dis. 2015;212:1345-7.
cure HIV-1. Science 2016;353:aaf6517. [8] Martin AR, Siliciano RF. Progress toward HIV eradication: case reports, current efforts,
AC C
359
SC
357
and the challenges associated with cure. Annu. Rev. Med. 2016;67:215-28.
368
[9] Coiras M, Lopez-Huertas MR, Perez-Olmeda M, Alcami J. Understanding HIV-1 latency
369
provides clues for the eradication of long-term reservoirs. Nat Rev Microbiol
370
2009;7:798-812.
371
[10] Williams SA, Chen LF, Kwon H, Ruiz-Jarabo CM, Verdin E, Greene WC. NF-kappaB p50
372
promotes HIV latency through HDAC recruitment and repression of transcriptional
17
ACCEPTED MANUSCRIPT 373
initiation. EMBO J. 2006;25:139-49. [11] Lucera MB, Tilton CA, Mao H, Dobrowolski C, Tabler CO, Haqqani AA, et al. The
375
histone deacetylase inhibitor vorinostat (SAHA) increases the susceptibility of uninfected
376
CD4+ T cells to HIV by increasing the kinetics and efficiency of postentry viral events. J.
377
Virol. 2014;88:10803-12.
RI PT
374
[12] Banga R, Procopio FA, Cavassini M, Perreau M. In vitro reactivation of
379
replication-competent and infectious HIV-1 by histone deacetylase inhibitors. J. Virol.
380
2015;90:1858-71.
M AN U
SC
378
381
[13] Deeks SG. HIV: Shock and kill. Nature 2012;487:439-40.
382
[14] Xu W, Li H, Wang Q, Hua C, Zhang H, Li W, et al. Advancements in developing strategies
383
for sterilizing and functional HIV cures. Biomed Res Int 2017;2017:6096134. [15] Laird GM, Bullen CK, Rosenbloom DI, Martin AR, Hill AL, Durand CM, et al. Ex vivo
385
analysis identifies effective HIV-1 latency-reversing drug combinations. J. Clin. Invest.
386
2015;125:1901-12.
TE D
384
[16] Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, et al.
388
Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy.
389
Nature 2012;487:482-5.
AC C
EP
387
390
[17] Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, et al. Activation
391
of HIV transcription with short-course vorinostat in HIV-infected patients on suppressive
392
antiretroviral therapy. PLoS Pathog. 2014;10:e1004473.
393
[18] Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A, et al.
394
Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected
18
ACCEPTED MANUSCRIPT 395
patients on suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. The
396
lancet. HIV 2014;1:e13-21. [19] Sogaard OS, Graversen ME, Leth S, Olesen R, Brinkmann CR, Nissen SK, et al. The
398
depsipeptide
romidepsin
399
2015;11:e1005142.
reverses
HIV-1
latency
in
vivo.
PLoS
Pathog.
RI PT
397
[20] Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. Eradicating HIV-1
401
infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 2014;12:750-64.
402
[21] Gama L, Abreu CM, Shirk EN, Price SL, Li M, Laird GM, et al. Reactivation of simian
403
immunodeficiency virus reservoirs in the brain of virally suppressed macaques. AIDS
404
2017;31:5-14.
M AN U
SC
400
[22] Gu R, Liu T, Zhu X, Gan H, Wu Z, Li J, et al. Development and validation of a sensitive
406
HPLC-MS/MS method for determination of chidamide (epidaza), a new benzamide class
407
of selective histone deacetylase inhibitor, in human plasma and its clinical application. J.
408
Chromatogr. B Analyt. Technol. Biomed. Life Sci. 2015;1000:181-6.
TE D
405
[23] Lu X, Ning Z, Li Z, Cao H, Wang X. Development of chidamide for peripheral T-cell
410
lymphoma, the first orphan drug approved in China. Intractable Rare Dis Res
411
2016;5:185-91.
AC C
EP
409
412
[24] Kobayashi Y, Gelinas C, Dougherty JP. Histone deacetylase inhibitors containing a
413
benzamide functional group and a pyridyl cap are preferentially effective human
414
immunodeficiency virus-1 latency-reversing agents in primary resting CD4+ T cells. J.
415
Gen. Virol. 2017;98:799-809.
416
[25] Jordan A, Bisgrove D, Verdin E. HIV reproducibly establishes a latent infection after
19
ACCEPTED MANUSCRIPT 417
acute infection of T cells in vitro. EMBO J. 2003;22:1868-77. [26] Ding D, Qu X, Li L, Zhou X, Liu S, Lin S, et al. Involvement of histone methyltransferase
419
GLP in HIV-1 latency through catalysis of H3K9 dimethylation. Virology
420
2013;440:182-9.
RI PT
418
[27] Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, et al. Tumor necrosis
422
factor alpha induces expression of human immunodeficiency virus in a chronically
423
infected T-cell clone. Proc. Natl. Acad. Sci. U. S. A. 1989;86:2365-8.
SC
421
[28] Clouse KA, Powell D, Washington I, Poli G, Strebel K, Farrar W, et al. Monokine
425
regulation of human immunodeficiency virus-1 expression in a chronically infected
426
human T cell clone. J. Immunol. 1989;142:431-8.
M AN U
424
[29] Contreras X, Schweneker M, Chen CS, McCune JM, Deeks SG, Martin J, et al.
428
Suberoylanilide hydroxamic acid reactivates HIV from latently infected cells. J. Biol.
429
Chem. 2009;284:6782-9.
TE D
427
[30] Zhu X, Liu S, Wang P, Qu X, Wang X, Zeng H, et al. Oxaliplatin antagonizes HIV-1
431
latency by activating NF-kappaB without causing global T cell activation. Biochem.
432
Biophys. Res. Commun. 2014;450:202-7.
434
AC C
433
EP
430
[31] Lu P, Qu X, Shen Y, Jiang Z, Wang P, Zeng H, et al. The BET inhibitor OTX015 reactivates latent HIV-1 through P-TEFb. Sci. Rep. 2016;6:24100.
435
[32] Li W, Yu F, Wang Q, Qi Q, Su S, Xie L, et al. Co-delivery of HIV-1 entry inhibitor and
436
nonnucleoside reverse transcriptase inhibitor shuttled by nanoparticles: cocktail
437
therapeutic strategy for antiviral therapy. AIDS 2016;30:827-38.
438
[33] Yu Y, Deng YQ, Zou P, Wang Q, Dai Y, Yu F, et al. A peptide-based viral inactivator
20
ACCEPTED MANUSCRIPT 439
inhibits Zika virus infection in pregnant mice and fetuses. Nat Commun 2017;8:15672. [34] Chou TC, Talalay P. Quantitative analysis of dose-effect relationships: the combined
441
effects of multiple drugs or enzyme inhibitors. Adv. Enzyme Regul. 1984;22:27-55.
442
[35] Jiang S, Lu H, Liu S, Zhao Q, He Y, Debnath AK. N-substituted pyrrole derivatives as
443
novel human immunodeficiency virus type 1 entry inhibitors that interfere with the gp41
444
six-helix bundle formation and block virus fusion. Antimicrob. Agents Chemother.
445
2004;48:4349-59.
447
SC
[36] Gan J, Wang C, Jin Y, Guo Y, Xu F, Zhu Q, et al. Proteomic profiling identifies the
M AN U
446
RI PT
440
SIM-associated complex of KSHV-encoded LANA. Proteomics 2015;15:2023-37. [37] Sun Z, Zhu Y, Wang Q, Ye L, Dai Y, Su S, et al. An immunogen containing four tandem
449
10E8 epitope repeats with exposed key residues induces antibodies that neutralize HIV-1
450
and activates an ADCC reporter gene. Emerg Microbes Infect 2016;5:e65.
TE D
448
[38] Victoriano AF, Imai K, Togami H, Ueno T, Asamitsu K, Suzuki T, et al. Novel histone
452
deacetylase inhibitor NCH-51 activates latent HIV-1 gene expression. FEBS Lett.
453
2011;585:1103-11.
EP
451
[39] Jiang G, Mendes EA, Kaiser P, Sankaran-Walters S, Tang Y, Weber MG, et al.
455
Reactivation of HIV latency by a newly modified Ingenol derivative via protein kinase
456
Cdelta-NF-kappaB signaling. AIDS 2014;28:1555-66.
AC C
454
457
[40] Jiang G, Mendes EA, Kaiser P, Wong DP, Tang Y, Cai I, et al. Synergistic reactivation of
458
latent HIV expression by ingenol-3-angelate, PEP005, targeted NF-κB signaling in
459
combination with JQ1 induced p-TEFb activation. PLoS Pathog. 2015;11:e1005066.
460
[41] Stroud JC, Oltman A, Han A, Bates DL, Chen L. Structural basis of HIV-1 activation by
21
ACCEPTED MANUSCRIPT 461
NF-kappaB--a higher-order complex of p50:RelA bound to the HIV-1 LTR. J. Mol. Biol.
462
2009;393:98-112.
464 465 466
[42] Wang P, Qu X, Wang X, Liu L, Zhu X, Zeng H, et al. As2O3 synergistically reactivate latent HIV-1 by induction of NF-kappaB. Antiviral Res. 2013.
RI PT
463
[43] Williams SA, Chen LF, Kwon H, Fenard D, Bisgrove D, Verdin E, et al. Prostratin antagonizes HIV latency by activating NF-kappaB. J. Biol. Chem. 2004;279:42008-17. [44] Ying H, Zhang Y, Zhou X, Qu X, Wang P, Liu S, et al. Selective histonedeacetylase
468
inhibitor M344 intervenes in HIV-1 latency through increasing histone acetylation and
469
activation of NF-kappaB. PLoS One 2012;7:e48832.
471
M AN U
470
SC
467
[45] Kutuk O, Basaga H. Aspirin inhibits TNFalpha- and IL-1-induced NF-kappaB activation and sensitizes HeLa cells to apoptosis. Cytokine 2004;25:229-37. [46] Alvarez S, Blanco A, Kern F, Fresno M, Munoz-Fernandez MA. HIV-2 induces
473
NF-kappaB activation and cyclooxygenase-2 expression in human astroglial cells.
474
Virology 2008;380:144-51.
EP
476
[47] Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. The challenge of finding a cure for HIV infection. Science 2009;323:1304-7.
AC C
475
TE D
472
477
[48] Gallo RC. Shock and kill with caution. Science 2016;354:177-8.
478
[49] Clutton G, Xu Y, Baldoni PL, Mollan KR, Kirchherr J, Newhard W, et al. The differential
479
short- and long-term effects of HIV-1 latency-reversing agents on T cell function. Sci. Rep.
480
2016;6:30749.
481 482
[50] Mottamal M, Zheng S, Huang TL, Wang G. Histone deacetylase inhibitors in clinical studies as templates for new anticancer agents. Molecules 2015;20:3898-941.
22
ACCEPTED MANUSCRIPT 483 484
[51] Folks TM, Justement J, Kinter A, Dinarello CA, Fauci AS. Cytokine-induced expression of HIV-1 in a chronically infected promonocyte cell line. Science 1987;238:800-2. [52] Mehla R, Bivalkar-Mehla S, Zhang R, Handy I, Albrecht H, Giri S, et al. Bryostatin
486
modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute
487
infection in a receptor independent manner. PLoS Onke 2010;5:e11160.
489
[53] Staszewski S. Update on study 006--EFV + AZT + 3TC versus the current 'standard of care' IDV + AZT + 3TC. Int. J. Clin. Pract. Suppl. 1999;103:10-5.
SC
488
RI PT
485
M AN U
490
AC C
EP
TE D
491
23
ACCEPTED MANUSCRIPT
Figure legends
493
Fig. 1. Reactivation of latent HIV-1 in different latently infected cells by chidamide. (A) J-Lat
494
clone C11 cells were treated with chidamide and vorinostat for 48 h at the indicated
495
concentrations. Results are expressed as a percentage of GFP-positive cells within the entire
496
population. (B) J-Lat clone A2 cells treated with chidamide and vorinostat for 48 h at the
497
indicated concentrations and analyzed as in (A). ACH-2 cells (C) or U1 (D) cells were
498
stimulated with increasing concentrations of chidamide and vorinostat for 48 h. Viral
499
replication was measured in the supernatant of the cells using p24 ELISA. Data show
500
dose-dependent effects of chidamide on HIV-1 production in different latently infected cells
501
and represent the means ± standard deviations of three independent experiments.
M AN U
SC
RI PT
492
502
Fig. 2. Time-dependent effects of chidamide on HIV-1 production. J-Lat A2 cells (A) or
504
ACH-2 cells (B) were treated with DMSO or with 1μM chidamide or vorinostat for 0, 24, 48,
505
72, 96 h. A time-dependent curve on HIV-1 transcription is represented by two latent cell
506
models. (C) and (D) represented the comparison of the effect on activating latently infected
507
cells between chidamide and vorinostat at the third day and the fourth day. Data show
508
time-dependent effects of chidamide on activating latently infected cell lines and represent the
509
means ± standard deviations of three independent experiments. (**P < 0.01 and***P < 0.001).
AC C
EP
TE D
503
510
511
Fig. 3. Viability of cells treated with chidamide or vorinostat. PBMCs from HIV-negative
512
donors (A), HEK293 T cells (B), J-Lat A2 cells (C) and C11 cells (D) were treated with
24
ACCEPTED MANUSCRIPT
chidamide and vorinostat from 0 to 100 μM for 48 h, and cytotoxicity was measured by
514
CCK-8 kit. The absorbance at 450 nm (OD450) of each well was determined as the readout of
515
cell viability. Data represent the means ± standard deviations of three independent
516
experiments.
RI PT
513
517
Fig. 4. Reactivation of HIV-1 latent provirus by chidamide through NF-κB pathway. (A) J-Lat
519
A2 cells were treated with 2 μM of chidamide for up to 3.5 h hours. Western blot analysis was
520
performed to detect the expression of p-p65 and IκBα. (B) Quantitation of phosphorylation of
521
p65 in J-Lat A2 cells after 3.5 hours treatment with chidamide in panel (A). Relative band
522
intensities from three independent experiments in J-Lat A2 cells, as determined using Image J
523
(NIH), are shown in the bar graph. (C) Quantitation of IκBα in J-Lat A2 cells after 3.5 hours
524
treatment with chidamide in panel (A). Relative band intensities from three independent
525
experiments in J-Lat A2 cells, as determined using ImageJ (NIH), are shown in the bar graph.
526
(D) J-Lat A2 cells were pretreated with various concentrations of Aspirin for 3 h and
527
subsequently treated with chidamide (1, 2 and 5 μM), TNF-a (10 ng/mL) or DMSO as control
528
for 48 h. The percentage of GFP-positive cells in drug-treated cells in the absence or presence
529
of the inhibitor Aspirin was measured by flow cytometry. (E) ACH-2 cells were pretreated
530
with increasing concentrations of PDTC for 2 h and subsequently treated with chidamide (2
531
μM); p24 production from latently infected cells was measured by p24 ELISA. Data represent
532
the means ± standard deviations of three independent experiments. (*P < 0.05, P < **0.01 and
533
***P < 0.001).
534
AC C
EP
TE D
M AN U
SC
518
25
ACCEPTED MANUSCRIPT
Fig. 5. Combination of chidamide with anti-HIV-1 drugs to suppress infection of reactivated
536
HIV-1. (a) 1×104 ACH-2 cells were treated with chidamide, vorinostat or PBS in the presence
537
or absence of the anti-HIV drugs AZT, NVP, or IDV for 4 days. The supernatants of ACH-2
538
cells containing the residual reactivated HIV-1 were cultured with TZM-Bl cells for 48 hours
539
before testing residual HIV-1 infectivity using the luciferase assay. Data represent the means ±
540
standard deviations of three independent experiments (***P < 0.001).
AC C
EP
TE D
M AN U
SC
RI PT
535
Figure 1
ACCEPTED MANUSCRIPT
Fig. 1. C11 cells
A2 cells
A
AC C
C
EP
ACH-2 cells
TE D
M AN U
SC
RI PT
B
U1 cells D
Figure 2
Fig. 2.
ACCEPTED MANUSCRIPT
A2 cells A A
ACH-2 cells
EP AC C
C C
TE D
M AN U
SC
RI PT
B
D
Figure 3
ACCEPTED MANUSCRIPT
Fig. 3. PBMCs
HEK 293 T cells
A
AC C
C
EP
A2 cells
TE D
M AN U
SC
RI PT
B
C11 cells
D
Figure 4
Fig. 4.
ACCEPTED MANUSCRIPT
B
Chidamide 0h
1h
2h
2.5 h 3 h
3.5 h
p-p65 (S536) β-actin
SC
C
M AN U
IκBα
AC C
EP
TE D
β-actin
D
RI PT
A
E
Figure 5
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
RI PT
Fig. 5.
*Detailed Response to Reviewers
ACCEPTED MANUSCRIPT
Responses to the Reviewers' comments
Reviewer #1: Minor comments
into that of resting…"
RI PT
1. Line 44: "integration of HIV-1 into into", change to "integration of HIV-1 genome
2. Line 51-52, "histone deacetylation" should be "histone deacetylase", because that
3. Line 111 "twenty µL" change to "20 µl".
SC
is what HDAC refers to in the rest part of the manuscript.
Response: We thank the reviewer for the constructive comments, and we have
M AN U
corrected these errors accordingly in the revised manuscript.
4. In the cytotoxicity assay (Figure 3), how were the CC50 values determined? If a nonlinear regression model is used for curving fitting, please describe the details in the method.
TE D
Response: We used the Calcusyn software program (Biosoft, Ferguson, MO) to calculate CC50 values and we have added the description of the method and
EP
references in the Materials and Methods section accordingly.
5. Line 190, "The lower toxicity compared …" change to "The lower toxicity of
AC C
chidamide compared …”
6. Line 236, "latently infected HIV-1" change to "reactivated HIV-1" 7. Line 240-243, "In the presence of anti-HIV-1 drugs …"should be rewritten to, e.g. "These results indicated that anti-HIV-1 drugs could prevent the spread of newly synthesized viruses reactivated by chidamide." Response: Thanks, we have changed these sentences in the revised manuscript according to the reviewer's suggestion.
Reviewer #2:
ACCEPTED MANUSCRIPT 1. In line 247 where the "shock and kill" strategy is described, the following reference (Gallo RC. Shock and kill with caution. Science 2016;354:177-8) should be cited and the related points should be discussed. Response: We appreciate the reviewer's insightful suggestion to improve our
RI PT
manuscript. We have cited this reference and added relevant discussion in the revised manuscript.
2. In the Materials and Methods, the method for Western blot should be provided.
SC
Response: We have added the following paragraph in the revised manuscript:
“Western blot analysis was performed as described [1]. Cells were harvested, washed
M AN U
once with ice-cold PBS, and lysed in ice-cold RIPA buffer [50 mM Tris (pH 8.0), 150 mM NaCl, 1% NP-40, and 2 mM EDTA)] containing PMSF and phosphatase inhibitor cocktails (Thermofisher Scientific, Omaha, NE) for 15 minutes. Protein samples were separated by SDS-PAGE and transferred onto nitrocellulose membrane (Pall Corporation, Port Washington, NY). After blocking with 5% non-fat dry milk in
TE D
PBS containing 0.1% Tween 20, proteins of interest were probed with the corresponding primary antibodies [pp65 (Ser536) and IκBα antibodies obtained from Cell Signaling (Danvers, MA)], followed by appropriate infrared-conjugated
EP
secondary antibodies, Alexa Fluor 800 or 680 (Carlsbad, CA). The immunoblotting
AC C
was carried out using the LiCOR (Lincoln, NE) Odyssey scanner.”
3. In line 208, the space between "Ser" and "536" should be deleted. Response: We have corrected this error.
4. In the Figure 5, the scale on the Y axis should be changed to scientific notation. 5. Figure 1A and 1B, Figure 2A and 2C, and Figure 4D, the "GFP-Positive cells (%)" in the Y-axis should be "GFP-positive cells (%)". Response: We have made correction accordingly.
Reviewer #3:
ACCEPTED MANUSCRIPT Minor comments 1. The authors could discuss in more detail about the advantages of chidamide as a LRA, compared with other HDACi-based anti-cancer drugs. Response: We appreciate the reviewer's insightful comment and have added the
RI PT
chidamide’s advantages in the Discussion section in the revised manuscript.
2. Some repeated sentences about the result in the Discussion section should be removed.
SC
Response: We have deleted the redundant parts in the revised manuscript.
M AN U
3. In the legend of Fig. 5, "ACH-2 cells were treated with chidamide, vorinostat or PBS in the presence or absence of the anti-HIV drugs AZT, NVP, or IDV for 96 h", while in the Results (lines 228-230), " ACH-2 cells were treated with chidamide or anti-HIV-1 drugs, including AZT (Zidovudine), NVP (Nevirapine), and IDV (Indinavir), for 4 days". To keep consistence, 96 h should be changed to 4 days.
TE D
Response: Thanks, we have corrected it accordingly.
4. The manuscripts need some proofreads in English.
EP
Response: We have thoroughly checked our revised manuscript, paying attention to English grammar, spelling, punctuation, subject/verb agreement, and proper use of
AC C
singular and plural as well as meaning.
References: [1]
Gan J, Wang C, Jin Y, Guo Y, Xu F, Zhu Q, et al. Proteomic profiling identifies the SIM-associated complex of KSHV-encoded LANA. Proteomics 2015;15:2023-37.