Introduction

PVs are established agents of human and animal cancers [1]. They infect cutaneous and mucous epithelia inducing benign tumours which usually regress. Occasionally, the tumours progress to malignancy. Over 120 types of HPVs have been identified so far and among these 15 have been defined as HR HPVs. These are consistently associated with cancer. Genital HPVs are sexually transmitted and HR genital HPVs are a necessary factor in the development of almost all cases of cervical cancer. HPV-16 and -18 are the viruses most frequently associated with cancer of the uterine cervix (CxCa) [2].

CxCa is the second most common cancer in women worldwide killing about 0.25 million women per year. However, in economically developed countries the rate of CxCa is dramatically reduced due to screening program based on exfoliative cervical cytology (PAP smears).Vaccines to prevent HR HPV infection are available, although their use should be implemented along with screening programmes to further reduce the incidence of such cancer [3]. HR HPVs are also involved in the etiopathogenesis of other anogenital cancer [4]. Furthermore HPV, particularly HR HPV-16 is strongly associated to oral squamous cell carcinoma and other potentially malignant oral lesions [5]. Growing evidence also suggests that HR HPV-16 is involved in the etiopathogenesis of head and neck squamous cell carcinomas, suggesting that HPV vaccines should be also considered for prevention of this type of cancer [5, 6]. Additionally, HPVs may be involved in the etiopathogenesis of others cancer types, including tumours of the upper respiratory tract, eye, esophagus, non-small-cell lung cancers [710]. The presence of HPV-16 has been reported also in colorectal carcinoma [11], breast cancer [12] and urinary bladder carcinoma [13]. Recently, HPV DNA has been associated also with prostatic tumours [14]. The widespread causal association of PVs with cancer makes their study worthwhile not only in humans but also in animal model systems which often provide new and profitable avenues of research [15]. The BPV system has been one of the most useful animal models in understanding the oncogenic potential of PVs. Furthermore, the mechanisms by which BPV induces tumors are an outstanding model to better understand the pathogenesis of other cancer types. The importance of the role of HPV in cancer etiology and development has been recognized by the assignment of the 2008 Nobel Prize for Medicine to Prof. Harald zur Hausen who firstly observed that infection with HPVs is responsible for CxCa development. The genome of PVs is a double stranded circular DNA roughly divided into three parts: the E region coding for early proteins (E1-E7) responsible for the pathogenicity of the virus; the L region coding for late structural proteins (L1, L2) and a non coding region which contains the cis-elements necessary for replication and transcription of the viral genome. Both in vivo and in vitro studies have pointed to E6 and E7 as the main HPV oncogenes, whereas E5 is the major oncogene of BPV. The E6 oncoprotein interacts with the cellular tumour suppressor p53 [16] and directs its degradation [17]. The primary target of the E7 oncoprotein is the retinoblastoma (Rb) proteins, the inactivation of which leads to tumour progression [18]. Both E6 and E7 also interact with many others cellular factors inducing genomic instability, tumour progression and immune evasion [18].

While the oncogenic activities of E6 and E7 are well characterised, the role of E5 is still rather nebulous. However, recent studies have highlighted the important role of this oncoprotein in cell transformation, tumourigenesis and immune modulation, thus implicating E5 in pivotal steps of carcinogenesis. Because of the perceived growing role of E5 in infection establishment and in cell transformation, it is worth considering the most salient features of the oncoprotein. This review will focus on the activities of E5, in particular from HPV-16, and from BPV. Targeting E5 as a possible therapeutic agent will also be briefly discussed.

HPV E5

E5 and the evolution of HPVs

HPVs have been classified into five genera (Alpha-, Beta-, Gamma-, Mu- and Nu-PVs), but not all HPV genera code for an E5 protein [19]. For instance αHPVs encode and express E5 but β HPVs do not. A phylogenetic analysis of known genital HPV types (derived using a Bayesian methodology, with notation of carcinogenic risk levels assigned by the large case-control study conducted by the International Agency for Research on Cancer [20]), suggested that at least 3 ancestral papillomaviruses are responsible for the current heterogeneous group of genital HPVs. The three major groups that emerged include alpha papillomavirus species and interestingly all carcinogenic types derive from a common ancestor and can code for an E5 protein, whereas the other HPVs either lack a definable E5 ORF (Open Reading Frame) or a translation start codon for E5. As notable exceptions, 10 types which cause benign venereal warts, can also code for an E5 protein. In addition, 16E5 variants with the greatest mitogenic activity in vitro[21] are most frequently detected in the population and most commonly associated with cervical lesions.

The HPV E5 ORF itself has been classified into four different groups: alpha, beta, gamma and delta [22], which correlate with different clinical manifestations, in particular with oncogenic potential [23]. Thus, the E5-alpha protein is encoded by HR αHPV, whereas the E5-gamma and E5-delta proteins are encoded by low-risk genital HPVs [24]. The phylogenetic analysis suggests that E5 must give some advantage to the virus expressing it and variants of this protein appear to increase the likelihood of oncogenic transformation following persistent infections. However, the E5 ORF is absent in the genome of many HPVs, such as beta-, gamma- and mu-HPVs, indicating that the protein is not essential for the life cycle of these viruses but rather can give some ADDED VALUE to favour infection and transformation (Figure 1). The HPV E5 ORF is expressed during the early phases of the viral life cycle but only as the fourth ORFs on polycistronic transcripts. Since HPVs are thought to use a leaky ribosome-scanning mechanism to translate proteins from polycistronic mRNAs, little E5 protein is likely to be synthesized from these transcripts. In contrast, on epithelial-cell differentiation, E5 is expressed as the second ORFs of late transcripts. It is therefore likely that E5 is synthesized highly in differentiating suprabasal epithelial cells [25] (Figure 1). In agreement, the BPV-1 E5 protein was detected at a low uniform level in basal layers and at a higher level in the uppermost layers of stratified squamous epithelium in papillomas productively replicating BPV-1 [26]. In addition, E5 was also shown to be expressed in basal and suprabasal layers of BPV4-induced papillomas [27] and of HPV-16 induced cervix lesions [28].

Figure 1
figure 1

E5 can improve HPV activity by altering host factors controlling the viral replication/persistence. A schematic view of the papillomavirus life cycle highlighting the expression of the HPV genes, which is tightly regulated and strictly linked to epithelial differentiation. E5 could contribute to a successful infection by inducing loss of surface MHC I expression in the infected basal cells preventing presentation of viral antigens to effector T-cells and thus, in addition to other mechanisms of immune avoidance, such as lack of inflammation, contributing to evasion of immune surveillance. Expression of E5 in the basal/suprabasal layers of the epithelium would lead to sustained cell proliferation to favour virus-infected cells, but extinction of its expression in the more superficial layers would permit cell differentiation and virion production. If E5 expression proceeds beyond early lesional stages, keratinocyte differentiation and immunological removal of infected cells would not take place and the lesion would be at greater risk for neoplastic progression. E5 actions on host factors controlling viral replication/persistence are indicated. Dashed dots indicate low levels of E5 gene expression.

In contrast to the highly transforming BPV-1 E5, the HPV E5 proteins display weak transforming activity in vitro (Table 1). Experiments with HPV-6 provided the first evidence that a HPV E5 protein had transforming activity in mammalian cells, as expression of HPV-6 E5 in established murine fibroblasts lead to anchorage independent growth [29]. Later it was shown that also 16E5 induces anchorage independence, more efficient growth in low serum and tumorigenic transformation of murine keratinocytes and fibroblasts [3032]. In addition, the acute expression of 16E5 stimulates cellular DNA synthesis in primary human keratinocytes, and in cooperation with E7, induces proliferation of primary rodent cells [3336]. The transforming activity of E5 from HPV-59 and rhesus papillomavirus has been demonstrated in various cell types and assays [37, 38].

Table 1 Comparison of function of HPV E5 proteins with BPV E5 proteins.

Structure and cell interaction

16E5 is 83 amino acid long. The detection of this protein has proved very difficult given its extreme hydrophobicity, membrane localisation and very low levels of expression. For these reasons expression of E5 is often inferred from the presence of E5 mRNA. 16E5 has been detected by immunohistochemistry by Chang et al. [28] in low-grade squamous intraepithelial lesions (LSILs), in high-grade SILs, CIN (cervical intraepithelial neoplasia) and paradoxically in cancer lesions although often this gene is deleted during viral integration into the host genome. Amino acid sequence analyses of 16E5 suggest it comprises 3 anchor-like α-helices (residues 8-30, 37-52 and 58-76), with only the first being long enough to span a lipid bilayer.

When over-expressed, 16E5 is present in the endoplasmic reticulum (ER), in the nuclear envelope and in the Golgi apparatus (GA) [39], whereas at a more physiological level of expression, as shown in primary human foreskin keratinocytes, it localizes almost exclusively to the ER and to lesser extent to GA and early endosomes [40]. Recently, it was reported that in HaCaT cells about 10% of total 16E5 can localize to plasma membrane with intracellular amino terminus and extracellular carboxyl-terminus. This observation suggests a fusogenic role of 16E5 with the induction of cell-cell fusion and the formation of binucleated HaCaT cells [4143]. In contrast, other authors report the presence of 16E5 protein only in the ER of COS and ectocervical cells in the opposite orientation: intraluminal amino-terminus and cytoplasmic carboxyl-terminus [44]. The cytoplasmic localization of the C terminus is further strengthened by the reported interaction of 10 amino acids at the C terminus of 16E5 with karyopherin β3 (KNβ3) [45], an abundant cellular protein localized mainly in the cytoplasm [46, 47]. An explanation for these contrasting data is the possible presence of 16E5 in the plasma membrane as a consequence of the over-expression of 16E5 by the adenovirus vector system used in HaCaT cells. In agreement, when BPV-1 E5 is expressed at very high levels, such as in baculovirus-infected cells, it is detectable on the plasma membrane in addition to the ER and GA [48].

16E5 self-associates in vitro[49] and in vivo[40, 50] and this oligomerization takes part mostly by hydrophobic interaction. The reported disulfide bonding [40] appears less likely because models of 16E5 membrane topology predict the localization of all six cysteine residues within the lipid bilayer [21, 51] rendering cysteine dimerization more difficult.

E5 and cell transformation

E5 interacts with a number of cellular proteins and these interactions are deemed important for the biological activity of the protein in cell transformation and evasion of the immune response. Thus the first transmembrane domain of 16E5 and HPV-31E5 interacts directly with the heavy chain component of the MHC I (Major Histocompatibility Complex class I) via the leucine pairs present in this region; interestingly this same transmembrane domain interacts with a chaperone of MHC I, Bap31 [52, 53]. A region within the second helix (residues 41-54) may be the binding site for the 16 kDa pore sub-unit of vacuolar-ATPase (V-ATPase) [54], although others claim this is located between residues 54-78 [55].

E5 is a weak transforming protein in vitro and its effects are seen best when in co-operation with the other viral oncoproteins: 16E5 together with E6 can induce the formation of koilocytes, large cells with cleared cytoplasm and pyknotic nuclei with inconspicuous nucleoli, a well known morphological marker of HPV infection [56]. The development of koilocytotic vacuoles may be linked to the E5-induced relocalization of calpactin I to the perinuclear region promoting perinuclear membrane fusion [57]. Hu et al. [43] confirmed previous reports describing the oncogenic capacity of 16E5 [30, 58] and offered mechanistic insights into how E5 expression brings about morphological changes in the cervical epithelium. Further, by identifying endoreplication as the mechanism by which the aberrant nuclei form and increased DNA synthesis arises, there is now a biological process that can be targeted to inhibit oncogenesis [43]. Fusogenic 16E5 is expressed on the plasma membrane of cells and both cells must express E5 for cell-cell fusion to occur, indicating that E5 cannot induce cell-cell fusion through an interaction with another protein (receptor) on a neighbouring cell, but rather forms E5-E5 dimers or complexes containing at least two 16E5 molecules. These findings provide important insight into how the fusogenic process is mediated [41].

E5 of HR HPV by itself can induce morphological and chromosomal changes that frequently accompany the progression of normal cells to cancerous cells. Increased nuclear size, increased DNA content and tetraploidy are characteristic of LSIL [59, 60]. All of these morphological changes have been detected in cell expressing 16E5 and, in particular, the aberrant nuclei formation seems to be due to endoreplication rather than a consequence of cell-cell fusion and failed cytokinesis [43]. Many of these changes are criteria used in the clinical detection of cervical cancer precursors in screening Pap tests [61]. Other morphological changes, such as the presence of binucleated cells, increased DNA content and polyploidy [62], are not used in the diagnosis of precancerous lesions despite being less subjective because they require larger amounts of material, time, and expenses.

HPV E5 interaction with host cell

HPVs infect stratified epithelia, and their whole life cycle is inextricably linked to keratinocyte differentiation and, in order to establish a persistent infection, HPVs have evolved to overcome many "obstacles":

  1. a)

    The stratified epithelium is associated with continuous cellular turnover and desquamation of the terminally differentiated keratinocytes, thus maintenance of the papillomavirus within the tissue requires the infection of basal epithelial cells and the propagation of these infected cells.

  2. b)

    Papillomaviruses use the cellular machinery for their replication and need to maintain cell division together with a delayed but not completely inhibited differentiation. HPV infected keratinocytes with imbalanced DNA synthesis/differentiation are forced to apoptosis by the cellular control mechanism.

  3. c)

    To establish persistent infection HPV must fight or evade immune-surveillance.

It is clear that the timely and epithelial differentiation-dependent expression of all E proteins is essential to favour viral replication and in turn to overcome the above mentioned obstacles. The "E5 added value" will be highlighted in the following paragraphs.

E5, growth factors and cell cycling

HPV infection is thought to take place when epithelial basal cells are directly exposed to the virus during microinjuries [63]. Infection of epithelial stem cells, which have the capacity for self-renewal, can ensure long term maintenance of the viral genome. During the productive stage of the HPV life cycle, the early viral proteins are expressed, maintaining 50-100 copies of episomal DNA per cell by synchronous replication with host cell DNA [6466]. The demonstrated ability of 16E5 to enhance ligand-dependent activation of the EGF-R [56, 5860] (Figure 2) and to stimulate EGF-dependent proliferation of cultured human keratinocytes [33, 35, 58] suggests that 16 E5 may play a major role in expanding populations of HPV-16-infected basal keratinocytes in vivo by enhancing ligand-dependent EGF-R activation. Confirmation of this E5 activity comes from a study on 16E5 transgenic mice showing that functional EGF-R is required for the induction of epidermal hyperplasia and formation of spontaneous skin tumours [67]. 16E5 oncoprotein binds and inhibits the activity of the 16 kDa subunit of V-ATPase, altering the endosomal acidification and degradation of EGF-R [34, 68] thus enhancing its recycling to the plasma membrane. 16E5 can also delay EGF-R degradation by interfering with membrane trafficking and the fusion of early and late endosomes [69]. Recently, 16E5 was shown to form a complex with KNβ3 which is localized mainly in the cytoplasm near the nuclear envelope [45]. It has been suggested that a KNβ3/16E5 complex plays an important role in vesicle trafficking, reinforcing the hypothesis that E5 has a central role in altering protein trafficking inside the cell. The EGF-R signalling pathway can be activated by 16E5 through either EGF-dependent or EGF-independent processes. 16E5 activates mitogen-activated protein kinase (MAPK) p38 and ERK1/2 in human keratinocytes in an EGF-independent manner [70]. Two different pathways, a receptor tyrosin kinase-mediated pathway and a protein kinase C (PKC)-dependent pathway, are involved in the MAPK activation [71, 72] which increases the transcription of c-fos and c-jun[33, 73, 74], forcing the cells through the cell cycle and stimulating transcription of the viral oncogenes E6 and E7. In contrast with EGF-R data, less is know about the role of the interaction of 16E5 with other ErbB family members such as ErbB2 or ErbB4 receptor [28, 7577].

Figure 2
figure 2

HPV 16 E5 enhances growth factor signalling pathways. Activation of EGF-R and the downstream Ras-Raf-MAP kinase pathway or PI3K-Akt pathway leads to altered cell proliferation, angiogenesis, and anti-apoptosis. The last two functions are further enhanced by the E5-induced upregulation of COX-2 expression. COX-2 expression inhibits hydrogen peroxide-induced apoptosis and induces PGE2 secretion that activates EP4 receptor. This, in turn, causes cAMP production, activating PKA; PKA contributes to E5-induced expression of EP4 by enhancing CREB binding to variant CRE of the EP4 promoter. PKA influences also apoptosis through the ubiquitin-proteasome degradation of BAX. Finally, E5 protein enhances cell proliferation through downregulation of tumor suppressor p21/p27 and through up-regulation of G protein-coupled endothelin receptor (ETA)/ET1 autocrine loop. Dotted arrow shafts indicate uncertain pathways. The possible cross-talking between ETAR and EGFR pathway through Src protein is also indicated.

16E5 is also capable of interacting with, and enhancing the signalling of, different classes of growth factor receptors like the G protein-coupled endothelin receptor (ETA) [36]. In growth factor-starved keratinocytes 16E5 enhances the mitogenic activity of endothelin-1 (ET-1) the specific ligand of ETA (Figure 2). Interestingly it was reported that the mitogenic activity of ET-1 may lead to the chronic stimulation of keratinocyte proliferation observed in psoriasis, an inflammatory/proliferative disorder of the skin, suggesting an important role for ET-1 in epithelial proliferation [78]. Furthermore cross-talking between ETAR and EGF-R pathways would have an amplifying effect on cell proliferation [79].

In contrast to the activation of EGF-R, 16E5 can down-modulate the keratinocyte growth factor receptor/fibroblast growth factor receptor 2b (KGF-R/FGF-R2b), through reduction of transcripts and protein with alteration of the receptor endocytic trafficking. This causes a decrease in the growth response to the receptor ligands, suggesting that 16E5 might have a role on HPV infection by perturbing the KGFR-mediated physiological behaviour of confluent keratinocytes committed to differentiation [80]. Thus, 16E5 could exert two opposite effects, both related to virus replication: (a) increasing the growth of the most basal undifferentiated cells by up-regulating the EGF-R pathway and (b) decreasing the physiological proliferation/differentiation of the suprabasal keratinocytes by down-modulation of KGFR expression and signalling.

16E5 can also down-regulate the expression of tumour suppressor p21 and p27, both of which are cyclin-dependent protein kinase inhibitors (CKIs), thus causing cell cycle progression and DNA synthesis (S-phase) (Figure 2). The p21Wafl/Sdil/Cipl down-regulation is at transcriptional level [81] whereas that of p27Kip1, one of the most abundant CKIs, is through reduction of the half-life of p27Kip1 protein [Full size image

Down-regulation of MHC expression

The down-regulation of surface MHC I and retention of the MHC I complex in the GA were first shown in cells expressing BPV-4 E5 [169] and then confirmed for other E5 proteins both from BPV and HPV. BPV E5 interferes with MHC I biosynthesis at multiple steps: transcriptional inhibition of the MHC class I heavy chain gene, degradation of the heavy chain and physical interaction with any residual heavy chain [169, 170]. Only the physical interaction with heavy chain is shared with 16E5 [99]. Moreover, in the case of BPV-4 E5 the down-regulation of surface MHC I is irreversible, as interferon cannot restore the transport of the MHC I complex to the cell surface [128], while this is not the case for 16E5. This may reflect the fact that E5 is a "stronger" protein in BPV than in HPV.

In any event, the finding of MHC down-regulation by E5 has proved of great importance since it defined a new biological activity shared by both BPV and HPV E5 (Table 1), which allows the infected cell not to be effectively recognised by cytotoxic CTL [106].

BPV E5: other biological activities

BPV E5 interacts with the 16 kDa protein, a component of the V-ATPase [147, 171]. In the case of BPV-1 E5, this protein association exists as a tri-component complex, which contains also PDGFβ-R molecules. Mutational analyses have demonstrated that glutamic acid 143 in the transmembrane domain of the 16 kDa protein and glutamine 17 in the transmembrane domain of BPV-1 E5 are essential for E5-16 kDa stable association [172]. The binding of the E5 with 16 kDa perturbs the correct assembly of V-ATPase and H+ pum**, with persistent alkalinization of the GA and endosomal vesicles [103] and may lead, directly or indirectly, to a marked loss of cell-cell communication through down-regulation of gap junctions (Table 1) [101, 173], rendering the transformed cells more insensitive to homeostatic growth control signals from adjacent normal cells. However, in the case of HPV-16 inhibition of gap junction intercellular communication has been attributed to the interaction of E5 with connexin 43 [84]. E5-expressing bovine urinary bladder tumours show activated calpain 3, suggesting a possible involvement of this protein in urothelial carcinogenesis [174].

Targeting HPV-16 E5 for cervical treatment

16E5 activates many different cellular pathways involved mostly in the early stage of cervical carcinogenesis [175]. Different studies have pointed out the possibility of E5 targeting for CaCx therapy. In animal studies, 16E5 delivered by an adenovirus vector reduces the growth of tumours and the E5 vaccine induces protection against tumours through CD8+ cytotoxic T cells (CTLs) [176]. The same research group has further demonstrated that the stimulating epitope was an E5 peptide (aminoacids 25-33) and vaccination with this peptide carrying CpG oligonucleotides reduced tumour growth [177]. 16E5 modulates different cellular pathways and the targeting of these relevant pathways may lead in a near future to a possible therapeutic approach. In addition, E5 is more consistently expressed in the early stage of viral infection and in precancerous lesions [178], and therefore E5 or E5 altered pathways could be targeted to cure the infection and to prevent precancerous lesions from progressing into invasive cancers. 16E5 interacts with many different pathways including upregulation of growth factor signalling, induction of inflammatory cell signalling, angiogenesis and antiapoptosis (Figure 2). Furthermore E5 participates in malignant transformation by supplementing (added value) the roles of E6 and E7. All these E5-induced signalling pathways (Figure 2) or E5 itself can be addressed by therapies already utilised against the other HPV oncogenes such as radioimmunotherapy, oncolytic adenoviruses [179], gene silencing using short interfering RNA (siRNA) [180] and others, which have been originally used to target the E5-upregulated pathways. EGF-R, COX-2 or ET1 inhibitors should be useful in controlling E5 activity in the precancerous lesions as well as other small molecules interfering with downstream molecules. Finally, E5 may be considered a viroporin, like the VP4 of SV40 [181] and, therefore, susceptible of therapy by compounds that affect similar structures produced by other viruses. Indeed amantidine, amiloride, long-alkyl-chain iminosugar derivatives and new compounds seem to affect, to various extent, the activity of the 16E5 protein in vitro (A. Macdonald, personal communication, 11 DNA TV Meeting, Trieste, Italy, 2011)