Journal of Antimicrobial Chemotherapy Advance Access published February 23, 2015

J Antimicrob Chemother doi:10.1093/jac/dkv045

Small molecule inhibitors of the annexin A2 heterotetramer prevent human papillomavirus type 16 infection Andrew W. Woodham1†, Julia R. Taylor1†, Andrew I. Jimenez1, Joseph G. Skeate2, Thomas Schmidt3, Heike E. Brand2, Diane M. Da Silva2,4 and W. Martin Kast1,2,4* 1

Department of Molecular Microbiology & Immunology, University of Southern California, 2011 Zonal Avenue HMR 401, Los Angeles, CA, USA; 2Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA, USA; 3Laboratories of Chemical Physics and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA; 4Department of Obstetrics & Gynecology, University of Southern California, 2020 Zonal Avenue Room 220, Los Angeles, CA, USA *Corresponding author. Tel: +1-323-442-3870; Fax: +1-323-442-7760; E-mail: [email protected] †These authors contributed equally to this work.

Received 18 December 2014; returned 27 January 2015; revised 30 January 2015; accepted 3 February 2015 Objectives: High-risk human papillomavirus (HPV) infection leads to the development of several human cancers that cause significant morbidity and mortality worldwide. HPV type 16 (HPV16) is the most common of the cancer-causing genotypes and gains entry to the basal cells of the epithelium through a non-canonical endocytic pathway that involves the annexin A2/S100A10 heterotetramer (A2t). A2t is composed of two annexin A2 monomers bound to an S100A10 dimer and this interaction is a potential target to block HPV16 infection. Here, recently identified small molecule inhibitors of A2t (A2ti) were investigated for their ability to prevent HPV16 infection in vitro. Methods: A2ti were added to HeLa cells in increasing concentrations prior to the addition of HPV16. Cytotoxicity was evaluated via trypan blue exclusion. HPV16 pseudovirion infection and fluorescently labelled HPV16 capsid internalization was measured with flow cytometry. Results: A2ti blocked HPV16 infection by 100% without substantial cellular toxicity or reduction in cell growth. Furthermore, A2ti blocked HPV16 entry into epithelial cells by 65%, indicating that the observed inhibition of HPV16 infection is in part due to a block in entry and that non-infectious entry may occur in the absence of A2t binding. Conclusions: These results demonstrate that targeting A2t may be an effective strategy to prevent HPV16 infection. Keywords: HPV16, annexin A2/S100A10 heterotetramer, A2t

Introduction High-risk human papillomavirus (hr-HPV) infection leads to the development of several infection-related cancers including cervical, anogenital and head and neck cancers that are a significant health burden worldwide. 1 – 4 HPV type 16 (HPV16), the most common of the hr-HPV genotypes, is an obligatory intracellular non-enveloped virus that must gain entry into host basal cells of the epithelium to deliver its double-stranded DNA to the nucleus and the HPV16 capsid proteins play a vital role in these steps. 5 We previously reported that the annexin A2/S100A10 heterotetramer (A2t) facilitates infectious entry of HPV16 into epithelial cells through a direct protein – protein interaction (PPI) between the S100A10 subunit of A2t and the HPV16

L2 minor capsid protein.6 PPIs are increasingly being explored for small molecule drug discovery and the identification of A2t as an HPV16 receptor makes it a promising target for inhibition. Annexin A2 (A2) is found cytoplasmically as a monomer or at the cell surface as a heterotetramer consisting of two A2 monomers bridged non-covalently to an S100A10 dimer.7 The dimeric S100A10 structure yields two binding pockets that accommodate the N-terminus of A28 and it is this interaction that preliminary drug discovery studies have targeted.9 Recently, inhibitors of A2t (A2ti) that specifically disrupt the PPI between A2 and S100A10 have been identified,10 but have not been explored in the context of HPV infection. Here, we investigated the ability of A2ti to inhibit HPV16 entry and infection of epithelial cells in vitro.

# The Author 2015. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy. All rights reserved. For Permissions, please e-mail: [email protected]

1 of 5

Woodham et al.

Materials and methods Cells, reagents and HPV16 pseudovirions (PsV) HeLa cells (ATCC, Manassas, VA, USA) were maintained in IMDM with 10% FBS and PenStrep (Lonza, Walkersville, MD, USA) at 378C with 5% CO2. Spontaneously transformed HaCaT keratinocytes (ATCC) were maintained in Keratinocyte Serum-Free Media (Life Technologies, Carlsbad, CA, USA) with manufacturer-provided growth supplement at 378C with 5% CO2. A2ti {A2ti-1: 2-[4-(2-ethylphenyl)-5-o-tolyloxymethyl-4H-[1,2,4]triazol3-ylsulfanyl]acetamide; and A2ti-2: 2-(4-phenyl-5-o-tolyloxymethyl-4H[1,2,4]triazol-3-ylsulfanyl)acetamide} were purchased from Asinex (Moscow, Russia) and reconstituted in DMSO. The ability of A2ti to disrupt A2t was verified via isothermal titration calorimetry following standard procedures.11,12 HPV16 PsV were produced by co-transfection of 293TT cells with plasmids encoding codon-optimized HPV16 L1 and L2 following published procedures.13

Toxicity assay HeLa cells were incubated at 378C in 5% CO2 with increasing concentrations of A2ti-1 or A2ti-2 for 72 h. Cells were then counted and viability was measured by trypan blue exclusion. In control experiments, cells were left untreated or were treated with DMSO at matched concentrations to A2ti delivery.

HPV16 PsV infection assay with A2ti HeLa or HaCaT cells seeded at 2×104 cells/well were incubated overnight in 24-well plates at 378C with increasing concentrations of A2ti-1 or A2ti-2. The following day, cells were incubated with HPV16 PsV containing a GFP reporter plasmid using an moi of 50. Using flow cytometry, infection was measured 48 h post-HPV16 PsV treatment as the percentage of GFP-positive cells. In control experiments, cells were left untreated or were treated with DMSO.

HPV16 PsV internalization assay with A2ti HPV16 PsV were labelled with a pH-dependent rhodamine fluorophore (pHrodo, Life Technologies) or carboxyfluorescein diacetate succinimidyl

(a)

ester (CFDA-SE, Life Technologies). HeLa cells seeded at 2×105 cells/well were incubated overnight with increasing concentrations of A2ti-1 or A2ti-2. The next day, pHrodo- or CFDA-SE-labelled HPV16 PsV were added at 1 mg/106 cells and incubated at 378C for 6 h. The mean fluorescence intensity (MFI) of the cells was analysed by flow cytometry. In control experiments, cells were left untreated or were treated with DMSO.

Results In the current study, we examined the ability of two A2ti to block HPV16 entry and infection in vitro. The first of these inhibitors (A2ti-1) has a reported IC50 of 24 mM10 and it was hypothesized that A2ti-1 would block HPV16 PsV infection within a reasonable range of this value. The second of the tested inhibitors (A2ti-2) is similar in chemical structure, but the deletion of an ethyl group results in an increased IC50 of 230 mM (Figure 1a) and was therefore predicted to be less effective in blocking HPV16 infection while having similar, if any, off-target effects. A2ti-1, A2ti-2 and DMSO matched to the concentration of A2ti delivery were found to be non-toxic at the maximum concentrations tested and did not affect total cells recovered after 72 h (Figure 1b and c). Before performing internalization and infection assays, we confirmed that A2ti targeted A2t with isothermal titration calorimetry and found that A2ti targeted the S100A10 dimer of A2t (Figure S1, available as Supplementary data at JAC Online). Next, we sought to determine the effect of A2ti on HPV16 PsV infection. We found that the higher-affinity A2ti-1 reduced HPV16 PsV infection of HeLa cells in a dose-dependent manner with 100% inhibition of infection observed at 100 mM (Figure 2a) and similar results were observed in HaCaT cells (data not shown). As predicted, lower-affinity A2ti-2 was less effective with ,50% reduction in HPV16 PsV infection achieved at 100 mM while DMSO vehicle had no effect. To determine whether the reduction in infection was due to a decrease in capsid entry, we next evaluated the ability of A2ti to block internalization of HPV16 into cells using PsV

(b) 100 000

A2ti-1 IC50 = 24 mM

N

100 mM A2ti-1

80 000

S

N

O

N

CH3

Total cell number

O

100

0.1% DMSO

NH2

80

100 mM A2ti-2

Viable cells (%)

CH3

CH3

(c) Untreated

60 000 40 000 20 000

60 40 20

NH2

A2ti-2 IC50 = 230 mM

S

N N

N

0 O

0 0

24

48

Hours in culture

72

Un tr ea te d 0. 1% DM 10 SO 0m M A2 ti10 1 0m M A2 ti2

O

Figure 1. A2ti do not affect cell growth and are non-toxic to HeLa cells. (a) Chemical structures and IC50 values of A2ti-1 and A2ti-2. HeLa cells were left untreated or treated with A2ti-1, A2ti-2 or DMSO. After 72 h, cells were counted (b) and viability was measured (c) via trypan blue exclusion. The mean+SD percentage viability is presented (n¼6). The graph is representative of three independent experiments.

2 of 5

JAC

A2t inhibitors prevent HPV16 infection

(a) 140

140

CH3

NH2

O

120 % HPV16 PsV infection (GFP+)

CH3

CH3

S

N N

O

NH2

O

120

N

N

100

S

N

O

N

100

80

80

***

60

***

** ***

60

*** 40

40

20

20

*** ***

CH3

CH3 O

S

N N

1.0

1.2

O

O

N

0 10

75

NH2

N

1.0

S

N N

**

*

O

*

0.8

** *** **

0.6

0.6

A2ti-1 concentration mM

0 10

75

50

25

Un tr ea te d 0. 1% DM SO

10

0

0.0 75

0.0 50

0.2

25

0.2

10

0.4

10

*** 0.4

Un tr ea te d 0. 1% DM SO

Fold change in MFI

CH3 NH2

(b) 1.2

50

A2ti-2 concentration mM

A2ti-1 concentration mM

0.8

25

Un tr ea te d 0. 1% DM SO

0 10

75

50

25

10

Un tr ea te d 0. 1% DM SO

10

0

0

A2ti-2 concentration mM

Figure 2. A2ti block HPV16 infection and entry into HeLa cells. (a) HeLa cells were treated with increasing concentrations of A2ti-1, A2ti-2 or DMSO control. The following day cells were infected with GFP-plasmid-containing HPV16 PsV. GFP-positive cells were measured after 48 h by flow cytometry. The mean+SD percentage of infected cells normalized to the PsV-only group is presented. (b) HeLa cells were incubated with increasing concentrations of A2ti-1, A2ti-2 or DMSO control. The next day, pHrodo-labelled HPV16 PsV were added to the cells for 6 h and the MFI of cells was analysed by flow cytometry. The mean+SD fold change in MFI normalized to the untreated group is presented (n¼ 4). Each graph is representative of three independent experiments (*P,0.05, **P,0.01 and ***P,0.001 as determined by a two-tailed, unpaired t-test compared with PsV only).

3 of 5

Woodham et al.

labelled with a pH-dependent dye (pHrodo) that fluoresces only in acidified late endosomes indicating HPV16 endocytosis. A2ti-1 significantly decreased HPV16 entry in a dose-dependent manner with a 65% reduction at 100 mM, whereas only a 20% reduction was observed with A2ti-2 and no effect on HPV16 internalization was observed with DMSO alone, indicating that the observed block in HPV16 infection with A2ti is due in part to a block in entry (Figure 2b). To ascertain that the decrease in pHrodo signal observed was not due to an effect of A2ti on endosomal acidification, entry of CFDA-SE-labelled PsV was also measured, which requires cleavage by cytoplasmic esterases for fluorescence independent of pH and, similarly, A2ti-1 significantly reduced CFDA-SE-labelled HPV16 PsV entry (Figure S2).

National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) of the NIH.

Discussion

Supplementary data

Our group previously identified A2t as an HPV16 uptake receptor using a combination of cellular, molecular and biochemical techniques including small hairpin RNA knockdown, antibody (Ab) neutralization and electron paramagnetic resonance. 6 Specifically, we demonstrated that the S100A10 subunit of A2t binds to amino acids 108 – 126 of HPV16 L2; A2t coimmunoprecipitates with HPV16 at the cell surface; A2t mediates HPV16 entry and infection in an L2-dependent manner; and a previously identified natural A2t ligand, secretory leucocyte protease inhibitor,14 reduced HPV16 PsV infection of epithelial cells.6 Dziduszko and Ozbun15 independently confirmed the role of A2t in HPV16 entry and infection by showing that (i) early HPV16 binding results in the translocation of A2t to the extracellular surface, (ii) A2t cointernalizes with HPV16 and mediates intracellular trafficking and (iii) anti-A2 and anti-S100A10 Abs block HPV16 PsV infection at different stages of HPV16 infection both pre- and post-entry. In the current study, we show a significant decrease in HPV16 PsV internalization and a complete reduction in HPV16 PsV infection in epithelial cells treated with a high-affinity A2ti (A2ti-1). Taken together, these results highlight the importance of A2t in HPV16 infection and demonstrate the potential for targeting A2t to block HPV16 infection. Additionally, A2t has been implicated in infection by other viruses including CMV, RSV, enterovirus 71 and HIV infection of macrophages. 14,16 – 18 Consequently, A2t inhibition has broad appeal as an antiviral strategy.

Transparency declarations None to declare.

Disclaimer The content is solely the responsibility of the authors and does not represent the official views of the NIH.

Figures S1 and S2 are available as Supplementary data at JAC Online (http://jac.oxfordjournals.org/).

References 1 Arbyn M, Castellsague X, de Sanjose S et al. Worldwide burden of cervical cancer in 2008. Ann Oncol 2011; 22: 2675 –86. 2 Haedicke J, Iftner T. Human papillomaviruses and cancer. Radiother Oncol 2013; 108: 397–402. 3 Hoffmann M, Quabius ES, Tribius S et al. Human papillomavirus infection in head and neck cancer: the role of the secretory leukocyte protease inhibitor. Oncol Rep 2013; 29: 1962 –8. 4 Forman D, de Martel C, Lacey CJ et al. Global burden of human papillomavirus and related diseases. Vaccine 2012; 30 Suppl 5: F12 – 23. 5 Raff AB, Woodham AW, Raff LM et al. The evolving field of human papillomavirus receptor research: a review of binding and entry. J Virol 2013; 87: 6062– 72. 6 Woodham AW, Da Silva DM, Skeate JG et al. The S100A10 subunit of the annexin A2 heterotetramer facilitates L2-mediated human papillomavirus infection. PLoS One 2012; 7: e43519. 7 Gerke V, Moss SE. Annexins: from structure to function. Physiol Rev 2002; 82: 331–71. 8 Becker T, Weber K, Johnsson N. Protein – protein recognition via short amphiphilic helices; a mutational analysis of the binding site of annexin II for p11. EMBO J 1990; 9: 4207 –13.

Acknowledgements

9 Reddy TR, Li C, Guo X et al. Design, synthesis, and structure– activity relationship exploration of 1-substituted 4-aroyl-3-hydroxy-5-phenyl-1Hpyrrol-2(5H)-one analogues as inhibitors of the annexin A2-S100A10 protein interaction. J Med Chem 2011; 54: 2080– 94.

W. M. K. holds the Walter A. Richter Cancer Research Chair. We acknowledge Sammie’s Circle for their support. We thank the Beckman Center for Immune Monitoring of the Norris Comprehensive Cancer Center for assistance with flow cytometry.

10 Reddy TR, Li C, Fischer PM et al. Three-dimensional pharmacophore design and biochemical screening identifies substituted 1,2,4-triazoles as inhibitors of the annexin A2-S100A10 protein interaction. ChemMedChem 2012; 7: 1435– 46.

Funding This work was supported by NIH grants R01 CA074397 and RC2 CA148298 (to W. M. K.), and NIH grant P30CA014089 (Cancer Center Support Grant). A. W. W. was supported by a TL1 Scholar award from the SC CTSI (NIH/NCRR/NCATS) Grant # TL1TR000132, and by the ARCS Foundation John and Edith Leonis Award. A. I. J. was supported by the Short Term Education Program for Underrepresented Persons (STEP-UP) from grant no. 2R25-DK078385-07, entitled ‘Kids Seek Cure for Kids’ issued by the

4 of 5

11 Situ AJ, Schmidt T, Mazumder P et al. Characterization of membrane protein interactions by isothermal titration calorimetry. J Mol Biol 2014; 426: 3670– 80. 12 Velazquez-Campoy A, Ohtaka H, Nezami A et al. Isothermal titration calorimetry. Curr Protoc Cell Biol 2004; Chapter 17: Unit 17.8. 13 Buck CB, Thompson CD. Production of papillomavirus-based gene transfer vectors. Curr Protoc Cell Biol 2007; Chapter 26: Unit 26.1. 14 Ma G, Greenwell-Wild T, Lei K et al. Secretory leukocyte protease inhibitor binds to annexin II, a cofactor for macrophage HIV-1 infection. J Exp Med 2004; 200: 1337– 46.

A2t inhibitors prevent HPV16 infection

JAC

15 Dziduszko A, Ozbun MA. Annexin A2 and S100A10 regulate human papillomavirus type 16 entry and intracellular trafficking in human keratinocytes. J Virol 2013; 87: 7502 –15.

17 Malhotra R, Ward M, Bright H et al. Isolation and characterisation of potential respiratory syncytial virus receptor(s) on epithelial cells. Microbes Infect 2003; 5: 123–33.

16 Wright JF, Kurosky A, Wasi S. An endothelial cell-surface form of annexin II binds human cytomegalovirus. Biochem Biophys Res Commun 1994; 198: 983– 9.

18 Yang SL, Chou YT, Wu CN et al. Annexin II binds to capsid protein VP1 of enterovirus 71 and enhances viral infectivity. J Virol 2011; 85: 11809–20.

5 of 5

Small molecule inhibitors of the annexin A2 heterotetramer prevent human papillomavirus type 16 infection.

High-risk human papillomavirus (HPV) infection leads to the development of several human cancers that cause significant morbidity and mortality worldw...
220KB Sizes 0 Downloads 9 Views