AAC Accepted Manuscript Posted Online 20 April 2015 Antimicrob. Agents Chemother. doi:10.1128/AAC.00574-15 Copyright © 2015, American Society for Microbiology. All Rights Reserved.

1

Histone Deacetylase Inhibitor Romidepsin

2

Inhibits de novo HIV-1 Infections

3 4

Kasper L. Jønsson1,2§, Martin Tolstrup3,4§, Johan Vad-Nielsen3, Kathrine Kjær1, Anders Laustsen1,2,

5

Morten N. Andersen1; Thomas A. Rasmussen3, Ole S. Søgaard3,4, Lars Østergaard3,4, Paul W.

6

Denton3,4,5# and Martin R. Jakobsen1,2#.

7 8

1

Institute of Biomedicine, Faculty of Health, Aarhus University, Denmark

9

2

Aarhus Research Centre of Innate Immunology, Denmark

10

3

Department of Infectious Diseases, Aarhus University Hospital Skejby, Denmark

11

4

Institute of Clinical Medicine, Aarhus University, Denmark

12 13

5

Aarhus Institute for Advanced Studies, Aarhus University, Denmark

15

§

These authors contributed equally to this work.

16

#

Equal contributors and co-corresponding authors:

14

Paul W. Denton, PhD Department of Infectious Diseases (Q) Aarhus University Hospital, Skejby Palle Juul-Jensens Boulevard 99 8200 Aarhus N, Denmark [email protected]

Martin R. Jakobsen, PhD Institute of Biomedicine Faculty of Health, Aarhus University Wilhelm Meyers Alle 4 8000 Aarhus C, Denmark [email protected]

17 18 19

Running title: Romidepsin Inhibits de novo HIV Infections

20 21

Keywords: HIV, romidepsin, viral outgrowth, latency, HDACi

1

22

ABSTRACT

23

Adjunct therapy with the histone deacetylase inhibitor (HDACi) romidepsin increases plasma

24

viremia in HIV patients on combination antiretroviral therapy (cART). However, a potential

25

concern is that reversing HIV latency with an HDACi may reactivate the virus in anatomical

26

compartments with sub-optimal cART concentrations leading to de novo infection of susceptible

27

cells in these sites. We tested physiologically relevant romidepsin concentrations known to

28

reactivate latent HIV in order to definitively address this concern. We found that romidepsin

29

significantly inhibited HIV infection in PBMCs and CD4+ T cells, but not in monocyte-derived-

30

macrophages. In addition, romidepsin impaired HIV spreading in CD4+ T cell cultures. When we

31

evaluated the impact of romidepsin on quantitative viral outgrowth assays with primary resting

32

CD4+ T cells, we found that resting CD4+ T cells exposed to romidepsin exhibited reduced

33

proliferation and viability. This significantly lowered assay sensitivity when measuring the efficacy

34

of romidepsin as an HIV latency reversal agent. All together our data indicate that romidepsin-

35

based HIV eradication strategies are unlikely to reseed a latent T cell reservoir, even under sub-

36

optimal cART conditions, because romidepsin profoundly restricts de novo HIV infections.

2

37 38

INTRODUCTION Combination antiretroviral therapy (cART) greatly reduces HIV disease-related mortality, but

39

does not cure HIV infection. During cART, transcriptionally silent HIV persists in resting CD4+ T

40

cells as a latent HIV reservoir (1). If cART is interrupted, viral replication is reinitiated from this

41

reservoir. The resumption of viral replication typically manifests clinically as rebound in plasma

42

viremia accompanied by a rapid decline in CD4+ T cells. Thus, lifelong cART is essential for

43

continued virus suppression. The primary goal of HIV eradication therapies is to eliminate the latent

44

HIV reservoir such that cART can be interrupted without viral rebound (2). Currently, activating

45

HIV from latency such that viral cytopathic effects or the host immune system kills the infected

46

cells is under investigation as a curative strategy. Key to the success of such strategies, collectively

47

referred to as “kick and kill” approaches, is the ability to effectively reverse HIV latency in vivo (3).

48 49

The mechanisms by which HIV establishes latency are complex and include enzymatic

50

processes that affect the chromatin organization of the HIV-promoter region, one of the key

51

determinants of virus transcriptional activity (4-7). Histone deacetylation mediated by histone

52

deacetylases (HDAC) leads to structural changes in chromatin that inhibit transcription (2, 6, 8).

53

Conversely, histone deacetylase inhibitors (HDACi) turn on gene transcription by promoting

54

acetylation of lysine residues on histones leading to chromatin relaxation (9). There are 4 classes of

55

HDACs into which the 18 described enzymes have been categorized (10). Relevant to HIV

56

eradication strategies, class I HDACs are particularly important for maintaining HIV latency (10,

57

11). HDACi, including romidepsin and panobinostat, may serve as latency reactivating agents

58

(LRA) via direct interference with HDAC maintained latency (9, 12). Romidepsin exhibits

59

inhibitory activity in the lower nanomolar range against class I HDACs and panobinostat exhibits

60

activity against class I/II HDACs (13). Consistent with the close interactions between HDACs and

3

61

the maintenance of HIV latency, HDACi have the ability to reactivate and induce HIV expression

62

from latently infected cells in both ex vivo and in vivo studies (9, 14-16).

63 64

Recently, Lucera, et al. reported that supra-physiological concentrations (200 nM) of

65

romidepsin increased the susceptibility of CD4+ T cells to HIV infection (17). These data, as well

66

as those of others (18), raise the concern that inducing latent HIV in anatomical compartments with

67

sub-optimal cART concentrations (19) could lead to the infection of new target cells and reseeding

68

of the latent reservoir. To determine the likelihood of such paradoxical outcomes, we

69

comprehensively analyzed the impact of HDACi on de novo virus infection employing a broad

70

panel of ex vivo experimental systems. We found that romidepsin-based HIV eradication strategies

71

are unlikely to reseed the latent reservoir because this drug profoundly restricted de novo HIV

72

infections. Further, resting CD4+ T cells exposed to romidepsin exhibited reduced proliferation and

73

viability in the viral outgrowth assay which led to significantly lower assay sensitivity when

74

measuring the efficacy of romidepsin as an HIV latency reversal agent.

4

75

METHODS AND MATERIALS

76

Source and Isolation of Primary Human Cells

77

Healthy blood donor buffy coat fractions were obtained from the Department of Clinical

78

Immunology Blood Bank at Aarhus University Hospital, Denmark. Peripheral blood mononuclear

79

cells (PBMCs) were isolated using Ficoll density separation. CD4+ T cells were enriched by

80

negative depletion from PBMCs using EasySep Human CD4+ T Cell Enrichment Kit (Cat #:

81

19052; Stem Cell Technologies) or CD4+ T Cell Isolation Kit (Cat #: 130-096-533, Miltenyi

82

Biotec) according to the respective manufacturer’s protocol. Monocyte-derived-macrophages

83

(MDMs) were generated from monocytes purified from PBMCs by plastic adherence, with

84

subsequent culture for 7 days in “RPMI growth medium” [RPMI 1640; 50,000 I.U. Penicillin;

85

50,000 µg Streptomycin; 10% fetal calf serum; 1% glutamine; IL-2 (20U/ml)] supplemented

86

with 10 ng/mL M-CSF and 1 ng/mL GM-CSF (PeproTech). Resting CD4+ T cells were enriched

87

from cryopreserved PBMCs by negative depletion via a 2-step protocol as previously described

88

(20). Briefly, the first step was to enrich CD4+ T cells from PBMCs using Miltenyi CD4+ T Cell

89

Isolation Kit (Cat #: 130-096-533) according to the manufacturer’s protocol. The second step was to

90

further enrich for resting CD4+ T cells via depletion of cells expressing CD69, CD25 or HLA-DR

91

(Miltenyi Cat #: CD69 Microbeads Kit II– 130-092-355; CD25 Microbeads II – 130-092-983;

92

HLA-DR Microbeads– 130-046-101). All cell incubations at 37°C unless otherwise noted. The

93

purity of enriched cells populations was assessed by flow cytometry using either a LSR Fortessa

94

(BD Biosciences) or a FACSVerse (BD Biosciences) flow cytometer and FlowJo software

95

(Treestar).

96 97

Viability Assay for TZM-bl Cells, PBMCs and Activated CD4+ T Cells

5

98

The viability of TZM-bl cells (21-25) and primary human cells was assessed following exposure to

99

romidepsin (Selleckchem) or panobinostat (Selleckchem) (26-28). We tested a range of drug doses

100

that spanned the physiologically relevant doses of each drug [i.e. 40nM romidepsin (14) and 30nM

101

panobinostat (15, 29)] and in select experiments, sub-physiological doses of romidepsin were

102

included along with media controls to facilitate the observation of dose-dependent effects on HIV

103

infection.

104 105

TZM-bl cells in “cDMEM” [DMEM; 50,000 I.U. Penicillin; 50,000 µg Streptomycin; 10% fetal

106

calf serum; 1% glutamine] were seeded (5x103 cells/well) in 96-well plates and incubated for 24

107

hours- and then primed (8 or 18 hours) with romidepsin or panobinostat at the indicated range of

108

concentrations (two-fold dilutions). Following priming, the cells were washed with cDMEM and

109

incubated for 48 hours. TZM-bl cells were trypsinized, resuspended in 100uL cDMEM, transferred

110

to 96-well ELISA plates and 20uL of MTS substrate (Promega - CellTiter 96® AQueous One

111

Solution Cell Proliferation Assay) was added to each well. Plates were incubated for 4 hours and

112

formazan production was then analyzed with an ELISA plate reader (Biotek - ELx808). Cell

113

viability was calculated according to the Promega CellTiter-protocol.

114 115

PBMCs CD4+ T cells were stimulated in “RPMI growth medium” supplemented with PHA

116

(5µg/mL) for 48 hours. PHA containing media was replaced with fresh RPMI growth medium and

117

cells were incubated for 24 hours. PBMCs or CD4+ T cells were then seeded (2x105 cells/well) in

118

96-well plates and primed (8 or 18 hours) with romidepsin or panobinostat at the indicated range of

119

concentrations (two-fold dilutions). Following priming, the cells were washed, resuspended in

120

RPMI growth medium and incubated for 48 hours. Cells were resuspended in 100µL RPMI growth

121

media and assayed for formazan production as described above.

6

122 123

Single Round, VSV-g Pseudotyped HIV Infection Assay

124

VSVg-pseudotyped HIV virions were produced by transfecting HEK293T cells with a packaging

125

system consisting of 4 plasmids: pCCL-PGK-eGFP, pMD.2G, pRSV-REV, and pMDlg/p-RRE.

126

Viral supernatants were harvested after 48 and 72 hours and concentrated through a 20% sucrose

127

cushion (25,000 x g for 2 hours). Viral pellets were resuspended in PBS, pooled and aliquots were

128

stored at -80oC.

129 130

PBMCs or enriched CD4+ T cells were stimulated in RPMI growth medium supplemented with

131

PHA (5ug/mL) for 48 hours. PHA containing media was replaced with fresh RPMI growth medium

132

and PBMCs or enriched CD4+ T cells were incubated for 24 hours. MDMs were grown as

133

described above. Cells were seeded (2x105 cells/well) in 48-well plates with RPMI growth medium

134

and pulsed for 2 hours with romidepsin (10nM; 40nM) or panobinostat (30nM). VSVg-pseudotyped

135

HIV virions were then inoculated into the cell cultures along with polybrene (6 µg/ml) followed by

136

incubation for an additional 6 hours. Cells were then washed to remove unbound virions and

137

resuspended in fresh RPMI growth medium and incubated for 48 hours. Cells were harvested by

138

aspiration for PBMCs and T cells or following EDTA treatment for MDMs. Cells were incubated

139

on ice with LIVE/DEAD Fixable Violet Dead Cell Stain Kit-405 (Life Technologies) according to

140

manufacturer’s instructions. Data were collected using a LSR Fortessa flow cytometer. Viable cells

141

exhibiting eGFP expression were characterized as infected.

142 143

HIV Spreading Assay

144

Enriched CD4+ T cells were stimulated in RPMI growth medium supplemented with PHA

145

(5µg/mL) for 48 hours. PHA containing media was replaced with fresh RPMI growth medium and

7

146

cells were incubated for 24 hours. CD4+ T cells were pulsed for 2 hours with romidepsin (10nM;

147

40nM) or panobinostat (30nM). Cells were then infected with HIV-1HXB2 (30). After an additional 4

148

hours of incubation, the HDACi and virus inoculum were removed and cells were resuspended in

149

fresh RPMI growth medium. At the indicated time points, culture supernatants were harvested and

150

evaluated for p24 antigen by ELISA to determine the replication kinetics of HIV as previously

151

described (9).

152 153

Romidepsin and Interferon-Stimulated Genes in PBMCs

154

PHA-activated PBMCs were seeded (1x105 cells/well) into a conical 96-well plate and primed for 2

155

or 8 hours with romidepsin (10nM; 40nM) or IFNβ (1000 IU/mL). Following priming, RNA was

156

collected from the cells using a High Pure RNA Isolation kit (Cat #: 11828665001, Roche)

157

according to manufacturer’s instructions. The RNA expression levels of 39 different genes were

158

subsequently analyzed using the Fluidigm 48-48 multiplex Gene array Biomark system, including

159

TaqMan probes (Fluidigm, USA) according to manufacturer’s instructions.

160 161

Resting CD4+ T Cell Proliferation and Viability Assay

162

Resting CD4+ T cells were incubated with eFluor-670 (Cat. #: 65-0840-85; eBioscience) according

163

to manufacturer’s instructions. Dyed resting CD4+ T cells (1x106 cells per well) were placed into a

164

24-well plate in “cRPMI+IL2 medium” [RPMI with L-glutamine; 1% streptomycin and penicillin;

165

10% fetal calf serum; IL-2 (10,000U/ml); conditioned media from a mix lymphocyte reaction

166

culture as described in (31)] plus PHA (0.5 µg/ml) and/or romidepsin (4 nM; 40 nM). Following an

167

18-hour incubation, the cultures were washed twice (2 hour interval) with cRPMI+IL2 medium and

168

then maintained in 2ml cRPMI+IL2 medium for 4 additional days. Cells were incubated on ice with

169

LIVE/DEAD Fixable Green Dead Cell Stain Kit-488 (Cat. #: L-23101; Life Technologies)

8

170

according to manufacturer’s instructions. Data were collected using a FACSVerse flow cytometer.

171

Viable cells exhibiting a dilution of eFluor-670 were characterized as having proliferated during the

172

culture period. Fold proliferation was determined relative to the respective human donor “media

173

only” culture. The percentage of viable cells represents the proportion of resting CD4+ T cells

174

which excluded the fixable green dead cell stain.

175 176

Activation of Latent HIV in ACH2 Cells

177

ACH2 cells (12, 32, 33) were incubated for 18 hours +/- PHA (0.5 µg/ml) or romidepsin (4 nM; 40

178

nM), washed twice (2 hour interval) with cRPMI+IL2 medium and then incubated in fresh

179

cRPMI+IL2 medium for 48 hours. Viral p24 antigen was measured in culture supernatants by

180

ELISA as an indicator of the ability of either romidepsin or PHA to induce virus production from

181

this latent HIV cell model (9).

182 183

Modified Viral Outgrowth Assay

184

The viral outgrowth assay was performed essentially as described (34). Briefly, resting CD4+ T

185

cells were isolated from healthy blood donor buffy coat fractions. On Day 0, 1x106 resting CD4+ T

186

cells per well from each donor were plated into a 24-well plate together with the indicated number

187

of ACH2 cells in cRPMI+IL2 medium with PHA (0.5 µg/ml) or romidepsin (4 nM; 40 nM). After

188

18 hours, the cultures were washed twice (2 hour interval) with cRPMI+IL2 medium, cultures were

189

moved into 6-well plates and fed with 5x105 MOLT-4/CCR5 cells in a final volume of 8ml

190

cRPMI+IL2 medium. On Day 7, 6ml of the culture volume was replaced with fresh cRPMI+IL2

191

medium. On Day 14, 200ul of each culture supernatant was transferred to TZM-bl cells in duplicate

192

and luciferase activity from these reporter cells was measured on Day 16 as an indicator of HIV

193

production within a given culture (15).

9

194 195

Statistics

196

Comparisons of infectivity within the single round infection assay were performed using t-tests of

197

each drug condition versus DMSO controls. Differences in replication (area-under-the-curve; AUC)

198

in the HIV spreading assay were evaluated by a one-way repeated measures ANOVA. Comparisons

199

between modified viral outgrowth conditions were made using a two-way ANOVA where the

200

sources of variation were either the latency reactivation treatment or the number of ACH2 cells per

201

well.

10

202

RESULTS

203

Limited HDACi-induced Cell Death in Primary Human Cells

204

Our research focus was to determine the extent of romidepsin’s impact on de novo HIV

205

infection. As a control for the generalizability of our observations with romidepsin to other HDACi,

206

we also evaluated the impact of panobinostat on de novo HIV infection in selected assays. To

207

design the best strategies for evaluating the impact of HDACi on de novo viral infection, we first

208

determined the toxicity of each drug in: (i) HIV reporter TZM-bl cells; (ii) primary human PBMCs;

209

and (iii) primary CD4+ T cells. The dose range examined included physiologically relevant doses

210

of each drug [i.e. 40nM romidepsin (14) and 30nM panobinostat (15, 29)]. Furthermore, we

211

designed these analyses to detect potentially delayed toxic effects of drug exposure by measuring

212

viability at both 24 and 48 hours following removal of the drug (Figure 1A-left and middle).

213

Romidepsin and panobinostat treatment of TZM-bl cells resulted in only a minor impact on cell

214

viability at 24 hours, with the exception of the very high doses of romidepsin. In contrast, cell

215

viability at 48 hours was dramatically affected by romidepsin treatment, where cell death exceeded

216

90% at 100nM and above (Figure 1A-middle). Cell death was less pronounced for panobinostat

217

even at high doses (Figure 1A-left and middle). In these experiments, TZM-bl cells were exposed to

218

the HDACi for 8 hours. Given that key ex vivo analyses have incorporated drug exposures with

219

these HDACi of 18 hours (34); we also evaluated 18 hours of drug exposure on TZM-bl cells and

220

observed a notably greater reduction in cell viability (Figure 1A-right). As little as 7nM romidepsin

221

resulted in more than 80% of TZM-bl cells dying within 24 hours. We observed ~50% cell death at

222

30nM panobinostat and 80% cell death at 125nM. Next, to explore whether HDACi toxicity also

223

occurred in primary human cells, we determined the viability of both PBMCs and CD4+ T cells

224

exposed to romidepsin or panobinostat for a total of 8 hours. We found that romidepsin-induced cell

225

death in primary cells was less pronounced versus TZM-bl cells. Specifically, approximately 70-

11

226

80% of PBMCs and CD4+ T cells remained viable for 48 hours following exposure to the drug

227

concentrations used in our subsequent virus infection assays (Figure 1B and C).

228 229 230

Romidepsin Inhibits VSV-g Pseudotyped HIV Infection of CD4+ T Cells To understand the impact of HDACi on de novo viral infection, we performed a series of

231

primary human cell experiments to measure de novo infection following HDACi treatment. In the

232

first line of experiments, IL-2/PHA activated PBMCs were primed with increasing amounts of

233

either romidepsin or panobinostat for 2 hours prior to incubation with VSVg-pseudotyped single-

234

round HIV virions expressing eGFP (Figure 2A). The live cells in each culture were examined

235

using flow cytometry to quantitate the proportion of eGFP positive cells as a measure of infectivity

236

(Figure 2B). We found that priming cells with panobinostat did not alter the proportion of infected

237

PBMCs (Figure 2B&C); however, romidepsin significantly reduced infectivity of VSVg-

238

pseudotyped HIV at a relatively low concentration (10nM, P ≤ 0.01; 95% CI of difference 1.874 to

239

8.240) as well as at the more physiologically relevant concentration (40nM, P ≤ 0.001; 95% CI of

240

difference 2.845 to 9.211) (Figure 2B&C). Collectively, our experiments with activated PBMCs

241

showed a substantial reduction in infectivity following exposure to HDACi although this

242

experimental approach did not permit cell lineage specific data interpretation.

243 244

To examine whether the observed inhibition of infectivity by romidepsin specifically affected

245

CD4+ T cells and/or macrophages, we sorted CD4+ T cell populations from activated PBMCs prior

246

to HDACi priming and generated MDMs. Similar to the results described above, significantly fewer

247

primary CD4+ T cells were infected following priming with 10nM or 40nM romidepsin (P ≤ 0.01;

248

95% CI of difference 3.017 to 13.32 and P ≤0.0001; 95% CI of difference 5.910 to 16.22;

249

respectively) (Figure 2C). In contrast, MDMs were generally permissive for infection by the VSVg-

12

250

pseudotyped single-round HIV virions regardless of HDACi treatment (Supplemental Figure 1).

251

Specifically, we observed more than 20% infection in DMSO, romidepsin and panobinostat treated

252

MDMs derived from one of the donor whose CD4+ T cells exhibited profoundly reduced infection

253

following exposure to romidepsin. Together, these data indicate that de novo infection of activated

254

CD4+ T cells by VSVg-pseudotyped HIV was inhibited by romidepsin.

255 256 257

Romidepsin Inhibits Wild-type HIV Infection of CD4+ T Cells Next we examined whether the mechanism of inhibition was specific to the pH-dependent

258

fusion that characterizes VSVg-pseudotyped virions or if wild-type HIV envelope fusion was

259

similarly affected by treatment. To do this, HDACi-primed activated CD4+ T cells were challenged

260

with replication competent HIV-1HXB2, a highly pathogenic, CXCR4-tropic, laboratory-adapted

261

viral isolate (30) (Figure 3A). CD4+ T cell cultures primed with 10nM romidepsin exhibited

262

significantly reduced infection after 2 days in culture as measured by Gagp24 in supernatants (P ≤

263

0.05) (Figure 3B&C). Similar effects were observed for romidepsin 40nM (P ≤0.05), but not for

264

30nM panobinostat. By Day 5, reduced HIV replication was only observed in the cultures primed

265

with 40nM romidepsin (P ≤0.001). This pronounced effect was maintained through 7 days of the

266

culture, despite the fact that the drug pulse was relatively short in duration (Figure 3B&C). Thus

267

exposure of activated CD4+ T cells to physiologically relevant doses of romidepsin significantly

268

decreased viral spreading and thus the effect of romidepsin on de novo HIV infection is not

269

dependent on the viral route of entry.

270 271

Romidepsin Selectively Activates Interferon-Stimulated Genes in PBMCs

272

To gain further insights into a potential mechanism for the HIV inhibition exhibited by

273

romidepsin, we investigated the gene profile in PBMCs challenged with either romidepsin or

13

274

panobinostat. We found that nearly all tested innate immune sensors, as well as other genes

275

examined, were affected by 2 or 8 hours of panobinostat exposure (e.g. downregulation of MX1,

276

Viperin, and APOBEC3G, NFkB and IRF7) (Figure 4). In contrast, whereas the expression of most

277

genes was unchanged following 2 hours of exposure to romidepsin, 8 hours of 40nM romidepsin

278

exposure led to major alterations in the gene profile (Figure 4-right). Expression was downregulated

279

for most of the examined genes; however, there were three notable exceptions: IFIT1, ISG15 and

280

STAT1. Expression of each of these genes was increased following 8 hours of exposure to 40nM

281

romidepsin (Figure 4-right), indicative of differential regulation of innate restriction factors

282

important for viral confinement.

283 284

Romidepsin Inhibits Proliferation of Resting CD4+ T Cells and Reduces the Sensitivity of

285

Viral Outgrowth Assays

286

Given our observations that romidepsin reduced de novo HIV infection and virus spread in

287

culture, we investigated the impact of this drug specifically on resting CD4+ T cells. We chose

288

these cells because they are the primary cell type included in the viral outgrowth assays used to

289

quantitate the size of the latent reservoir in HIV patients (20, 31, 34-39). The impact of romidepsin

290

on the proliferation of magnetically-enriched, HIV-negative blood donor derived resting CD4+ T

291

cells was profound. We first confirmed a previously reported observation (40) that exposure to

292

romidepsin interferes with T cell proliferation when combined with mitogenic stimulation

293

(Supplemental Figure 2). However, it was essential that we also evaluate the impact of romidepsin

294

(18 hour drug exposure) in the absence of mitogenic stimulation to more closely approximate the

295

conditions utilized in viral outgrowth assays (34) (Figure 5A). We found that both proliferation

296

(Figure 5B-C) and cell viability (Figure 5D) of resting CD4+ T cells were dramatically reduced by

297

18 hours of exposure to romidepsin.

14

298 299

Latent HIV infection in resting CD4+ resting T cells is infrequent (~1 infectious unit per

300

million resting CD4+ T cells tested [IUPM]). Viral outgrowth assays are extended duration (~14

301

days), stimulatory cultures of resting CD4+ T cells that are optimized for the quantitation of the

302

latent HIV reservoir (20, 31, 34-39). To test our hypothesis that romidepsin’s broad impact on cell

303

viability, cell proliferation and de novo HIV infection could alter the sensitivity of viral outgrowth

304

assays and to experimentally overcome the limitation posed by the fact that latency events in cART-

305

suppressed HIV patients are rare, we established a modified viral outgrowth assay with 1x106

306

resting CD4+ T cells per culture from HIV-negative blood donors spiked with a defined number of

307

ACH2 cells. ACH2 cells are a model for HIV latency (12, 32, 33), where cells can be stimulated to

308

produce HIV progeny virions by romidepsin exposure [as with latently infected resting CD4+ T

309

cells (14)], but not the mitogenic stimulator phytohemagglutinin (PHA) [unlike latently infected

310

resting CD4+ T cells (Figure 5E)]. Three different treatment conditions were evaluated in this

311

modified viral outgrowth system: media only, 0.5 µg PHA and 40nM romidepsin (Figure 5F).

312

When we tested the typical patient condition of 1 IUPM, we observed viral outgrowth in only 17%

313

(1 of 6) of PHA-treated wells tested and 0% in the media only (0 of 6) or romidepsin wells (0 of 8).

314

When a 10-fold higher IUPM was stimulated, we observed 0% viral outgrowth in the romidepsin

315

wells (0 of 8) whereas 33% of media only wells (2 of 6) and 50% of PHA-stimulated cultures (3 of

316

6) exhibited viral outgrowth. When 100 IUPM was evaluated only 50% of romidepsin cultures (4 of

317

8) versus 83% of media only wells (5 of 6) and 100% percent of PHA wells (6 of 6) were positive

318

for viral outgrowth. Even at 1000 IUPM, not all wells treated with romidepsin (5 of 6) were positive

319

for viral outgrowth as was the case for both the media control (6 of 6) and the PHA (6 of 6) treated

320

wells. We conclude from these data that romidepsin reduced the sensitivity of the viral outgrowth

15

321

assay and that romidepsin’s induction effects are obscured within this assay at the typical patient

322

condition of 1 IUPM.

16

323

DISCUSSION

324

The rationale for incorporation of HDACi into HIV eradication strategies is that epigenetic

325

modulation of molecular mechanisms blocking transcription of integrated HIV DNA can reactivate

326

HIV expression in resting infected memory CD4+ T cells and disrupt latency (29). Numerous in

327

vitro, ex vivo and clinical studies have shown that HDACi reactivate latent HIV infection (9, 12-16,

328

37, 38, 40-48). However, a theoretical concern is that HDACi-induced viral production in cART

329

sanctuary sites could result in de novo HIV infections and reseeding of the latent reservoir (17, 18).

330

In the current study, we addressed this concern by comprehensively exploring the impact of

331

romidepsin on HIV infection. We found that this effective LRA profoundly restricts de novo HIV

332

infections of primary human T cells.

333 334

After demonstrating that the physiologically relevant dosages of the drugs that we used were not

335

overtly cytotoxic (Figure 1), we challenged primary human cells with romidepsin and then infected

336

the cells with either VSV-g pseudotyped HIV or highly pathogenic wild-type HIV, which in both

337

cases were virus models with robust infectivity rates (Figures 2&3). To our surprise, we observed

338

that de novo infection of activated PBMCs and activated CD4+ T cells by VSVg-pseudotyped HIV

339

was inhibited by physiological drug levels of romidepsin. In contrast, when we performed a similar

340

experiment with donor-matched MDM, we observed no appreciable HDACi effect on infection by

341

these VSVg-pseudotyped HIV virions which agrees with recent findings presented by Campbell, et

342

al. (18). When activated CD4+ T cells were treated with romidepsin and then infected with wild-

343

type HIV, we observed a significant reduction in viral spread within the cell culture. Taken

344

together, these two observations reveal that the mechanism of inhibiting de novo HIV was not

345

envelope-dependent. However, further investigations are needed to dissect the differences in

346

infectivity observed between romidepsin-treated CD4+T cells and MDMs.

17

347 348

Next we used Fluidigm gene expression analyses to begin to elucidate whether the mechanism of

349

inhibiting de novo HIV is dependent upon an HDACi-induced alteration in the innate antiviral

350

activities of the target cells. To do this we utilized a multiplex gene array (49) designed to

351

investigate mRNA levels for: (i) molecules involved in innate immune sensing pathways; (ii)

352

interferon stimulated genes (ISG); (iii) innate immunity signal transduction mediators; and (iv)

353

cytokines related to innate immune responses. We noted that after 8 hours of exposure to 40nM

354

romidepsin expression of IFIT1, ISG15 and STAT1 in PBMCs were significantly upregulated

355

(Figure 4-right). This is important, in short, because of known antiviral effects for each of these

356

proteins. Specifically, the members of the family of IFITs, including IFIT1, are known to be

357

upregulated after IFN treatment, viral infections and pathogen-associated molecular pattern

358

recognition (50-52). ISG15 is an ubiquitin-like protein shown to protect cells in numerous viral

359

infection models, but for HIV it is believed mainly to impair viral release (53, 54). STAT1

360

activation can often be correlated with cellular proinflammatory, anti-proliferative, and apoptotic

361

activities (55). While this analysis was focused on a subset of genes involved in innate immunity

362

and did not address other potential mechanisms of virus inhibition, the observation that romidepsin-

363

induced increases in expression for these genes suggests that HDACi inhibition of de novo HIV

364

infections could be mediated through HDACi-altered innate immunity versus direct interference

365

with virus replication.

366 367

In selected assays, we included panobinostat as a control to assess the generalizability of our

368

romidepsin-related findings to other HDACi. Romidepsin and panobinostat target class I and class

369

I/II HDAC, respectively (13). These two HDACi had similar cell viability profiles in activated,

370

primary PBMCs and CD4+ T cell cultures (Figure 1). When we examined the impact of these 2

18

371

HDACi on infection with VSV-g pseudotyped HIV and wild-type HIV, we observed a dose

372

dependent effect with romidepsin where the most profound inhibition of de novo infection was

373

observed at the highest tested dose (40nM). The dose of panobinostat (30nM) evaluated herein was

374

matched to the concentration of panobinostat that was attained in vivo with the dosing strategy

375

employed in a recent clinical trial (9, 15). At this dose (and also at the supra-physiological dose of

376

>100nM; data not shown), panobinostat did not exhibit the same level of inhibition of de novo HIV

377

infection that we observed for romidepsin (Figures 2&3).

378 379

The cells that compose the latent HIV reservoir are relatively infrequent (20, 31, 34-39). Because

380

positive signals detected by viral outgrowth assays are rare, we hypothesized that romidepsin’s

381

broad impacts on cell viability, cell proliferation and de novo HIV infection could alter the

382

sensitivity of this assay. Such an outcome could lead to under-estimation of efficacy when this

383

assay is utilized ex vivo to evaluate the function of potential LRA like romidepsin. Our modified

384

viral outgrowth assay was designed with defined IUPM values to allow us to test this hypothesis.

385

Two relevant comparisons were incorporated into the experiment. The “PHA versus media only”

386

comparison was included to evaluate whether resting CD4+ T cells that were activated by PHA

387

would in turn activate ACH2 cell HIV production while the “media only versus romidepsin”

388

comparison permitted us to discern whether romidepsin reduced the ability of this assay to detect

389

viral outgrowth from latently infected cells or not. Our results showed that ex vivo romidepsin

390

exposure profoundly impacted the ability of the viral outgrowth assay to quantitate the latent HIV

391

reservoir at the typical patient condition of 1 IUPM. Thus, these data indicate that caution should be

392

exercised when determining whether a given molecule is capable of reactivating latent HIV

393

infection from HIV patient-derived resting CD4+ T cells following ex vivo stimulation of with

394

potential LRA. We further demonstrate that the inhibition of HIV spreading in cell culture that

19

395

results from romidepsin treatment creates a technical barrier for the use of virus spreading assays,

396

such as the viral outgrowth assay, for assessing the potential of romidepsin to reverse HIV latency.

397 398

In conclusion, we show that romidepsin inhibits HIV infection in an envelope-independent manner.

399

Furthermore, we provide evidence that this inhibition could result from a romidepsin-mediated

400

enhancement in antiviral innate immunity. Altogether our data indicate that reseeding of the latent

401

reservoir is an unlikely outcome of a romidepsin-based HIV eradication strategy. Translation of our

402

ex vivo findings to the clinic, may best be facilitated through the use of an animal model of HIV

403

persistence where ART sanctuaries may exist (1). The in vivo testing of adjunct therapy with

404

romidepsin as an HIV eradication strategy in such a model is warranted to fully characterize the

405

likelihood of adjunct romidepsin treatment to enhance the risk of HIV infection and reservoir

406

reseeding during a kick and kill HIV eradication therapy.

20

407

ACKNOWLEDGEMENTS

408

This work was supported in part by: The Lundbeck foundation (KJ and MRJ), Aarhus University

409

Research Foundation (MRJ), The Danish Strategic Research Council Grant 0603-00521B (LØ) and

410

Danish Research Council Grant 12-133887 (OSS). The funders had no role in study design, data

411

collection, and analysis, decision to publish, or writing of the manuscript. The following reagents

412

were obtained through the NIH AIDS Reagent Program, Division of AIDS, NIAID, NIH: TZM-bl

413

cells (from Dr. John C. Kappes, Dr. Xiaoyun Wu and Tranzyme Inc.); ACH-2 cells (from Dr.

414

Thomas Folks); MOLT-4/CCR5 cells (from Dr. Masanori Baba, Dr. Hiroshi Miyake, Dr. Yuji

415

Iizawa).

416 417

We thank the following for their technical assistance: Lene Svinth Jøhnke; Ane Kjeldsen; and the

418

Flow Cytometry core facilities at the Aarhus University Institute of Biomedicine.

21

419

REFERENCES

420

1.

421

422

Eisele E, Siliciano RF. 2012. Redefining the viral reservoirs that prevent HIV-1 eradication. Immunity 37:377-388.

2.

423

Richman DD, Margolis DM, Delaney M, Greene WC, Hazuda D, Pomerantz RJ. 2009. The challenge of finding a cure for HIV infection. Science 323:1304-1307.

424

3.

Deeks SG. 2012. HIV: Shock and kill. Nature 487:439-440.

425

4.

Deeks SG, Autran B, Berkhout B, Benkirane M, Cairns S, Chomont N, Chun TW, Churchill M,

426

Mascio MD, Katlama C, Lafeuillade A, Landay A, Lederman M, Lewin SR, Maldarelli F,

427

Margolis D, Markowitz M, Martinez-Picado J, Mullins JI, Mellors J, Moreno S, O'Doherty U,

428

Palmer S, Penicaud MC, Peterlin M, Poli G, Routy JP, Rouzioux C, Silvestri G, Stevenson M,

429

Telenti A, Lint CV, Verdin E, Woolfrey A, Zaia J, Barre-Sinoussi F. 2012. Towards an HIV cure: a

430

global scientific strategy. Nat Rev Immunol 12:607-614.

431

5.

432

433

Immunol 33:554-562.

6.

434

435

438

Dahl V, Josefsson L, Palmer S. 2010. HIV reservoirs, latency, and reactivation: prospects for eradication. Antiviral Res 85:286-294.

7.

436

437

Durand CM, Blankson JN, Siliciano RF. 2012. Developing strategies for HIV-1 eradication. Trends

Archin NM, Sung JM, Garrido C, Soriano-Sarabia N, Margolis DM. 2014. Eradicating HIV-1 infection: seeking to clear a persistent pathogen. Nat Rev Microbiol 12:750-764.

8.

Colin L, Van Lint C. 2009. Molecular control of HIV-1 postintegration latency: implications for the development of new therapeutic strategies. Retrovirology 6:111.

22

439

9.

Rasmussen TA, Schmeltz Sogaard O, Brinkmann C, Wightman F, Lewin SR, Melchjorsen J,

440

Dinarello C, Ostergaard L, Tolstrup M. 2013. Comparison of HDAC inhibitors in clinical

441

development: Effect on HIV production in latently infected cells and T-cell activation. Human

442

vaccines & immunotherapeutics 9:993-1001.

443

10.

Keedy KS, Archin NM, Gates AT, Espeseth A, Hazuda DJ, Margolis DM. 2009. A limited group of

444

class I histone deacetylases acts to repress human immunodeficiency virus type 1 expression. J

445

Virol 83:4749-4756.

446

11.

Archin NM, Keedy KS, Espeseth A, Dang H, Hazuda DJ, Margolis DM. 2009. Expression of

447

latent human immunodeficiency type 1 is induced by novel and selective histone deacetylase

448

inhibitors. AIDS 23:1799-1806.

449

12.

Wightman F, Lu HK, Solomon AE, Saleh S, Harman AN, Cunningham AL, Gray L, Churchill M,

450

Cameron PU, Dear AE, Lewin SR. 2013. Entinostat is a histone deacetylase inhibitor selective for

451

class 1 histone deacetylases and activates HIV production from latently infected primary T cells.

452

AIDS 27:2853-2862.

453

13.

Huber K, Doyon G, Plaks J, Fyne E, Mellors JW, Sluis-Cremer N. 2011. Inhibitors of histone

454

deacetylases: correlation between isoform specificity and reactivation of HIV type 1 (HIV-1) from

455

latently infected cells. J Biol Chem 286:22211-22218.

456

14.

Wei DG, Chiang V, Fyne E, Balakrishnan M, Barnes T, Graupe M, Hesselgesser J, Irrinki A,

457

Murry JP, Stepan G, Stray KM, Tsai A, Yu H, Spindler J, Kearney M, Spina CA, McMahon D,

458

Lalezari J, Sloan D, Mellors J, Geleziunas R, Cihlar T. 2014. Histone deacetylase inhibitor

459

romidepsin induces HIV expression in CD4 T cells from patients on suppressive antiretroviral

460

therapy at concentrations achieved by clinical dosing. PLoS Pathog 10:e1004071.

23

461

15.

Rasmussen TA, Tolstrup M, Brinkmann CR, Olesen R, Erikstrup C, Solomon A, Winckelmann

462

A, Palmer S, Dinarello C, Buzon M, Lichterfeld M, Lewin SR, Østergaard L, Søgaard OS. 2014.

463

Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on

464

suppressive antiretroviral therapy: a phase 1/2, single group, clinical trial. The Lancet HIV 1:e13-

465

e21.

466

16.

Søgaard O, Graversen M, Leth S, Brinkmann C, Kjær A-S, Olesen R, Denton P, Nissen S,

467

Sommerfelt M, Rasmussen T, Østergaard L, Tolstrup M. 2014. The HDAC inhibitor romidepsin is

468

safe and effectively reverses HIV-1 latency in vivo as measured by standard clinical assays, 20th

469

International AIDS Conference (AIDS 2014), Melbourne, Australia.

470

17.

Lucera MB, Tilton CA, Mao H, Dobrowolski C, Tabler CO, Haqqani AA, Karn J, Tilton JC. 2014.

471

The histone deacetylase inhibitor vorinostat (SAHA) increases the susceptibility of uninfected CD4+

472

T cells to HIV by increasing the kinetics and efficiency of postentry viral events. J Virol 88:10803-

473

10812.

474

18.

475

476

Campbell GR, Bruckman RS, Chu YL, Spector SA. 2015. Autophagy Induction by Histone Deacetylase Inhibitors Inhibits HIV Type 1. J Biol Chem 290:5028-5040.

19.

Fletcher CV, Staskus K, Wietgrefe SW, Rothenberger M, Reilly C, Chipman JG, Beilman GJ,

477

Khoruts A, Thorkelson A, Schmidt TE, Anderson J, Perkey K, Stevenson M, Perelson AS,

478

Douek DC, Haase AT, Schacker TW. 2014. Persistent HIV-1 replication is associated with lower

479

antiretroviral drug concentrations in lymphatic tissues. Proc Natl Acad Sci U S A 111:2307-2312.

480

20.

Laird GM, Eisele EE, Rabi SA, Lai J, Chioma S, Blankson JN, Siliciano JD, Siliciano RF. 2013.

481

Rapid quantification of the latent reservoir for HIV-1 using a viral outgrowth assay. PLoS Pathog

482

9:e1003398.

24

483

21.

Platt EJ, Bilska M, Kozak SL, Kabat D, Montefiori DC. 2009. Evidence that ecotropic murine

484

leukemia virus contamination in TZM-bl cells does not affect the outcome of neutralizing antibody

485

assays with human immunodeficiency virus type 1. J Virol 83:8289-8292.

486

22.

487

488

Takeuchi Y, McClure MO, Pizzato M. 2008. Identification of gammaretroviruses constitutively released from cell lines used for human immunodeficiency virus research. J Virol 82:12585-12588.

23.

Wei X, Decker JM, Liu H, Zhang Z, Arani RB, Kilby JM, Saag MS, Wu X, Shaw GM, Kappes JC.

489

2002. Emergence of resistant human immunodeficiency virus type 1 in patients receiving fusion

490

inhibitor (T-20) monotherapy. Antimicrob Agents Chemother 46:1896-1905.

491

24.

Derdeyn CA, Decker JM, Sfakianos JN, Wu X, O'Brien WA, Ratner L, Kappes JC, Shaw GM,

492

Hunter E. 2000. Sensitivity of human immunodeficiency virus type 1 to the fusion inhibitor T-20 is

493

modulated by coreceptor specificity defined by the V3 loop of gp120. J Virol 74:8358-8367.

494

25.

Platt EJ, Wehrly K, Kuhmann SE, Chesebro B, Kabat D. 1998. Effects of CCR5 and CD4 cell

495

surface concentrations on infections by macrophagetropic isolates of human immunodeficiency virus

496

type 1. J Virol 72:2855-2864.

497

26.

498

499

summary. National Center for Biotechnology Information. [Online.]

27.

500

501 502

2015, posting date. PubChem Compound Database; CID=6918837; panobinostat compound

2015, posting date. PubChem Compound Database; CID=5352062; romidepsin compound summary. National Center for Biotechnology Information. [Online.]

28.

Atadja P. 2009. Development of the pan-DAC inhibitor panobinostat (LBH589): successes and challenges. Cancer letters 280:233-241.

25

503

29.

Rasmussen TA, Tolstrup M, Winckelmann A, Ostergaard L, Sogaard OS. 2013. Eliminating the

504

latent HIV reservoir by reactivation strategies: advancing to clinical trials. Human vaccines &

505

immunotherapeutics 9:790-799.

506

30.

Ratner L, Fisher A, Jagodzinski LL, Mitsuya H, Liou RS, Gallo RC, Wong-Staal F. 1987.

507

Complete nucleotide sequences of functional clones of the AIDS virus. AIDS Res Hum Retroviruses

508

3:57-69.

509

31.

Siliciano JD, Siliciano RF. 2005. Enhanced culture assay for detection and quantitation of latently

510

infected, resting CD4+ T-cells carrying replication-competent virus in HIV-1-infected individuals.

511

Methods Mol Biol 304:3-15.

512

32.

Folks TM, Clouse KA, Justement J, Rabson A, Duh E, Kehrl JH, Fauci AS. 1989. Tumor

513

necrosis factor alpha induces expression of human immunodeficiency virus in a chronically infected

514

T-cell clone. Proc Natl Acad Sci U S A 86:2365-2368.

515

33.

Clouse KA, Powell D, Washington I, Poli G, Strebel K, Farrar W, Barstad P, Kovacs J, Fauci

516

AS, Folks TM. 1989. Monokine regulation of human immunodeficiency virus-1 expression in a

517

chronically infected human T cell clone. J Immunol 142:431-438.

518

34.

Bullen CK, Laird GM, Durand CM, Siliciano JD, Siliciano RF. 2014. New ex vivo approaches

519

distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat Med

520

20:425-429.

521

35.

Chun TW, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, Hermankova M, Chadwick K,

522

Margolick J, Quinn TC, Kuo YH, Brookmeyer R, Zeiger MA, Barditch-Crovo P, Siliciano RF.

523

1997. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature

524

387:183-188.

26

525

36.

Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, Quinn TC, Chadwick K,

526

Margolick J, Brookmeyer R, Gallant J, Markowitz M, Ho DD, Richman DD, Siliciano RF. 1997.

527

Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science

528

278:1295-1300.

529

37.

Archin NM, Eron JJ, Palmer S, Hartmann-Duff A, Martinson JA, Wiegand A, Bandarenko N,

530

Schmitz JL, Bosch RJ, Landay AL, Coffin JM, Margolis DM. 2008. Valproic acid without

531

intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells.

532

AIDS 22:1131-1135.

533

38.

Lehrman G, Hogue IB, Palmer S, Jennings C, Spina CA, Wiegand A, Landay AL, Coombs RW,

534

Richman DD, Mellors JW, Coffin JM, Bosch RJ, Margolis DM. 2005. Depletion of latent HIV-1

535

infection in vivo: a proof-of-concept study. Lancet 366:549-555.

536

39.

Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DI, Lai J, Blankson JN,

537

Siliciano JD, Siliciano RF. 2013. Replication-competent noninduced proviruses in the latent

538

reservoir increase barrier to HIV-1 cure. Cell 155:540-551.

539

40.

Jones RB, O'Connor R, Mueller S, Foley M, Szeto GL, Karel D, Lichterfeld M, Kovacs C,

540

Ostrowski MA, Trocha A, Irvine DJ, Walker BD. 2014. Histone deacetylase inhibitors impair the

541

elimination of HIV-infected cells by cytotoxic T-lymphocytes. PLoS Pathog 10:e1004287.

542

41.

Elliott JH, Wightman F, Solomon A, Ghneim K, Ahlers J, Cameron MJ, Smith MZ, Spelman T,

543

McMahon J, Velayudham P, Brown G, Roney J, Watson J, Prince MH, Hoy JF, Chomont N,

544

Fromentin R, Procopio FA, Zeidan J, Palmer S, Odevall L, Johnstone RW, Martin BP, Sinclair

545

E, Deeks SG, Hazuda DJ, Cameron PU, Sekaly RP, Lewin SR. 2014. Activation of HIV

546

Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral

547

Therapy. PLoS Pathog 10:e1004473.

27

548

42.

549

550

compounds to disrupt HIV latency. Cytokine Growth Factor Rev 23:159-172.

43.

551

552

Remoli AL, Marsili G, Battistini A, Sgarbanti M. 2012. The development of immune-modulating

Rigby L, Muscat A, Ashley D, Algar E. 2012. Methods for the analysis of histone H3 and H4 acetylation in blood. Epigenetics : official journal of the DNA Methylation Society 7:875-882.

44.

Savarino A, Mai A, Norelli S, El Daker S, Valente S, Rotili D, Altucci L, Palamara AT, Garaci E.

553

2009. "Shock and kill" effects of class I-selective histone deacetylase inhibitors in combination with

554

the glutathione synthesis inhibitor buthionine sulfoximine in cell line models for HIV-1 quiescence.

555

Retrovirology 6:52.

556

45.

Archin NM, Espeseth A, Parker D, Cheema M, Hazuda D, Margolis DM. 2009. Expression of

557

latent HIV induced by the potent HDAC inhibitor suberoylanilide hydroxamic acid. AIDS Res Hum

558

Retroviruses 25:207-212.

559

46.

Archin NM, Liberty AL, Kashuba AD, Choudhary SK, Kuruc JD, Crooks AM, Parker DC,

560

Anderson EM, Kearney MF, Strain MC, Richman DD, Hudgens MG, Bosch RJ, Coffin JM, Eron

561

JJ, Hazuda DJ, Margolis DM. 2012. Administration of vorinostat disrupts HIV-1 latency in patients

562

on antiretroviral therapy. Nature 487:482-485.

563

47.

564

565

Human Immunodeficiency Virus Type 1 Latency in CD4(+) T Cells. J Infect Dis 197:1162-1170.

48.

566

567

Choudhary SK, Archin NM, Margolis DM. 2008. Hexamethylbisacetamide and Disruption of

Ylisastigui L, Archin NM, Lehrman G, Bosch RJ, Margolis DM. 2004. Coaxing HIV-1 from resting CD4 T cells: histone deacetylase inhibition allows latent viral expression. AIDS 18:1101-1108.

49.

Jang JS, Simon VA, Feddersen RM, Rakhshan F, Schultz DA, Zschunke MA, Lingle WL,

568

Kolbert CP, Jen J. 2011. Quantitative miRNA expression analysis using fluidigm microfluidics

569

dynamic arrays. BMC genomics 12:144.

28

570

50.

571

572

Sarkar SN, Sen GC. 2004. Novel functions of proteins encoded by viral stress-inducible genes. Pharmacology & therapeutics 103:245-259.

51.

Jager S, Cimermancic P, Gulbahce N, Johnson JR, McGovern KE, Clarke SC, Shales M,

573

Mercenne G, Pache L, Li K, Hernandez H, Jang GM, Roth SL, Akiva E, Marlett J, Stephens M,

574

D'Orso I, Fernandes J, Fahey M, Mahon C, O'Donoghue AJ, Todorovic A, Morris JH, Maltby

575

DA, Alber T, Cagney G, Bushman FD, Young JA, Chanda SK, Sundquist WI, Kortemme T,

576

Hernandez RD, Craik CS, Burlingame A, Sali A, Frankel AD, Krogan NJ. 2012. Global landscape

577

of HIV-human protein complexes. Nature 481:365-370.

578

52.

579

580

eIF3f. Proc Natl Acad Sci U S A 106:4071-4078.

53.

581

582

585

Okumura A, Lu G, Pitha-Rowe I, Pitha PM. 2006. Innate antiviral response targets HIV-1 release by the induction of ubiquitin-like protein ISG15. Proc Natl Acad Sci U S A 103:1440-1445.

54.

583

584

Valente ST, Gilmartin GM, Mott C, Falkard B, Goff SP. 2009. Inhibition of HIV-1 replication by

Morales DJ, Lenschow DJ. 2013. The antiviral activities of ISG15. Journal of molecular biology 425:4995-5008.

55.

Decker T. 1999. Introduction: STATs as essential intracellular mediators of cytokine responses. Cellular and molecular life sciences : CMLS 55:1505-1508.

586 587 588 589

29

590 591

FIGURE LEGENDS

592

Figure 1. Differential impact of HDACi in immortalized versus primary human cells. (A-C)

593

The viability of TZM-bl cells (A) and primary human cells (B&C) was assessed by flow cytometry

594

after the cells were pulsed with either romidepsin or panobinostat for 8 or 18 hours and then

595

incubated for an additional 24 or 48 hours, as indicated in each sub-panel. Panel (A) presents

596

compiled results from (two) experiments performed in triplicate. Panels (B) and (C) present

597

compiled data for PBMCs or CD4+ T cells, triplicate measures respectively from 2 human donors.

598

Error bars: SD. Gray squares: romidepsin. Black circles: panobinostat.

599 600

Figure 2. Single round HIV infection of primary human cells with VSV-g pseudotyped virus

601

was inhibited by romidepsin. (A) Schematic representation of the experimental approach. (B)

602

Representative flow cytometry data highlight the reduced infection that results from romidepsin

603

treatment. Percentages of infected, eGFP positive cells are presented with each dot plot. (C)

604

Compiled data for PBMCs and for CD4+ T cells from 4 human donors are shown (mean +/- SEM).

605

**p

Histone deacetylase inhibitor romidepsin inhibits de novo HIV-1 infections.

Adjunct therapy with the histone deacetylase inhibitor (HDACi) romidepsin increases plasma viremia in HIV patients on combination antiretroviral thera...
3MB Sizes 2 Downloads 8 Views