1. Introduction
Human papillomaviruses (HPV) are important pathogens that play an etiological role in approximately 5% of all cancers worldwide, including at least 99% of cervical cancers [
1]. There are effective prophylactic vaccines against some HPV types, but these vaccines have no effect on ongoing HPV infection, do not protect against all HPV types, and are not widely deployed in much of the world. Therefore, study of HPV infection may lead to important new therapeutics to reduce the spread of the virus and disease burden. In addition, thorough investigation of HPV infection is likely to provide new insights into many aspects of cell biology and biochemistry [
2].
The HPV virion is non-enveloped and contains 360 molecules of the major capsid protein, L1, and up to 72 molecules of the minor capsid protein, L2 [
3]. Upon binding to heparan sulfate proteoglycans on the cell surface or extracellular matrix, HPV undergoes conformational changes and proteolytic cleavage and is transferred to an unidentified cell surface receptor for internalization [
4,
5]. HPV entry then occurs through the retrograde pathway: after cell internalization, the virus traffics from endosomes to the trans-Golgi network (TGN) and Golgi apparatus, possibly the endoplasmic reticulum (ER), and finally the nucleus. Internalized HPV remains within vesicular compartments until breakdown of the nuclear envelope during mitosis, which allows the viral genome to access cellular DNA replication machinery [
6]. Virions assembled from L1 only are noninfectious and do not reach the nucleus, and many L2 mutants have been described that display trafficking defects [
7,
8,
9,
10,
11,
12,
13]. These results show that L2 plays an important role in trafficking of the viral particle during HPV entry. Cellular proteins such as retromer and -secretase also play important roles in HPV entry.
A cell-penetrating peptide (CPP) in the C-terminus of L2 is also required for proper trafficking of HPV [
9,
14]. CPPs are short peptide segments that can transfer proteins and other cargo into cells [
15]. Our results showed that the HPV L2 CPP mediates transfer of protein segments across intracellular membranes. Specifically, when internalized HPV is in the endosome, the CPP penetrates the endosomal membrane, and a segment of L2 upstream of the CPP protrudes through the endosomal membrane into the cytoplasm [
9,
16]. This allows L2 to bind to cytoplasmic entry factors such as the retromer complex, which normally mediates retrograde trafficking of cellular transmembrane (TM) cargo proteins from the endosome to TGN. Retromer-L2 binding is essential for HPV exiting the endosome and entering the retrograde transport pathway [
8,
17].
Early during entry when HPV resides in the endosome, HPV engages with γ-secretase, a protein complex comprised of four subunits (nicastrin [NCT], presenilin 1 [PS1], PEN2, and APH1), each of which have at least one TM domain [
7,
18]. Typically, γ-secretase recognizes and cleaves TM proteins such as Notch and the amyloid precursor protein (APP) within their TM domain [
19]. Knockdown of any of the four γ-secretase subunits or treatment with chemical inhibitors of γ-secretase proteolytic activity, such as compound XXI, cause a severe defect in HPV infection and block intracellular HPV trafficking during entry [
17,
20,
21,
22]. Additionally, we showed that γ-secretase binds to L2 and promotes stable insertion of L2 into the endosome membrane [
7]. The cellular protein p120 catenin is required for HPV-γ-secretase interaction [
23]. The mechanistic details of how γ-secretase associates with L2 and promotes insertion of L2 into the membrane have not fully been elucidated. Dopachrome tautomerase (DCT), a γ-secretase substrate, supports HPV entry, but it is not known whether cleavage of DCT is required for HPV trafficking [
24].
In vitro experiments with purified components showed that a short N-terminal segment of L2 appended to GFP binds directly to γ-secretase [
7]. This segment of L2 contains a conserved hydrophobic, glycine-rich segment that appears to act as a TM domain, but it is less hydrophobic than canonical TM domains. Although γ-secretase can cleave within this putative TM domain, cleavage of L2 by γ-secretase is not required for successful infection. Glycine to valine mutations in this segment of HPV L2 inhibit the association of HPV with γ-secretase and retromer in infected cells, severely inhibit infection, and impair proper HPV trafficking to the TGN [
7,
10]. Others have also reported that some mutations in this TM segment of L2 drastically block HPV entry [
10,
11]. The ability of these mutants to associate with cellular factors such as γ-secretase was not investigated, and the precise nature of their trafficking defect was not determined. This segment of bovine papillomavirus (BPV) L2 is also important for infectivity and engages the SNARE protein syntaxin 18, but it is not known if syntaxin 18 is required for BPV or HPV infection [
25,
26].
Most of the information summarized above was obtained through studies of HPV pseudoviruses (PsVs). HPV PsVs are comprised of the L1 and L2 proteins, which assemble into a capsid that encapsidates a plasmid that expresses a reporter protein such as green fluorescent protein (GFP). The reporter gene is expressed upon successful infection of target cells and delivery of the reporter plasmid to the nucleus. PsVs provide several advantages for the study of HPV entry. They are easy to produce in monolayer cultures of cells, whereas the production of authentic virus requires stratified epithelial cells. Second, PsVs are non-pathogenic because they lack viral genes. Third, infectivity is simple to quantify, for example, by flow cytometry for reporter protein fluorescence. Finally, unlike the situation with authentic virus, it is relatively simple to generate and characterize specific viral mutants, including L2 proteins containing an epitope tag, L2 mutants that are defective for entry, and PsVs that lack L2. Although PsVs do not express any viral genes, they appear to act remarkably similarly to authentic virions during entry [
27]. Thus, they are well suited to investigate the entry process, which is mediated solely by the viral capsid proteins in the incoming virus particle working in concert with cellular proteins.
In this paper, we use HPV PsVs to show that infection by divergent HPV types stabilizes the γ-secretase complex in multiple cell lines. γ-secretase stabilization requires the L2 protein and is tightly correlated with the infectivity of L2 mutant PsVs. Additionally, we found that γ-secretase activity and the HPV TM domain are required for protrusion of L2 into the cytoplasm. Taken together, our findings suggest that stabilization of γ-secretase by HPV may promote stable membrane protrusion of L2 and that specific residues within the putative TM domain of L2 play a critical role in this process.
2. Materials and Methods
2.1. Cell Lines
HeLa S3 cells were purchased from American Type Culture Collection (Manassas, VA, USA) HaCaT cells were purchased from AddexBio Technologies (San Diego, CA, USA), and 293TT cells were obtained from Christopher Buck (NIH). All cell lines were cultured at 37 °C and 5% CO2 in DMEM supplemented with 2% HEPES, 10% fetal bovine serum, l-glutamine, and 100 units/mL penicillin–streptomycin.
2.2. Generation of Mutant HPV16 Pseudovirus Packaging Plasmids
A silent unique AvrII restriction site was inserted downstream of the putative L2 TM domain in the p16SheLL-3X FLAG vector using the Q5 site-directed mutagenesis protocol and Phusion High Fidelity Polymerase. This plasmid was used to generate the putative TM domain mutants by inserting gBlocks (Integrated DNA Technologies, Coralville, IA, USA) containing the relevant mutation between the AvrII restriction site and an XbaI site upstream of the L2 start codon. The GV mutant was previously described [
7]. For split GFP experiments, we generated PsVs from p16sheLL-CPP-GFP11, which contains seven copies of GFP11 appended to the C-terminus of L2 in wild-type or mutant PsV preparations [
9].
2.3. HPV Pseudovirus Production
HPV PsVs were produced by using polyethylene imine (PEI) (MilliporeSigma, Burlington, MA, USA) to transfect 293TT cells with wild-type or mutant p16L1-GFP (to generate L1 only PsVs), p16SheLL, p16SheLL-3XFLAG, p5SheLL, or p16SheLL-GFP11 together with pCAG-HcRed or pCINeo-GFP as a reporter gene. PsV was collected and purified using an OptiPrep gradient (MilliporeSigma), as described previously [
28]. In brief, cells were collected 72 h post transfection in siliconized tubes and lysed. Crude PsV preps were incubated overnight at 37 °C to allow capsid maturation. Matured PsV preps were loaded on an OptiPrep gradient and centrifuged at 50,000×
g for 4 h at 4 °C in a SW-55Ti Beckman Ultracentrifuge rotor (Beckman Coulter, Indianapolis, IN, USA) rotor. Fractions were collected, and L1 and L2 levels of purified PsV preparations were assessed by SDS-PAGE followed by Coomassie blue staining or by immunoblotting with antibodies recognizing L1 and L2.
2.4. qPCR for Reporter Gene Quantitation
Encapsidated HcRed genomes were quantitated by qPCR as described previously [
8]. In brief, gradient-purified PsV preps were treated with DNAse I to remove unencapsidated DNA, followed by proteinase K to digest capsids. The reporter plasmid was purified with the Qiagen DNeasy Blood and Tissue kit (Qiagen, Germantown, MD, USA) followed by qRT-PCR with primers specific to HcRed in comparison to a standard curve to determine viral genome quantity.
2.5. Infectivity
One day before infection, 1 × 105 HeLa S3 or HaCaT cells were plated in 12-well plates. Cells were mock-infected or infected with wild-type or mutant PsV, and flow cytometry was used to measure reporter gene expression 48 h post infection (h.p.i.). The amount of mutant PsV used to infect cells was normalized to wild-type HPV16 PsV by using equal levels of L1 and L2 in purified PsV or qRT-PCR for the encapsidated reporter plasmid.
2.6. Co-Immunoprecipitation of L2-FLAG and γ-Secretase
Twenty-four hours before infection, 1 × 10
6 HeLa S3 cells were plated in 6 cm dishes. Cells were infected with wild-type or mutant HPV PsV at a multiplicity of infection (MOI) of 50 for 16 h, unless otherwise indicated. Cells were scraped off the dishes and lysed in HN-DMNG lysis buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 1% decyl maltose neopentyl glycol (DMNG) (Anatrace, Inc., Maumee, OH, USA) supplemented with HALT protease inhibitors) on ice for 45 min. For the co-immunoprecipitation experiment, in which γ-secretase subunits were stripped away from HPV using different amounts of detergents, samples were prepared the same, except HN lysis buffer with combinations of
N-Dodecyl-B-
d-Maltoside (DDM) (Anatrace) and NP40 was used (total detergent concentration, 1%). Cell debris was removed from the sample by centrifuging at 16.1×
g for 15 min at 4 °C. The supernatant was incubated with anti-FLAG antibody for 4–6 h at 4 °C on a rotating tube rack. Then, 50 µL of Protein G magnetic beads (Thermo Fisher Scientific, Waltham, MA, USA) were washed in TBS-T, added to the lysate, and incubated at 4 °C overnight. Samples were washed three times with TBS-T and eluted from the beads using 2X Laemmli sample buffer (4% SDS, 20% glycerol, 0.004% bromophenol blue, 0.125 M Tris-Cl, pH 6.8, 10% Dithiothreitol (DTT)) at 100 °C. The entire sample was subjected to SDS-PAGE, transferred to a 0.2 µM PVDF membrane, blocked with 5% milk in TBS-T, and immunoblotted with antibodies recognizing the FLAG tag on the C-terminus of HPV16 L2 and γ-secretase substrates. See
Table S2 for antibody information and dilutions.
2.7. Assay for γ-Secretase–HPV Association and γ-Secretase Stabilization
Twenty-four hours before infection, 1 × 106 HeLa S3 cells were plated in 6 cm dishes. Cells were infected with wild-type or mutant HPV PsV at a MOI of 50 for 16 h. Cells were scraped off the dishes and lysed in HN-DDM lysis buffer (50 mM HEPES pH 7.5, 150 mM NaCl, 1% DDM supplemented with HALT protease inhibitors) on ice for 45 min. Where indicated, cells were lysed in HN lysis buffer containing 1% of the indicated detergent (CHAPSO, DDM, or NP40). Cell debris was removed from the sample by centrifuging at 16.1× g for 15 min at 4 °C. The supernatant was incubated with anti-PS1 or -APH1 antibody for 4–6 h at 4 °C on a rotating tube rack. Then, 50 µL of Protein G magnetic beads were washed, added to the lysate, and incubated at 4 °C overnight. Samples were washed three times and eluted from the beads using 2X Laemmli sample buffer at 100 °C. The entire sample was subjected to SDS-PAGE, transferred to a 0.2 µM PVDF membrane, blocked with 5% milk in TBS-T, and immunoblotted with antibodies recognizing HPV16 L1, the FLAG tag on the C-terminus of HPV16 L2 or γ-secretase substrates.
2.8. Split GFP Assay
HaCaT cells expressing GFP1-10NES were generated as described previously [
9]. For the split GFP assay, 2.5 × 10
4 GFP1-10NES-expressing cells were plated in eight-chambered glass slides overnight. Cells were treated with 1 µM of XXI γ-secretase inhibitor (MilliporeSigma) for 30 min prior to infection or left untreated, then infected with wild-type HPV16 PsV with a FLAG tag or with seven copies of GFP11 fused to the C-terminus of L2, or with mutant HPV PsV with seven copies of GFP11 fused to the C-terminus of L2. All PsVs were infected at a MOI of 2000. Live cells were stained 3 h post infection with Hoescht 33342 for 15 min at 37 °C to visualize DNA. Live cells were analyzed for reconstituted GFP fluorescence using a Leica SP5 confocal microscope.
4. Discussion
Our study of the role of γ-secretase in HPV entry generated several interesting findings. First, HPV is in a complex with all four subunits of the γ-secretase complex and interacts with the catalytic subunit PS1. This result is consistent with recently published crystal structures of other substrates bound to γ-secretase, which showed that the TM helix of Notch and the amyloid precursor protein (APP) are surrounded by TM helices of PS1 [
30,
31]. The interaction of HPV with the catalytic subunit of γ-secretase appears vital for HPV entry, consistent with the fact that inhibitors of γ-secretase activity block HPV infection, even though protease activity is not required for successful infection.
Second, co-IP experiments with antibodies recognizing two different γ-secretase subunits showed that infection with HPV capsids containing L2 stabilizes the γ-secretase complex, whereas PsV capsids devoid of L2 do not stabilize the complex. Both α- and β-papillomaviruses stabilize γ-secretase, stabilization occurs at multiple MOIs in a dose-dependent manner, and stabilization occurs in HeLa cervical cancer cells and HaCaT immortalized keratinocytes. Thus, the ability of HPV to stabilize γ-secretase is conserved among diverse papillomavirus types in relevant cell types.
Third, we confirmed that the putative TM domain in the N-terminal portion of L2 is required for infection and γ-secretase binding. In addition, we showed that this segment of L2 is required for γ-secretase stabilization and for protrusion of L2 through the endosome membrane. By using the PsV system to generate and test TM domain mutants, we showed that the L2 TM domain could not be removed or replaced with the TM domain of a known γ-secretase substrate, and that single residue substitution mutations within the TM domain can have a large effect on HPV infection, HPV–γ-secretase interaction, and HPV trafficking. There is a correlation between γ-secretase stabilization, γ-secretase binding to HPV L2, and infectivity: the mutants that were the least infectious failed to bind to or stabilize the γ-secretase complex, while those that were partially infectious both bound to and stabilized γ-secretase. We did not identify a mutant that could bind to γ-secretase without stabilizing it. These results show that the intact L2 TM domain is vital for recognition of L2 by γ-secretase and imply that γ-secretase–L2 binding is required for γ-secretase stabilization and for the subsequent membrane protrusion and trafficking events. They further raise the possibility that γ-secretase stabilization plays a role in infection.
Our results extend the findings of others that point mutations in this portion of the L2 TM domain can inhibit infection by HPV16 and BPV PsVs [
11,
25]. The Jung group found that an L46K mutant HPV16 PsV was almost as infectious as wild-type PsV, whereas PsV containing a triple mutation of L46, Q47, and Y48 to three alanines was essentially noninfectious. Additionally, they showed that a replacement of residues 49–53 with five alanines retained roughly 90% infectivity of wild-type HPV PsV. Residues other than L46 within this six-amino acid region were not tested individually. We found that L46, G49, and S50 are critical for infection, and that mutations at Q47, Y48, and M51 impaired but did not abrogate infection. There are two key differences between the two studies that could contribute to these conflicting results. The Jung group assessed infectivity of their mutants in CHO-K1 hamster kidney cells, which is not a natural target for papillomavirus infection, whereas we assessed mutants in cells representative of the natural target of HPV. In addition, the Jung group replaced the residues with alanine, and we replaced them with leucine (the most abundant residue within TM domains [
32]). In addition to the importance of individual residues, the sequence context of the L2 M domain also appears important. Mutations in the HPV16 L2 TM domain that replace the native residue with a residue found in other papillomavirus types, such as the G49L mutant, are much less infectious than the wild-type HPV16 PsV, even though leucine is present at this position in other HPV types, such as HPV95. Perhaps glycine but not leucine at this position in the context of the HPV16 TM domain allows for proper γ-secretase association, whereas leucine at this position in HPV95 allows association in the presence of the other sequence differences in the HPV95 TM domain.
The stabilization of γ-secretase by HPV L2 is intriguing. To our knowledge, stabilization of γ-secretase by substrates has not been previously reported, but some γ-secretase inhibitors have been reported to stabilize the γ-secretase complex by increasing interactions between γ-secretase subunits [
33]. The structure of the γ-secretase complex bound to Notch or APP provides insight into substrate recognition [
30,
31]. Many of the residues in PS1 that contact Notch and APP differ, but there are some common elements. Both substrates interact with multiple TM domains of PS1 and induce the formation of two β-sheets in PS1 that interact with a β-sheet in the substrate, and hydrogen bonds involving S169 or G384 of PS1 anchor the interaction with the substrate TM domain. These common features may be important for substrate cleavage. While the structure of HPV L2 bound to γ-secretase is unknown, L2 likely interacts with PS1 in a different way than either canonical substrate, because L2 cleavage is minimal, the L2 TM domain scores lower than confirmed TM domains in TM prediction programs, and the TM domain of a known γ-secretase substrate cannot functionally substitute for the L2 TM domain [
7,
10]. We speculate that the interactions between HPV L2 and γ-secretase produce different structural arrangements of PS1 that stabilize its interactions with the other γ-secretase subunits. Alternatively, L2 itself may act as a “glue” by simultaneously contacting multiple γ-secretase subunits. It will be interesting to identify other cellular proteins that stabilize γ-secretase and to determine whether their TM domains can replace the L2 TM domain and support HPV infection.
Finally, protrusion of L2 into the cytoplasm requires γ-secretase activity, consistent with our previous finding that L2 membrane insertion requires γ-secretase. This suggests that the infectivity defect caused by γ-secretase inhibition is ultimately due to impaired L2 protrusion. The TM domain of L2 is also important for protrusion of L2 into the cytoplasm, presumably at least in part because it binds γ-secretase. The non-canonical TM sequence may be beneficial to the virus. As the L2 TM domain is less hydrophobic than typical TM domains, L2 might be able to move into and out of the membrane as necessary for proper viral trafficking. L2 protrudes through the membrane to bind γ-secretase and retromer, but perhaps it is released from membrane association in order to traffic to distal retrograde compartments. Experiments are underway to test these possibilities.
Overall, we have characterized the interaction between γ-secretase and HPV L2 and discovered that HPV infection stabilizes the γ-secretase complex, identified specific residues in the L2 TM domain that are required for stabilization, and provided evidence that stabilization may be important for infection. Additionally, γ-secretase activity and the L2 TM domain are required for L2 protrusion into the cytoplasm, a vital step in infection that allows L2 to bind to cytoplasmic cellular trafficking factors. γ-secretase stabilization may also be important for the processing, trafficking, and modulation of cellular γ-secretase substrates, and it will be interesting to investigate whether stabilization contributes to the pathogenesis of disorders related to improper γ-secretase function.