Open access

The Role of E2 Proteins in Papillomavirus DNA Replication

Written By

Reet Kurg

Submitted: 04 November 2010 Published: 01 August 2011

DOI: 10.5772/19609

From the Edited Volume

DNA Replication - Current Advances

Edited by Herve Seligmann

Chapter metrics overview

6,324 Chapter Downloads

View Full Metrics

1. Introduction

Papillomaviruses are small, nonenveloped, double-stranded DNA viruses, which infect a wide variety of vertebrate species and induce proliferative lesions in their host. The viruses are species-specific and to date, more than 150 different types of papillomaviruses have been identified; each virus infects a specific region of cutaneous or internal mucosal epithelium in its host (McLaughlin-Drubin and Münger, 2009). Papillomaviruses are widely spread in nature and in most cases, papillomavirus infections are cleared by the host immune system, however, sometimes papillomaviruses establish persistent infections. Papillomavirus classification is based on nucleotide sequence homology (de Villiers et al., 2004). Human papillomavirus (HPV) types that infect the genital epithelia belong to subgroup A (alpha-papillomaviruses), and are classified into high- or low-risk types, depending upon their oncogenic potential. Persistent infection with high-risk HPVs (HPV16, 18, 31, 33, 45) can lead to cervical cancer, the second most common cancer in women (Durst et al., 1983; zur Hausen, 2000). Low-risk HPVs, such as HPV6 and 11, can infect genital tract as well as oral sites where they are generally associated with benign papillomas. The second major group of HPVs, supergroup B, also known as beta-papillomaviruses, infect skin epithelia and may develop skin cancers at the site of HPV infection (McLaughlin-Drubin and Münger, 2009).

Advertisement

2. Papillomavirus life cycle

Papillomaviruses, with the help of only few genes, can achieve a complete replication cycle in the epidermal and mucosal keratinocytes. Most viral types are predominantly trophic for one or the other cell types, but certain genotypes can infect and persist in both. Papillomavirus virions enter the epithelial tissue through microwounds and infect a subset of basal cells, probably stem cells, at low copy number (Egawa, 2003). HPV virions migrate to the cell nucleus and establish their genomes as episomes. Next, the HPV early promoter is activated and the viral early proteins, E1 and E2, are transcribed, the synthesis of viral DNA is initiated and the copy number of viral episomes is raised up to 20-100 genomes per cell. During this amplification stage, rapid viral replication is required to quickly reach an optimal copy number. As basal cells divide, HPV genomes are replicated and distributed evenly between daughter cells in mitosis. This is a phase of episomal maintenance with minimal viral gene expression, viral replication proceeds at moderate level and is synchronized to cellular proliferation (De Geest et al., 1993). Papillomavirus persistence is set up through the maintenance of a constant copy number of extrachromosomal viral genomes in the nuclei of dividing host cells. The expression of late genes encoding the capsid proteins and virus assembly are tightly coupled to the differentiation of epithelial tissue (Longworth and Laimins, 2004). When the infected keratinocyte enters the differentiating compartment, exiting the cell cycle, the vegetative phase of the HPV life cycle leading to amplification of the viral genome is initiated. This phase includes amplification of viral copy number to at least 1000 copies per cell and expression of late transcripts encoding proteins for viral capsid. The viral DNA is packed into virions, which are released with dead cells after the infected cells reach the epithelial surface (for review, see Chow et al. 2010; Doorbar, 2005).

The dependence of papillomavirus life cycle on epithelial differentiation and difficulties in reproducing this process in cell culture has complicated the studies of papillomavirus replication. Short-term replication assays closely mimic the initial amplification phase of replication, while long-term replication assays mimic viral maintenance in undifferentiated basal cells. DNA amplification and virion assembly can be analyzed only in differentiating epithelial cells. The most extensively studied papillomavirus is bovine papillomavirus type-1 (BPV-1) because of its capacity to transform rodent cell lines in culture (Law et al., 1981). The mouse fibroblast cell line C127 transfected with the BPV1 genome maintaining the viral DNA as extrachromosomal plasmid with the constant copy number was historically the first model system for studying mechanisms of papillomavirus DNA replication. Our knowledge of initiation of papillomavirus DNA replication and maintenance of virus genomes has primarily derived from studies with BPV1. However, BPV-1 belongs to delta-papillomaviruses and is evolutionary distinct from the human papillomaviruses.

Figure 1.

Schematic representation of epithelia showing various differentiated layers and PV replication, and cell culture models mimicking papillomavirus DNA replication.

The limited availability of appropriate cell culture systems supporting HPV DNA replication has hampered research of HPV replication and regulatory pathways involved in these processes. Primary keratinocytes, transfected with circulized HPV genome, replicate HPV genomes at low copy number. Productive HPV replication needs culture systems where epithelial cells are able to differentiate. Adaptation of organotypic culture techniques, „raft“ culture systems, provide the basis for propagation of HPVs, although the yield of virions is very low (Dollard et al., 1992; McCance et al., 1988; Meyers et al., 1992). Introduction of adenovirus recombinants carrying the HPV genome flanked by loxP sites and Cre recombinase into PHK cells grown in raft cultures results in efficient establishment of cells harboring thousands of copies of the HPV18 genome. Virions obtained from these cultures efficiently infect PHKs at a multiplicity of infection (MOI) of <50, resulting in expression of spliced viral mRNAs (Wang et al., 2009a). However, organotypic culture of HPV-infected cells is not a very convenient system for long-term viral persistence and latency studies. Cell lines from mild dysplasia harboring episomal HPV genomes (W12 and CIN-612) have been very useful tools for studying stable episomal HPV replication, and also loss of episomes and integration of HPV into the host genome, a process accompanied by tumourigenesis (Pett and Coleman, 2007). At early passages in monolayer culture, the W12 cells retain HPV16 episomes at 100 to 200 copies per cell, but in long-term cultivation in the absence of feeder cells, the spontaneous loss of episomes and integration of HPV16 occurs (De Geest et al., 1993; Pett et al., 2004; Stanley et al., 1989). The genomes of both high- and low-risk alpha-papillomaviruses as well as cutaneous beta-papillomaviruses replicate in human osteosarcoma cell line U2OS, and are able to establish persistent replication in these cells. These cell lines might be a valuable and efficient tool for studying fundamental properties of HPV DNA replication, especially the long-term viral persistence, and for the development of inhibitors of HPV genome replication (Geimanen et al., 2011).

2.1. Papillomavirus genome

All papillomaviruses have a similar genomic organization. The papillomavirus genome of approximately 8000 base pairs in length contains a non-coding upstream regulatory region (URR), early (E) region of open reading frames (ORFs) and late (L) region of ORFs. The circular papillomavirus genome encodes roughly eight ORFs from a single DNA strand. The URR alone is 600-900 base pairs in length and contains the origin of replication and binding sites for transcription factors and enhancers (Fig. 2). All transcription takes place in the same direction (clockwise on the circular map) using multiple promoters. The E region and L region are both followed by a poly-A addition site. Within the virions, the papillomavirus genome is associated with cellular histones forming chromatin-like complexes.

Papillomavirus transcription is complex and involves the usage of different promoters, multiple splice patterns and differential production of mRNA species in different cells. In BPV1, seven different promoters, six of which being active in undifferentiated cells, have been identified (Ahola et al., 1987; Baker and Howley, 1987). In the high-risk HPV types, transcripts are initiated at two major viral promoters, one for early and the other for late transcripts. In contrast, early genes of low-risk HPVs are expressed from two independent promoters (Chow et al., 1987). The HPV early promoter (p105 in case of HPV18) is activated by binding of cellular transcription factors; however the factors that determine the cell specificity are still not fully understood. Among the main players involved in PV replication are the ubiquitous transcription factors Sp1 and AP1 (Gloss and Bernard, 1990; Hubert et al., 1999; Thierry, 2009; Thierry et al., 1992). In addition to the binding sites for the cellular transcription factors, the URR contains binding sites for the virally encoded E2 regulatory proteins and E1 helicase. Early region proteins, E6, E7, E1 and E2 are transcribed from the HPV early promoter active in basal, nonproductively infected cells and in transformed cells, and all four early proteins are translated from the same polycistronic mRNA (Hummel et al., 1992). E1 and E2 are directly involved in viral DNA replication; the other early proteins - oncoproteins E6 and E7 - support viral DNA amplification indirectly by inactivating major tumor suppressor proteins and activating signal transduction pathways (for review, see (McLaughlin-Drubin and Münger, 2009; Moody and Laimins, 2010).

Figure 2.

Organization of the HPV18 circular genome showing the cis-sequences and viral proteins required for initiation of papillomavirus DNA replication. Open reading frames are indicated by dark grey (early region) and light grey (late region) boxes. The origin of replication, early promoter (p105) and E2 binding sites (boxes with numbers) are shown on the scheme of URR. Papillomavirus E1 and E2 proteins, transcribed from the early promoter, form a complex and bind cooperatively to their binding sites within the origin of replication locating in the URR.

Advertisement

3. Papillomavirus DNA replication

3.1. Initiation of replication

The double-stranded circular DNA replicates as a multi-copy extrachromosomal plasmid in the nucleus of infected cells. The replication of papillomavirus DNA is initiated from the origin of replication consisting of binding sites for E1 and E2 proteins (Del Vecchio et al., 1992; Ustav et al., 1993; Ustav et al., 1991). Transient, short-term replication requires the origin of replication in cis and the expression of viral E1 and E2 proteins, all other replication enzymes and proteins are supplied by the host cells (Remm et al., 1992; Ustav and Stenlund, 1991). Papillomavirus DNA replication is not cell-type specific, despite of high degrees of host and cell-type specificity for infection. The BPV1 genome can replicate and is maintained in murine C127 cells. HPV11 and HPV18 genomic DNA can replicate not only in its natural host cells, primary foreskin keratinocytes, but also in the human tongue squamous cell carcinoma cell line SCC4 and osteosarcoma cell line U2OS (Del Vecchio et al., 1992; Geimanen et al., 2011; Mungal et al., 1992). Transient replication of BPV1 and HPV origin-containing plasmids can take place in a number of mammalian cells if E1 and E2 proteins are provided from heterologous expression vectors (Chiang et al., 1992). HPV transcription exhibits stringent cell specificity and the lack of regulated gene expression of viral early proteins from HPV genomes could be the reason that restricts HPV genome replication to certain cell types. BPV1 DNA replication in vitro with purified proteins and cell extracts from murine, simian and human cells has been reconstituted showing that all cellular factors essential for papillomavirus DNA replication are provided by the host cell (Liu et al., 1995; Muller et al., 1994; Yang et al., 1991a). The cellular replication factors required for papillomavirus DNA replication include replication protein A (RPA), a single-stranded-DNA-binding protein, replication factor C (RFC), proliferating-cell nuclear antigen (PCNA) and DNA polymerase α–primase. It has been shown that E1 recruits Topo I to the papillomavirus replication fork and Topo I specifically stimulates the origin binding activity of papillomavirus E1 protein (Clower et al., 2006; Hu et al., 2006; Melendy et al., 1995; Park et al., 1994).

3.2. Origin of replication

The papillomavirus origin of replication consists of three E2 binding sites (E2BS), from which only one is absolutely required for replication, and an A/T-rich region containing an array of E1 protein binding sites (E1BS) (Remm et al., 1992; Ustav et al., 1991). The overall structure of BPV1 and HPV origins is conserved and various combinations of E1 and E2 proteins from different papillomaviruses can initiate DNA replication from several papillomavirus origins (Chiang et al., 1992; Del Vecchio et al., 1992; Kadaja et al., 2007). A relationship appears to exist between the affinity of the E2BS and the ability to function at a distance from the binding site for the E1 protein. A low-affinity site appears to be functional only when located close to the E1 binding site, while for function at greater distances higher affinities are required. In multimerized form, the high affinity E2BSs are able to function even when placed at a distance of one kilobase from the rest of the origin (Ustav et al., 1993).

High-risk HPV genomes have been found integrated into host chromatin in tumors and cervix dysplasia biopsy specimens of patients (Dall et al., 2008; Kristiansen et al., 1994). Integration of the HPV genome is often accompanied by the disruption of E2 open reading frame and the loss of E2 activity, leading to enhanced expression of virus oncogenes E6 and E7. The HPV regulatory sequences are active in cervical cancer cells as expression of E7 promotes cell survival (Jiang and Milner, 2002). In patients, the HPV16 genome may exist at the same time in episomal as well as in an integrated form. Co-existence of the replicating episomal viral genome expressing the viral E2 protein, and integrated HPV with viral replication origin raises the question about the functionality of the integrated origins. Recent works have shown that expression of E1 and E2 proteins from expression vectors or from different HPV genomes can induce replication of the genomic integrated HPV origin. The replication forks initiated at the integrated HPV origins extend into the flanking regions of cellular DNA, and these amplified genomic sequences could be targets for the recombination and repair system. This suggests that replication induced from the papillomavirus integrated origin may induce genomic changes of the host cell (Kadaja et al., 2009; Kadaja et al., 2007).

Figure 3.

Origin of replication of BPV1 and HPV18. E1 binding sites are depicted with arrows and E2 binding sites with boxes. The minimal origin of replication consisting of E1BS and E2BSs is shown for BPV1 and HPV18, and the minichromosome maintenance element MME, required for maintenance and segregation of viral DNA, for BPV1.

3.3. Assembly of the DNA replication initiation complex

E1 is an initiator protein of papillomavirus DNA replication. E1 is responsible for recognition of the replication origin, melting of the DNA at the origin as well as for subsequent unwinding of the double helix during progression of the replication fork. The viral E1 protein is an ATP-dependent helicase which binds as a dimer to pairs of its binding sites (Yang et al., 1993). The binding sites for the E1 replication helicase are short sequences, 5-6 base pairs in length, arranged as two pairs of inverted repeats (Chen and Stenlund, 1998; Chen and Stenlund, 2001). E1 has low sequence specificity and therefore it can initiate DNA replication in vitro, and from non-specific DNA sequences (Bonne-Andrea et al., 1995). Within the cells, the E1 and E2 proteins form a complex through multiple protein-protein interactions and bind cooperatively to adjacent binding sites in the origin of replication (Berg and Stenlund, 1997; Mohr et al., 1990). E1 by itself binds to the origin with low degree of sequence specificity, but in the presence of E2 the sequence specificity is increased (Sedman and Stenlund, 1995; Sedman et al., 1997). In this process, E2 functions transiently and “catalytically”, providing sequence specificity for the formation of the E1-ori complex. In addition, E2 enhances E1 binding to DNA through the DNA-binding domain by inhibiting the non-specific DNA binding of the E1 helicase domain (Stenlund, 2003a). The E1-E2-ori complex is able to bind DNA with high specificity but lacks other biochemical activities. In the next step, additional E1 molecules are added by displacing E2 from the DNA-bound complex in an ATP-dependent manner (Sanders and Stenlund, 1998; Sanders and Stenlund, 2000). Two additional E1 molecules are recruited to the origin, which results in the formation of two E1 trimers on the ori, followed by formation of two hexamers in the presence of ATP. E1 hexameric complex has the DNA helicase activity which is able to unwind the DNA and initiate the papillomavirus DNA replication (Fouts et al., 1999; Schuck and Stenlund, 2005; Sedman and Stenlund, 1998).

Figure 4.

Assembly of the replication initiation complex. The E1 and E2 proteins bind cooperatively to the origin of replication, which contains binding sites for both proteins (E1BS and E2BS). The resulting E1:E2 complex binds DNA with high affinity and specificity. In the next step, additional E1 molecules are recruited to the complex and E2 is displaced in the presence of ATP. ATP hydrolysis is also required for conversion of E1 complex into a double hexamer possessing DNA helicase activity (for review, see Stenlund, 2003b).

3.4. Stable maintenance replication

Papillomaviruses have the capacity to establish a persistent infection in mammalian epithelial cells. The BPV1-transformed C127 cells maintain the viral genome as a multicopy nuclear plasmid thus being a valuable tissue culture system for investigating the establishment and maintenance of papillomavirus genomic DNA. The studies with BPV1-C127 cells have revealed that during the maintenance of virus genomes BPV1 DNA is replicating by a conventional bi-directional (theta-type) replication mode throughout S phase of the cell cycle in a random-choice fashion (Gilbert and Cohen, 1987; Ravnan et al., 1992). Some molecules replicate once per cell cycle, some replicate more than once, and others do not replicate at all during a given cell cycle, resulting in statistically “once per cell cycle” replication of the viral DNA. Stable HPV DNA replication seems to be more complicated. This has been studied in human keratinocytes, where the HPV DNA is stably maintained at high copy number over several passages. HPV16 and HPV31 DNA can replicate randomly or in an ordered once-per-S-phase fashion depending on the cell line in which it is located. In W12 cells, which are derived from a natural cervical lesion, the HPV16 DNA replicates only once per S phase, but in another immortalized keratinocyte cell line, NIKS, it replicates randomly (Hoffmann et al., 2006). At later stages of the papillomavirus life cycle, there is a shift from the theta replication mode in the proliferating keratinocytes to the rolling-circle replication mode after the cells start to differentiate (Flores and Lambert, 1997).

Studies of subclones of BPV1-transformed C127 cells as well as U2OS cells maintaining HPV genomes have demonstrated cell to cell variation in the extent and state of genomic DNA. In addition to the monomeric form of genomic DNA, dimeric and sometimes higher oligomeric forms of BPV1 and mucosal and cutaneous HPV genomes are detected. Oligomeric forms of papillomavirus DNA are organized in a head-to-tail configuration and replication is initiated at only some of the origins (Geimanen et al., 2011; Schvartzman et al., 1990). In patient samples, HPV16 DNA, which is maintained in carcinoma cells as episome, is always multimeric, suggesting that oligomerization of HPV genomes is common during viral infections in vivo (Cullen et al., 1991; Kristiansen et al., 1994).

It is widely accepted that the E1 and E2 proteins are essential for long-term stable replication. However, some studies suggest that E1 is necessary to establish the viral genome as a nuclear plasmid, but is not required at the maintenance stage. The replication of the BPV1 genome containing temperature-sensitive mutation in the E1 gene was first initiated at the permissive temperature and then switched to the non-permissive temperature, where ts-E1 genomes were found to persist as nuclear plasmids for multiple cell generations (Kim and Lambert, 2002).

The functions of viral oncoproteins E6 and E7 are also essential for the maintenance of the extrachromosomal forms of HPV DNA (Thomas et al., 1999). Both E6 and E7 stimulate cell cycle progression and may create a cellular environment permissive to HPV maintenance and abrogate the checkpoints that would block the long-term retention of viral DNA (Garner-Hamrick et al., 2004). Oncogene expression in basal epithelial cells is shown to inhibit cellular differentiation thus promoting long-term persistence of HPV episomes (Hudson et al., 1990; Jones et al., 1997; Sherman and Schlegel, 1996). The E6 and E7 proteins of high-risk HPV types act as viral oncogenes. E6 binds the p53 tumor suppressor protein, which regulates the expression of proteins involved in cell cycle control, leading to degradation of p53 (Scheffner et al., 1990; Werness et al., 1990). Another important function of the high-risk E6 protein is the activation of telomerase in infected cells. High-risk HPV E6 has been shown to increase telomeric length by activating the catalytic subunit of the telomerase hTERT. This extends the life of epithelial cells containing HPV genomes (Oh et al, 2001; Stoppler et al., 1997). The function of the high-risk E7 protein is the binding and degradation of the RB family proteins, which are the major regulators of the cell cycle (Boyer et al., 1996; Schmitt et al., 1994). E7 interaction with histone deacetylases HDACs plays also an essential role in the viral life cycle, as cells harboring HPV genomes with mutations abolishing the E7-HDAC interaction display slower growth and a loss of episomal maintenance (Longworth and Laimins, 2004a). Integration of high-risk HPV into the host genome and loss of E2 expression leads to constitutive activation of the viral oncogenes. Reintroduction of E2 into HPV-associated cervical carcinoma cells, resulting in reactivation of p53 and pRB pathways, has shown to suppress cellular growth, because of cell cycle arrest in G1, apoptosis and senescence (Desaintes et al., 1997; Goodwin and DiMaio, 2000; Goodwin et al., 2000; Hwang et al., 1996; Thierry et al., 2004; Wells et al., 2000).

Advertisement

4. E2 as a viral regulatory protein

The proteins encoded by the papillomavirus E2 ORF play a crucial role in the viral life cycle and serve as major viral regulators of transcription, replication and segregation of the viral genome in the infected cells. The E2 protein is a modular protein consisting of three different structural as well as functional domains; the N-terminal transactivation domain (TAD) (aa 1-200), the C-terminal DNA-binding dimerization domain (DBD) (aa 310-410), and flexible unstructured “hinge region” which functions as a linker between the two domains. The carboxy-terminal DBD binds as a dimer to consensus sequence ACCN6GGT (Androphy et al., 1987) and the N-terminal activation domain is required for the replication, transactivation and segregation function of the protein (Abroi et al., 2004; Bastien and McBride, 2000; McBride et al., 1989; Ustav and Stenlund, 1991). The three-dimensional structures of the C-terminal DBD and N-terminal transactivation domain of several E2 proteins have been reported revealing a tight dimer of the DBD bound to DNA and a L-shape structure of activation domain (Antson et al., 2000; Harris and Botchan, 1999; Hegde et al., 1992). The papillomavirus E2 proteins can function as activators or repressors of transcription depending on the concentration of proteins within the cell. At low concentrations, E2 activates, and at high concentrations, suppresses transcription from homologous and heterologous promoters containing E2 binding sites (Abroi et al., 1996; Schweiger et al., 2007). E2 has been demonstrated to be a transcriptional activator of early genes in BPV1 and a repressor of early genes in case of HPV16 (Soeda et al., 2006; Spalholz et al, 1985; Thierry and Yaniv, 1987).

E2 is a multifunctional protein. The transactivation domain of E2 is responsible for interactions with viral helicase E1 and with several cellular proteins. The transactivation and replication functions of E2 are separable by point-mutations in the N-terminal activation domain (Abroi et al., 1996; Brokaw et al., 1996; Ferguson and Botchan, 1996; Grossel et al., 1996). The X-ray crystal structures of a complex containing the activation domain of E2 and the helicase domain of E1, and the activation domain of E2 together with Brd4 have been solved and confirm that E2 interacts with E1 and Brd4 through different interaction surfaces (Abbate et al., 2004; Abbate et al., 2006). The cellular bromodomain protein Brd4 is the major cellular partner for E2, and interaction with Brd4 is crucial for both E2 transactivation and repression functions (Ilves et al., 2006; McPhillips et al., 2006; Schweiger et al., 2006). Brd4 is a component of the HPV11 E2 transcriptional silencing complex involved in repression of the E6/E7 promoter (Wu et al., 2006). Through the interaction of Brd4 and transactivation domain, E2 is associated with transcriptionally active cellular chromatin. This association is driving the E2-mediated tethering of viral genomes to host chromatin, and at the same time is retaining the viral genomes in transcriptionally active regions of the nucleus to escape silencing (Jang et al., 2009; Kurg et al., 2005). The transactivation domain mediates functional interactions with cellular transcription factors Sp1 and AP1, histone acetylase complexes containing CBP/p300 and pCAF, and with nucleosome assembly protein hNAP1 (Lee et al., 2002a; Lee et al., 2000; Li et al., 1991; Müller et al., 2002; Rehtanz et al., 2004; Thierry et al., 1992). E2-dependent activities are also modulated by interactions with Brm, a chromatin remodeling protein associated with SWI/SNF ATP-dependent chromatin remodeling complex, and with EP400, a component of the NuA4/TIP60 histone acetylase complex, and SMCX, also known as histone demethylase JARID1C, and Tax1BP1 (Kumar et al., 2007; Smith et al., 2010; Wang et al., 2009b).

E2 binds to the papillomavirus oncoproteins E6 and E7, leading to the modulation of their functions (Gammoh et al., 2006; Grm et al., 2005). E2 has activities that inhibit cell growth. High-risk, but not low-risk HPV E2 proteins can induce themselves growth arrest and apoptotic cell death in several HPV-negative carcinoma cell lines. Apoptosis can occur via a p53-dependent as well as independent pathways (Demeret et al., 2003; Parish et al., 2006b). E2 binds to the cellular protein p53. Expression of p53 can inhibit papillomavirus DNA replication and alter the transcriptional activity of E2 (Frattini et al., 1997; Lepik et al., 1998; Massimi et al., 1999). Interestingly, p53 inhibits the initial, amplificational replication, but not the stable, long-term replication of BPV1 (Ilves et al., 2003). E2 can modulate the activity of cellular proteins, like activators of the Anaphase Promoting Complex (APC) Cdh1 and Cdc20 inducing genomic instability (Bellanger et al., 2005). E2 is associated with transcriptionally active chromatin and as a transcription factor may directly regulate the expression of cellular genes. The HPV E2 proteins are reported to repress the hTERT promoter activity (Lee et al., 2002b). HPV8 E2 protein suppresses β4-integrin expression and HPV16 E2 protein transcriptionally activates the promoter of a key cellular splicing factor SF2/ASF (Mole et al., 2009; Oldak et al., 2004). The expression of HPV8 E2 protein in transgenic mice can induce the formation of skin tumors through an unknown mechanism (Pfefferle et al., 2008).

The E2 proteins bind to inverted repeats with consensus sequence 5’ ACCGN4CGGT, where N4 represents a 4 bp central sequence (Li et al., 1989). Although the base pairs in the central spacer sequence are not in contact with the protein, they affect protein binding. The E2 DNA binding is accompanied by bending of DNA and depends on DNA flexibility (Hines et al., 1998). HPV E2 proteins bind with higher affinity to sites with A/T-rich central spacer while BPV1 E2 has no preference (Dell et al., 2003). E2 binding can be inhibited by CpG methylation of the E2 binding site (Kim et al., 2003; Thain et al., 1996). The BPV1 genome contains 17 E2 binding sites from which 12 sites are located within the URR region. In genital HPV genomes, there are four E2 binding sites with conserved sequences and positions. E2 remains associated with the BPV1 URR throughout the cell cycle including mitosis (Melanson and Androphy, 2009).

Figure 5.

Papillomavirus genomes encode multiple E2 proteins. In addition to the full-length E2 protein, BPV1 and HPV18 genomes encode truncated E2 proteins, which lack the activation domain and serve as repressors of replication and transcription. The full-length and truncated E2 proteins are able to form dimers through their common C-terminal DNA-binding-dimerization domain.

4.1. E2 repressors and heterodimers

In addition to the full-length E2 protein, the BPV-1 and mucosal HPV genomes encode truncated E2 proteins, which lack the activation domain, but maintain the DNA-binding-dimerization domain (DBD). For BPV1, mRNA for E2C is transcribed from a promoter internal to the open reading frame, and E8/E2 is created by splicing E8 ORF sequences to an acceptor located within the E2 ORF. The repressor proteins encoded by HPVs are similar to the BPV1 E8/E2 protein, since they contain a small conserved E8 ORF (HPV 11, 18, 31) or fragment of E1 ORF (HPV 11) fused to the C-terminus of E2. Transient over-expression assays have suggested that shorter E2 proteins act as negative regulators of E2 and function as repressors of transcription and replication (Chiang et al., 1991; Doorbar et al., 1990; Hubbert et al., 1988; Kurg et al., 2010; Lim et al., 1998; Stubenrauch et al., 2000). The shorter E2s antagonize the function of full-length E2 by competing for E2 DNA binding sites. In addition, the E8 of HPV 31 E8/E2 protein itself is a transcriptional repressor domain that functions independently of binding site competition inhibiting transcription and DNA replication by interacting with co-repressor molecules such as NCoR1/HDAC3, the histone methyltransferase SETDB1, and the TRIM28 protein (Ammermann et al., 2008; Powell et al., 2010).

The full-length and truncated E2 proteins are able to form dimers through their common C-terminal DNA-binding-dimerization domain (McBride et al., 1989). E2 heterodimers with single activation domain bind DNA sequence-specifically and serve as activators of transcription and replication in cell culture model systems (Kurg et al., 2006). E2 heterodimers can interact with viral helicase E1, and are able to recruit E1 to the origin of replication and activate the papillomavirus DNA replication in a cell-free system (Lim et al., 1998). Replacing the open reading frame of E2 with “single-chain” E2 in the context of BPV1 and HPV18 genome revealed that E2 heterodimer with single activation domain could support transient, but not long-term, replication in cell culture model systems (Kurg et al., 2009; Kurg et al., 2010). The full-length E2 protein is required for long-term papillomavirus DNA replication, the E2 heterodimer with single activation domain is crippled in this function. E2 requires two activation domains for interaction with Brd4, the cellular receptor for BPV1 E2 on mitotic chromosomes. Brd4 interacts efficiently with the BPV1 homodimer with two activation domains and with low affinity with the E2 heterodimer with single activation domain and with E2 mutants unable to form dimers between N-terminal activation domains (Cardenas-Mora et al., 2008; Kurg et al., 2006; You et al., 2004).

4.2. The role of E2 in initiation of DNA replication

The expression of E2 protein is required for initiation of papillomavirus DNA replication. The role of E2 in initiation of viral DNA replication is relatively well understood, E2 helps to recruit the viral helicase E1 to the viral replication origin by direct protein-protein and protein-DNA interactions as discussed above. The initiation step and interactions mediating the formation of the replication initiation complex are well studied and are conserved between BPV1 and alpha-papillomaviruses.

4.3. The role of E2 in stable maintenance

Segregation of papillomavirus genomic DNA is achieved through its attachment to mitotic host chromosomes during cell division. Non-covalent association of viral DNA with chromosomes is a general mechanism used by all papillomaviruses studied so far. This mechanism ensures that the replicated virus episomes are retained inside the nuclei of dividing host cells and faithfully partitioned to the daughter cells during mitosis (You, 2010).

Long-term replication and maintenance of BPV1 episomes requires the sequence of URR - the minichromosome maintenance element (MME) - consisting of at least six E2 binding sites and the minimal origin of replication as cis-elements. The plasmids containing BPV1 URR can be maintained as extrachromosomal elements in hamster CHO cells stably expressing the viral E1 and E2 proteins (Piirsoo et al., 1996). Extrachromosomal MME-containing plasmids containing ten oligomerized E2 binding sites segregate efficiently between daughter cells in the presence of E2 protein expressed from the same plasmid (Abroi et al., 2004; Silla et al., 2005). MME consisting of E2 binding sites and the E2 protein are responsible for anchoring of BPV1 genomes as well as URR reporter plasmids to host cell chromosomes (Ilves et al., 1999; Lehman and Botchan, 1998; Skiadopoulos and McBride, 1998). The functional organization of the HPV URR is significantly different from that of the BPV1. Alpha-papillomavirus genomes have four E2 binding sites in this region and only three of them are required for stable replication (Stubenrauch et al., 1998). The exact mechanism of segregation of HPV genomes is not yet known and needs further investigations.

Figure 6.

Papillomaviruses establish persistent infection by maintaining viral genomes as episomes in host cell. (A) During mitosis, papillomavirus genomes are associated with cellular mitotic chromosomes. (B) For many papillomaviruses, association with mitotic chromosomes is mediated by the viral E2 protein and cellular bromodomain protein Brd4.

The role of E2 in long-term stable replication is not yet fully understood. The maintenance of papillomavirus genomes during latency is achieved by tethering viral genomes to host the mitotic apparatus in dividing cells. In this, BPV1 and HPVs may use different targets to achieve their goal. In BPV1, the activation domain of E2 is attached to chromosomes and the DNA-binding-dimerization domain tethers viral genomes to achieve their accurate segregation during mitosis. The point-mutations in the activation domain of E2, disrupting the transcription activity of E2, affect the chromatin attachment, suggesting that this activity is required for efficient segregation and maintenance of MME-containing plasmids (Abroi et al., 2004). The cellular receptor for BPV1 E2 on mitotic chromosomes is the bromodomain protein Brd4 (You et al., 2004). However, E2 proteins from different papillomaviruses interact with Brd4 with different affinities. E2 proteins of alpha-papillomaviruses interact with Brd4 weakly and do not co-localize with Brd4 on host mitotic chromosomes, suggesting that HPV E2 proteins may use different cellular targets for tethering virus genomes to host chromosomes (McPhillips et al., 2005; Oliveira et al., 2006). DNA helicase ChR1, a member of the mammalian cohesion complex, is a candidate partner for cellular receptor of HPV E2. ChlR1 and E2 co-localize at early stages of mitosis, however, during prometaphase, ChlR1 is localized to the spindle poles, suggesting that ChlR1 may help to load the papillomavirus E2 protein onto mitotic chromosomes during early mitosis (Parish et al., 2006a). HPV16 E2 co-localizes with TopBP1, a cellular protein involved in DNA damage response, on chromatin and centrosomes during late telophase, suggesting that TopBP1 could be the mitotic chromatin receptor for HPV16 E2 (Donaldson et al., 2007). On the other hand, E2 proteins of HPV11, HPV16 and HPV18 have been found to localize to centrosomes and mitotic spindles during cell division (Van Tine et al., 2004). MKlp2, mitotic kinesin-like protein 2, a kinesin-like motor protein of the central mitotic spindle, binds and co-localizes with papillomavirus E2 during mitosis (Yu et al., 2007). The beta-papillomavirus HPV8 E2 protein binds to the repeated ribosomal DNA loci that are found on the short arm of human acrocentric chromosomes. These speckles do not contain Brd4, the E2 protein co-localizes with UBF, the RNA polymerase I transcription factor (Poddar et al., 2009). A recent study using chimeric BPV1 E2 proteins has shown that attachment of the protein to chromatin is not sufficient for proper segregation. Successful partitioning of virus genomes during cell division is determined by effective formation of the segregation-competent complex which does not necessarily involve the Brd4 protein (Silla et al., 2010).

4.4. Regulation of replication by E2

The relative abundance of E2 proteins within the cell is an important factor regulating papillomavirus DNA replication. In BPV1-transformed cells, the relative ratio of E2 proteins is 1:5:1.5 for E2-E2C-E8/E2. The truncated E2 repressor proteins predominate in the steady state and the E2 heterodimers with single activation domain formed between the full-length and truncated E2 proteins are the preferential form for E2 (Kurg et al., 2006). The promoters for full-length E2 as well as for repressors are themselves controlled and differently regulated by E2, and furthermore, the ratio of the repressors to activators changes throughout the cell cycle. In G1 cells, the repressors dominate, but in late S phase and G2/M, the activator is present at about equal levels to that of the repressors. So, the level of E2 activators and repressors as well as E2 homo- and heterodimers is changing suggesting that the balance of different E2 proteins is a key event in the regulation of papillomavirus DNA replication (Szymanski and Stenlund, 1991; Yang et al., 1991b).

In papillomaviruses, there is a link between transcription and replication control, the protein that binds specifically to origin of replication also functions in control of transcription. BPV1 E2 protein levels are regulated by E2 itself and E2 activators and/or repressors have a positive or negative feedback to virus DNA replication. Initially the level of E2 activators is high to facilitate DNA amplification after infection, but later, the E2 expression is strictly controlled to avoid over-replication. Genetic studies have shown that the E8/E2 protein of HPV18, 31 and at least one of the BPV1 repressors are required for the long-term maintenance of virus episomes, demonstrating the important role of E2 repressors in the viral life cycle. Deletion of the E8 ORF results in robust initial replication of HPV genomes followed by rapid loss of virus episomes from dividing cells. The role of E2 repressors in the virus life cycle is to modulate the activity of full-length E2 protein by preventing the E2 binding to E2BS via binding site competition, and by acting as a repressor recruiting host co-repressor molecules (Kurg et al., 2010; Lambert et al., 1990; Stubenrauch et al., 2000). However, the persistent replication and maintenance of virus genomes is not affected in E8 knock-out genomes of HPV16 and cotton-tail rabbit papillomavirus (CRPV), suggesting that E2 proteins of different papillomaviruses may regulate their expression and through this replication by different ways (Jeckel et al., 2003; Lace et al., 2008).

The efficiency of papillomavirus DNA replication is also controlled by the level of the E1 protein. E2 regulates E1 expression level through modulation of the activity of viral early promoters (Hubert and Laimins, 2002; Szymanski and Stenlund, 1991). However, it is still not clear to what extent HPV E2 proteins regulate HPV URRs. The HPV16 E2 protein does not repress HPV16 transcription when the URR is contained within an episomal HPV genome (Bechtold et al., 2003). Another group has shown that transcription activation function of the HPV31 E2 protein is not required for the viral life cycle (Stubenrauch et al., 1998). In addition to the transcriptional regulation by E2, the expression of E1 is regulated post-transcriptionally by mRNA splicing. In high-risk HPVs, E1 is translated by a discontinuous scanning mechanism and mRNA splicing within the E6 ORF is required for efficient expression of E1 (Hubert and Laimins, 2002)(Remm et al., 1999).

Advertisement

5. Conclusion

E2 is the master regulator of extrachromosomal replication of papillomaviruses. E2 regulates papillomavirus replication at multiple levels and through different mechanisms. First, E2 is essential for initiation of papillomavirus DNA replication. Second, E2 is required for long-term stable replication and is involved in maintenance and segregation of viral genomic DNA. Third, the abundance of E2 proteins and formation of homo- and heterodimers possessing different activities as well as the expression level of viral helicase E1 is regulated by E2. In addition to direct involvement in replication, E2 indirectly regulates papillomavirus replication through modulation of expression and activity of viral oncogenes E6 and E7, and the cellular environment.

Advertisement

Acknowledgments

I thank members of the Ustav lab for stimulating discussions and I am very grateful to Ivar Ilves for valuable comments. RK is supported by the Estonian Science Foundation (ETF7556) and by the European Regional Development Fund through the Center of Excellence in Chemical Biology.

References

  1. 1. AbbateE. A.BergerJ. M.BotchanM. R. 2004 The X-ray structure of the papillomavirus helicase in complex with its molecular matchmaker E2. Genes Dev 18(16), 1981 EOF96 EOF .
  2. 2. AbbateE. A.VoitenleitnerC.BotchanM. R. 2006 Structure of the papillomavirus DNA-tethering complex E2:Brd4 and a peptide that ablates HPV chromosomal association. Mol Cell 24(6), 877 EOF889 EOF .
  3. 3. AbroiA.IlvesI.KiviS.UstavM. 2004 Analysis of chromatin attachment and partitioning functions of the bovine papillomavirus type 1 E2 protein. J.Virol. 78 21002113 .
  4. 4. AbroiA.KurgR.UstavM. 1996 Transcriptional and replicational activation functions in the bovine papillomavirus type 1 E2 protein are encoded by different structural determinants. J Virol 70(9), 6169-6179.
  5. 5. AholaH.StenlundA.Moreno-LopezJ.PetterssonU. 1987 Promoters and processing sites within the transforming region of bovine papillomavirus type 1. J Virol 61(7), 2240-2244.
  6. 6. AmmermannI.BrucknerM.MatthesF.IftnerT.StubenrauchF. 2008 Inhibition of transcription and DNA replication by the papillomavirus E8 -E2C protein is mediated by interaction with corepressor molecules. J Virol 82(11), 5127-5136.
  7. 7. AndrophyE.LowyD.SchillerJ. 1987 Bovine papillomavirus E2 trans-activating gene product binds to specific sites in papillomavirus DNA. Nature 325(6099), 70 EOF3 EOF .
  8. 8. AntsonA. A.BurnsJ. E.MorozO. V.ScottD. J.SandersC. M.BronsteinI. B.DodsonG. G.WilsonK. S.MaitlandN. J. 2000 Structure of the intact transactivation domain of the human papillomavirus E2 protein. Nature 403(6771), 805 EOF9 EOF .
  9. 9. BakerC.HowleyP. 1987 Differential promoter utilization by the bovine papillomavirus in transformed cells and productively infected wart tissues. EMBO J 6(4), 1027-1035.
  10. 10. BastienN.Mc BrideA. 2000 Interaction of the papillomavirus E2 protein with mitotic chromosomes. Virology 270 124134 .
  11. 11. BechtoldV.BeardP.RajK. 2003 Human papillomavirus type 16 E2 protein has no effect on transcription from episomal viral DNA. J Virol 77(3), 2021 EOF8 EOF .
  12. 12. BellangerS.BlachonS.MechaliF.Bonne-AndreaC.ThierryF. 2005 High-risk but not low-risk HPV E2 proteins bind to the APC activators Cdh1 and Cdc20 and cause genomic instability. Cell Cycle 4(11), 1608 EOF1615 EOF .
  13. 13. BergM.StenlundA. 1997 Functional interactions between papillomavirus E1 and E2 proteins. J Virol 71(5), 3853-3863.
  14. 14. Bonne-AndreaC.SantucciS.ClertantP. 1995 Bovine papillomavirus E1 protein can, by itself, efficiently drive multiple rounds of DNA synthesis in vitro. J Virol 69(5), 3201 EOF5 EOF .
  15. 15. BoyerS. N.WazerD. E.BandV. (1996). E7 protein of human papilloma virus-16 induces degradation of retinoblastoma protein through the ubiquitin-proteasome pathway. Cancer Res 56(20), 4620-4624.
  16. 16. BrokawJ.BlancoM.Mc BrideA. 1996 Amino acids critical for the functions of the bovine papillomavirus type 1 E2 transactivator. J Virol 70(1), 23 EOF9 EOF .
  17. 17. Cardenas-MoraJ.SpindlerJ. E.JangM. K.Mc BrideA. A. 2008 Dimerization of the papillomavirus E2 protein is required for efficient mitotic chromosome association and Brd4 binding. J Virol 82(15), 7298-7305.
  18. 18. ChenG.StenlundA. 1998 Characterization of the DNA-binding domain of the bovine papillomavirus replication initiator E1. J Virol 72(4), 2567 EOF76 EOF .
  19. 19. ChenG.StenlundA. 2001 The E1 initiator recognizes multiple overlapping sites in the papillomavirus origin of DNA replication. J Virol 75(1), 292-302.
  20. 20. ChiangC.BrokerT.ChowL. 1991 An E1M--E2C fusion protein encoded by human papillomavirus type 11 is a sequence-specific transcription repressor. J Virol 65(6), 3317-3329.
  21. 21. ChiangC.UstavM.StenlundA.HoT.BrokerT.ChowL. 1992 Viral E1 and E2 proteins support replication of homologous and heterologous papillomaviral origins. Proc Natl Acad Sci U S A 89(13), 5799-5803.
  22. 22. ChowL.NasseriM.WolinskyS.BrokerT. 1987 Human papillomavirus types 6 and 11 mRNAs from genital condylomata acuminata. J Virol 61(8), 2581-2588.
  23. 23. ChowL. T.BrokerT. R.SteinbergB. M. 2010 The natural history of human papillomavirus infections of the mucosal epithelia. Apmis 118(6-7), 422 EOF449 EOF .
  24. 24. ClowerR. V.FiskJ. C.MelendyT. 2006 Papillomavirus E1 protein binds to and stimulates human topoisomerase I. J Virol 80(3), 1584-1587.
  25. 25. CullenA. P.ReidR.CampionM.LorinczA. T. 1991 Analysis of the physical state of different human papillomavirus DNAs in intraepithelial and invasive cervical neoplasm. J Virol 65(2), 606-612.
  26. 26. DallK. L.ScarpiniC. G.RobertsI.WinderD. M.StanleyM. A.MuralidharB.HerdmanM. T.PettM. R.ColemanN. 2008 Characterization of naturally occurring HPV16 integration sites isolated from cervical keratinocytes under noncompetitive conditions. Cancer Res 68(20), 8249 EOF8259 EOF .
  27. 27. De GeestK.TurykM. E.HoskenM. I.HudsonJ. B.LaiminsL. A.WilbanksG. D. 1993 Growth and differentiation of human papillomavirus type 31b positive human cervical cell lines. Gynecol Oncol 49(3), 303 EOF310 EOF .
  28. 28. de VilliersE. M.FauquetC.BrokerT. R.BernardH. U.zurHausen. H. 2004 Classification of papillomaviruses. Virology 324(1), 17-27.
  29. 29. Del VecchioA.RomanczukH.HowleyP.BakerC. 1992 Transient replication of human papillomavirus DNAs. J Virol 66(10), 5949-5958.
  30. 30. DellG.WilkinsonK. W.TranterR.ParishJ.LeoBrady. R.GastonK. 2003 Comparison of the structure and DNA-binding properties of the E2 proteins from an oncogenic and a non-oncogenic human papillomavirus. J Mol Biol 334(5), 979-991.
  31. 31. DemeretC.Garcia-CarrancaA.ThierryF. 2003 Transcription-independent triggering of the extrinsic pathway of apoptosis by human papillomavirus 18 E2 protein. Oncogene 22(2), 168 EOF75 EOF .
  32. 32. DesaintesC.DemeretC.GoyatS.YanivM.ThierryF. 1997 Expression of the papillomavirus E2 protein in HeLa cells leads to apoptosis. EMBO J 16(3), 504-514.
  33. 33. DollardS. C.WilsonJ. L.DemeterL. M.BonnezW.ReichmanR. C.BrokerT. R.ChowL. T. 1992 Production of human papillomavirus and modulation of the infectious program in epithelial raft cultures.. Genes Dev 6(7), 1131-1142.
  34. 34. DonaldsonM. M.BonerW.MorganI. M. 2007 TopBP1 regulates human papillomavirus type 16 E2 interaction with chromatin. J Virol 81(8), 4338-4342.
  35. 35. DoorbarJ. 2005 The papillomavirus life cycle. J Clin Virol 32 Suppl 1, S715 .
  36. 36. DoorbarJ.PartonA.HartleyK.BanksL.CrookT.StanleyM.CrawfordL. 1990 Detection of novel splicing patterns in a HPV16 -containing keratinocyte cell line. Virology 178(1), 254-262.
  37. 37. DurstM.GissmannL.IkenbergH.zurHausen. H. 1983 A papillomavirus DNA from a cervical carcinoma and its prevalence in cancer biopsy samples from different geographic regions. Proc Natl Acad Sci U S A 80(12), 3812 EOF -3815.
  38. 38. EgawaK. 2003 Do human papillomaviruses target epidermal stem cells? Dermatology 207 251254 .
  39. 39. FergusonM.BotchanM. 1996 Genetic analysis of the activation domain of bovine papillomavirus protein E2: its role in transcription and replication. J Virol 70(7), 4193-4199.
  40. 40. FloresE. R.LambertP. F. 1997 Evidence for a switch in the mode of human papillomavirus type 16 DNA replication during the viral life cycle. J Virol 71(10), 7167-7179.
  41. 41. FoutsE. T.YuX.EgelmanE. H.BotchanM. R. 1999 Biochemical and electron microscopic image analysis of the hexameric E1 helicase. J Biol Chem 274(7), 4447 EOF58 EOF .
  42. 42. FrattiniM. G.HurstS. D.LimH. B.SwaminathanS.LaiminsL. A. 1997 Abrogation of a mitotic checkpoint by E2 proteins from oncogenic human papillomaviruses correlates with increased turnover of the 53 tumor suppressor protein. Embo J 16(2), 318-331.
  43. 43. GammohN.GrmH. S.MassimiP.BanksL. 2006 Regulation of human papillomavirus type 16 E7 activity through direct protein interaction with the E2 transcriptional activator. J Virol 80(4), 1787-1797.
  44. 44. Garner-HamrickP. A.FostelJ. M.ChienW. M.BanerjeeN. S.ChowL. T.BrokerT. R.FisherC. 2004 Global effects of human papillomavirus type 18 E6/E7 in an organotypic keratinocyte culture system. J Virol 78(17), 9041 EOF50 EOF .
  45. 45. GeimanenJ.Isok-PaasH.PipitchR.SalkK.LaosT.OravM.ReinsonT.UstavM.Jr UstavM.UstavE. 2011 Development of a cellular assay system to study the genome replication of high- and low-risk mucosal and cutaneous human papillomaviruses. J Virol., in press.
  46. 46. GilbertD.CohenS. 1987 Bovine papilloma virus plasmids replicate randomly in mouse fibroblasts throughout S phase of the cell cycle. GilbertD.CohenS. (1987). Bovine papilloma virus plasmids replicate randomly in mouse fibroblasts throughout S phase of the cell cycle. Cell 50(1), 59-68. 50(1), 59 EOF68 EOF .
  47. 47. GlossB.BernardH. U. 1990 The E6/E7 promoter of human papillomavirus type 16 is activated in the absence of E2 proteins by a sequence-aberrant Sp1 distal element. J Virol 64(11), 5577 EOF84 EOF .
  48. 48. GoodwinE. C.Di MaioD. 2000 Repression of human papillomavirus oncogenes in HeLa cervical carcinoma cells causes the orderly reactivation of dormant tumor suppressor pathways. Proc Natl Acad Sci U S A 97(23), 12513-12518.
  49. 49. GoodwinE. C.YangE.LeeC. J.LeeH. W.Di MaioD.HwangE. S. 2000 Rapid induction of senescence in human cervical carcinoma cells. Proc Natl Acad Sci U S A 97(20), 10978-10983.
  50. 50. GrmH. S.MassimiP.GammohN.BanksL. 2005 Crosstalk between the human papillomavirus E2 transcriptional activator and the E6 oncoprotein. Oncogene 24(33), 5149-5164.
  51. 51. GrosselM.SverdrupF.BreidingD.AndrophyE. 1996 Transcriptional activation function is not required for stimulation of DNA replication by bovine papillomavirus type 1 E2. J Virol 70(10), 7264 EOF9 EOF .
  52. 52. HarrisS. F.BotchanM. R. 1999 Crystal structure of the human papillomavirus type 18 E2 activation domain. Science 284(5420), 1673-1677.
  53. 53. HegdeR.GrossmanS.LaiminsL.SiglerP. 1992 Crystal structure at 1.7 A of the bovine papillomavirus-1 E2 DNA-binding domain bound to its DNA target [see comments]. Nature 359(6395), 505-512.
  54. 54. HinesC.MeghooC.ShettyS.BiburgerM.BrenowitzM.HegdeR. 1998 DNA structure and flexibility in the sequence-specific binding of papillomavirus E2 proteins. J Mol Biol 276(4), 809 EOF18 EOF .
  55. 55. HoffmannR.HirtB.BechtoldV.BeardP.RajK. 2006 Different modes of human papillomavirus DNA replication during maintenance. J Virol 80(9), 4431-4439.
  56. 56. HuY.ClowerR. V.MelendyT. 2006 Cellular topoisomerase I modulates origin binding by bovine papillomavirus type 1 E1. J Virol 80(9), 4363-4371.
  57. 57. HubbertN.SchillerJ.LowyD.AndrophyE. 1988 Bovine papilloma virus-transformed cells contain multiple E2 proteins. Proc Natl Acad Sci U S A 85(16), 5864 EOF -5868.
  58. 58. HubertW. G.KanayaT.LaiminsL. A. 1999 DNA replication of human papillomavirus type 31 is modulated by elements of the upstream regulatory region that lie 5’ of the minimal origin. J Virol 73(3), 1835-1845.
  59. 59. HubertW. G.LaiminsL. A. 2002 Human papillomavirus type 31 replication modes during the early phases of the viral life cycle depend on transcriptional and posttranscriptional regulation of E1 and E2 expression. J Virol 76(5), 2263-2273.
  60. 60. HudsonJ. B.BedellM. A.Mc CanceD. J.LaiminisL. A. 1990 Immortalization and altered differentiation of human keratinocytes in vitro by the E6 and E7 open reading frames of human papillomavirus type 18. J Virol 64(2), 519-526.
  61. 61. HummelM.HudsonJ. B.LaiminsL. A. 1992 Differentiation-induced and constitutive transcription of human papillomavirus type 31b in cell lines containing viral episomes. J Virol 66(10), 6070 EOF80 EOF .
  62. 62. HwangE.NaegerL.Di MaioD. 1996 Activation of the endogenous 53 growth inhibitory pathway in HeLa cervical carcinoma cells by expression of the bovine papillomavirus E2 gene. Oncogene 12(4), 795-803.
  63. 63. IlvesI.KadajaM.UstavM. 2003 Two separate replication modes of the bovine papillomavirus BPV1 origin of replication that have different sensitivity to 53 Virus Res 96(1-2), 75-84.
  64. 64. IlvesI.KiviS.UstavM. 1999 Long-term episomal maintenance of bovine papillomavirus type 1 plasmids is determined by attachment to host chromosomes, which Is mediated by the viral E2 protein and its binding sites. J Virol 73(5), 4404-4412.
  65. 65. IlvesI.MaemetsK.SillaT.JaniksonK.UstavM. 2006 Brd4 is involved in multiple processes of the bovine papillomavirus type 1 life cycle. J Virol 80(7), 3660-3665.
  66. 66. JangM. K.KwonD.Mc BrideA. A. 2009 Papillomavirus E2 proteins and the host BRD4 protein associate with transcriptionally active cellular chromatin. J Virol 83(6), 2592-2600.
  67. 67. JeckelS.LoetzschE.HuberE.StubenrauchF.IftnerT. 2003 Identification of the E9/E2C cDNA and functional characterization of the gene product reveal a new repressor of transcription and replication in cottontail rabbit papillomavirus. J Virol 77(16), 8736-8744.
  68. 68. JiangM.MilnerJ. 2002 Selective silencing of viral gene expression in HPV-positive human cervical carcinoma cells treated with siRNA, a primer of RNA interference. Oncogene 21(39), 6041 EOF8 EOF .
  69. 69. JonesD. L.AlaniR. M.MungerK. 1997 The human papillomavirus E7 oncoprotein can uncouple cellular differentiation and proliferation in human keratinocytes by abrogating 21Cip1 inhibition of cdk2. Genes Dev 11(16), 2101-2111.
  70. 70. KadajaM.Isok-PaasH.LaosT.UstavE.UstavM. 2009 Mechanism of genomic instability in cells infected with the high-risk human papillomaviruses. PLoS Pathog 5(4), e1000397.
  71. 71. KadajaM.SumerinaA.VerstT.OjarandM.UstavE.UstavM. 2007 Genomic instability of the host cell induced by the human papillomavirus replication machinery. Embo J 26(8), 2180-2191.
  72. 72. KimK.Garner-HamrickP. A.FisherC.LeeD.LambertP. F. 2003 Methylation patterns of papillomavirus DNA, its influence on E2 function, and implications in viral infection. J Virol 77(23), 12450-12459.
  73. 73. KimK.LambertP. 2002 E1 protein of bovine papillomavirus 1 is nor required for the maintenance of viral plasmid DNA replication. Virology 293 1014 .
  74. 74. KristiansenE.JenkinsA.HolmR. 1994 Coexistence of episomal and integrated HPV16 DNA in squamous cell carcinoma of the cervix. J Clin Pathol 47(3), 253-256.
  75. 75. KumarR. A.NaiduS. R.WangX.ImbalzanoA. N.AndrophyE. J. 2007 Interaction of papillomavirus E2 protein with the Brm chromatin remodeling complex leads to enhanced transcriptional activation. J Virol 81(5), 2213-2220.
  76. 76. KurgR.SildK.IlvesA.SeppM.UstavM. 2005 Association of bovine papillomavirus E2 protein with nuclear structures in vivo. J Virol 79(16), 10528-10539.
  77. 77. KurgR.TekkelH.AbroiA.UstavM. 2006 Characterization of the functional activities of the Bovine Papillomavirus type 1 E2 protein "single-chain" heterodimers. J Virol 80(22), 11218-11225.
  78. 78. KurgR.UusenP.SeppT.SeppM.AbroiA.UstavM. 2009 Bovine papillomavirus type 1 E2 protein heterodimer is functional in papillomavirus DNA replication in vivo. Virology 386(2), 353-359.
  79. 79. KurgR.UusenP.VosaL.UstavM. 2010 Human papillomavirus E2 protein with single activation domain initiates HPV18 genome replication, but is not sufficient for long-term maintenance of virus genome. Virology 408(2), 159-166.
  80. 80. LaceM. J.AnsonJ. R.ThomasG. S.TurekL. P.HaugenT. H. 2008 The E8--E2 gene product of human papillomavirus type 16 represses early transcription and replication but is dispensable for viral plasmid persistence in keratinocytes. J Virol 82(21), 10841-10853.
  81. 81. LambertP.MonkB.HowleyP. 1990 Phenotypic analysis of bovine papillomavirus type 1 E2 repressor mutants. J Virol 64(2), 950-956.
  82. 82. LawM.LowyD.DvorezkyI.HowleyP. 1981 Mouse cells transformed by bovine papillomavirus contain only extra-chromosomal viral DNA sequences. PNAS 78 27272731 .
  83. 83. LeeD.HwangS.KimJ.ChoeJ. 2002a Functional interaction between p/CAF and human papillomavirus E2 protein. J. Biol. Chem. 277 64836489 .
  84. 84. LeeD.KimH.JeongK.ShimY.HorikawaI.BarrettJ.ChoeJ. 2002b Human papillomavirus E2 down-regulates the human telomerase reverse transcriptase promoter. J. Biol. Chem. 277 2774827756 .
  85. 85. LeeD.LeeB.KimJ.KimD.ChoeJ. 2000 cAMP response element-binding protein-binding protein binds to human papillomavirus E2 protein and activates E2-dependent transcription. J. Biol. Chem. 275 70457051 .
  86. 86. LehmanC. W.BotchanM. R. 1998 Segregation of viral plasmids depends on tethering to chromosomes and is regulated by phosphorylation. Proc Natl Acad Sci U S A 95(8), 4338-4343.
  87. 87. LepikD.IlvesI.KristjuhanA.MaimetsT.UstavM. 1998 53 protein is a suppressor of papillomavirus DNA amplificational replication. J Virol 72(8), 6822-6831.
  88. 88. LiR.KnightJ.BreamG.StenlundA.BotchanM. 1989 Specific recognition nucleotides and their DNA context determine the affinity of E2 protein for 17 binding sites in the BPV-1 genome. Genes Dev 3(4), 510-526.
  89. 89. LiR.KnightJ.JacksonS.TjianR.BotchanM. 1991 Direct interaction between Sp1 and the BPV enhancer E2 protein mediates synergistic activation of transcription. Cell 65(3), 493-505.
  90. 90. LimD.GossenM.LehmanC.BotchanM. 1998 Competition for DNA binding sites between the short and long forms of E2 dimers underlies repression in bovine papillomavirus type 1 DNA replication control. J Virol 72(3), 1931-1940.
  91. 91. LiuJ.KuoS.BrokerT.ChowL. 1995 The functions of human papillomavirus type 11 E1, E2, and E2C proteins in cell-free DNA replication. J Biol Chem 270(45), 27283-27291.
  92. 92. LongworthM. S.LaiminsL. A. 2004a The binding of histone deacetylases and the integrity of zinc finger-like motifs of the E7 protein are essential for the life cycle of human papillomavirus type 31. J Virol 78(7), 3533-3541.
  93. 93. LongworthM. S.LaiminsL. A. 2004b Pathogenesis of human papillomaviruses in differentiating epithelia. Microbiol Mol Biol Rev 68(2), 362-372.
  94. 94. MassimiP.PimD.BertoliC.BouvardV.BanksL. 1999 Interaction between the HPV-16 E2 transcriptional activator and 53 Oncogene 18(54), 7748-7754.
  95. 95. Mc BrideA.ByrneJ.HowleyP. 1989 E2 polypeptides encoded by bovine papillomavirus type 1 form dimers through the common carboxyl-terminal domain: transactivation is mediated by the conserved amino-terminal domain. Proc Natl Acad Sci U S A 86(2), 510-514.
  96. 96. Mc CanceD. J.KopanR.FuchsE.LaiminsL. A. 1988 Human papillomavirus type 16 alters human epithelial cell differentiation in vitro. Proc Natl Acad Sci U S A 85(19), 7169-7173.
  97. 97. Mc Laughlin-DrubinM.MüngerK. 2009 Oncogenic activities of human papillomaviruses. Virus Research 143 195208 .
  98. 98. Mc PhillipsM. G.OliveiraJ. G.SpindlerJ. E.MitraR.Mc BrideA. A. 2006 Brd4 is required for e2 -mediated transcriptional activation but not genome partitioning of all papillomaviruses. J Virol 80(19), 9530-9543.
  99. 99. Mc PhillipsM. G.OzatoK.Mc BrideA. A. 2005 Interaction of bovine papillomavirus E2 protein with Brd4 stabilizes its association with chromatin. J Virol 79(14), 8920-8932.
  100. 100. MelansonS. M.AndrophyE. J. 2009 Topography of bovine papillomavirus E2 protein on the viral genome during the cell cycle. Virology 393(2), 258-264.
  101. 101. MelendyT.SedmanJ.StenlundA. 1995 Cellular factors required for papillomavirus DNA replication. J Virol 69(12), 7857-7867.
  102. 102. MeyersC.FrattiniM. G.HudsonJ. B.LaiminsL. A. 1992 Biosynthesis of human papillomavirus from a continuous cell line upon epithelial differentiation. Science 257(5072), 971-973.
  103. 103. MohrI.ClarkR.SunS.AndrophyE.MacPherson. P.BotchanM. 1990 Targeting the E1 replication protein to the papillomavirus origin of replication by complex formation with the E2 transactivator. Science 250(4988), 1694-1699.
  104. 104. MoleS.MilliganS.GrahamS. 2009 Human papillomavirus type 16 E2 protein transcriptionally activates the promoter of a key cellular splicing factor, SF2/ASF J Virol 83(1), 357-367.
  105. 105. MoodyC.LaiminsL. 2010 Human papillomavirus oncoproteins: pathways to transformation. Nat Rev Cancer 10 550560 .
  106. 106. MullerF.SeoY.HurwitzJ. 1994 Replication of bovine papillomavirus type 1 origin-containing DNA in crude extracts and with purified proteins. J Biol Chem 269(25), 17086-17094.
  107. 107. MungalS.SteinbergB. M.TaichmanL. B. 1992 Replication of plasmid-derived human papillomavirus type 11 DNA in cultured keratinocytes. J Virol 66(5), 3220-3224.
  108. 108. MüllerA.RitzkowskyA.StegerG. 2002 Cooperative activation of human papillomavirus type 8 gene expression by the E2 protein and the cellular coactivator 300 J. Virol. 76, 11042-11053.
  109. 109. OhS. T.KyoS.LaiminsL. A. 2001 Telomerase activation by human papillomavirus type 16 E6 protein: induction of human telomerase reverse transcriptase expression through Myc and GC-rich Sp1 binding sites. J Virol 75(12), 5559-5566.
  110. 110. OldakM.SmolaH.AumailleyM.RiveroF.PfisterH.Smola-HessS. 2004 The human papillomavirus type 8 E2 protein suppresses beta4 -integrin expression in primary human keratinocytes. J Virol 78(19), 10738-10746.
  111. 111. OliveiraJ. G.ColfL. A.Mc BrideA. A. 2006 Variations in the association of papillomavirus E2 proteins with mitotic chromosomes. Proc Natl Acad Sci U S A 103(4), 1047-1052.
  112. 112. ParishJ. L.BeanA. M.ParkR. B.AndrophyE. J. 2006a ChlR1 is required for loading papillomavirus E2 onto mitotic chromosomes and viral genome maintenance. Mol Cell 24(6), 867-876.
  113. 113. ParishJ. L.KowalczykA.ChenH. T.RoederG. E.SessionsR.BuckleM.GastonK. 2006b E2 proteins from high- and low-risk human papillomavirus types differ in their ability to bind 53 and induce apoptotic cell death. J Virol 80(9), 4580-4590.
  114. 114. ParkP.CopelandW.YangL.WangT.BotchanM.MohrI. 1994 The cellular DNA polymerase alpha-primase is required for papillomavirus DNA replication and associates with the viral E1 helicase. Proc Natl Acad Sci U S A 91(18), 8700-8704.
  115. 115. PettM. R.AlazawiW. O.RobertsI.DowenS.SmithD. I.StanleyM. A.ColemanN. 2004 Acquisition of high-level chromosomal instability is associated with integration of human papillomavirus type 16 in cervical keratinocytes. Cancer Res 64(4), 1359-1368.
  116. 116. PettM.ColemanN. 2007 Integration of high-risk human papillomavirus: a key event in cervical carcinogenesis? J Pathol 212 356367 .
  117. 117. PfefferleR.MarcuzziG. P.AkgulB.KasperH. U.SchulzeF.HaaseI.WickenhauserC.PfisterH. 2008 The human papillomavirus type 8 E2 protein induces skin tumors in transgenic mice. J Invest Dermatol 128(9), 2310-2315.
  118. 118. PiirsooM.UstavE.MandelT.StenlundA.UstavM. 1996 Cis and trans requirements for stable episomal maintenance of the BPV-1 replicator. EMBO J 15(1), 1-11.
  119. 119. PoddarA.ReedS. C.Mc PhillipsM. G.SpindlerJ. E.Mc BrideA. A. 2009 The human papillomavirus type 8 E2 tethering protein targets the ribosomal DNA loci of host mitotic chromosomes. J Virol 83(2), 640-650.
  120. 120. PowellM. L.SmithJ. A.SowaM. E.HarperJ. W.IftnerT.StubenrauchF.HowleyP. M. 2010 NCoR1 mediates papillomavirus E8;E2C transcriptional repression. J Virol 84(9), 4451-4460.
  121. 121. RavnanJ.GilbertD.TenHagen. K.CohenS. 1992 Random-choice replication of extrachromosomal bovine papillomavirus (BPV) molecules in heterogeneous, clonally derived BPV-infected cell lines. J Virol 66(12), 6946-6952.
  122. 122. RehtanzM.SchmidtH.WarthorstU.StegerG. 2004 Direct interaction between nucleosome assembly protein 1 and the papillomavirus E2 proteins involved in activation of transcription. Mol.Cell.Biol. 24 21532168 .
  123. 123. RemmM.BrainR.JenkinsJ. 1992 The E2 binding sites determine the efficiency of replication for the origin of human papillomavirus type 18. Nucleic Acids Res 20(22), 6015-6021.
  124. 124. RemmM.RemmA.UstavM. 1999 Human papillomavirus type 18 E1 protein is translated from polycistronic mRNA by a discontinuous scanning mechanism. J Virol 73(4), 3062-3070.
  125. 125. SandersC. M.StenlundA. 1998 Recruitment and loading of the E1 initiator protein: an ATP-dependent process catalysed by a transcription factor. Embo J 17(23), 7044-7055.
  126. 126. SandersC. M.StenlundA. 2000 Transcription factor-dependent loading of the E1 initiator reveals modular assembly of the papillomavirus origin melting complex. J Biol Chem 275(5), 3522-3534.
  127. 127. ScheffnerM.WernessB. A.HuibregtseJ. M.LevineA. J.HowleyP. M. 1990 The E6 oncoprotein encoded by human papillomavirus types 16 and 18 promotes the degradation of 53 Cell 63(6), 1129-1136.
  128. 128. SchmittA.HarryJ. B.RappB.WettsteinF. O.IftnerT. 1994 Comparison of the properties of the E6 and E7 genes of low- and high-risk cutaneous papillomaviruses reveals strongly transforming and high Rb-binding activity for the E7 protein of the low-risk human papillomavirus type 1. J Virol 68(11), 7051-7059.
  129. 129. SchuckS.StenlundA. 2005 Assembly of a double hexameric helicase. Mol Cell 20(3), 377-89.
  130. 130. SchvartzmanJ. B.AdolphS.Martin-ParrasL.SchildkrautC. L. 1990 Evidence that replication initiates at only some of the potential origins in each oligomeric form of bovine papillomavirus type 1 DNA. Mol Cell Biol 10(6), 3078-3086.
  131. 131. SchweigerM. R.OttingerM.YouJ.HowleyP. M. 2007 Brd4 -independent transcriptional repression function of the papillomavirus e2 proteins. J Virol 81(18), 9612-9622.
  132. 132. SchweigerM. R.YouJ.HowleyP. M. 2006 Bromodomain protein 4 mediates the papillomavirus E2 transcriptional activation function. J Virol 80(9), 4276-4285.
  133. 133. SedmanJ.StenlundA. 1995 Co-operative interaction between the initiator E1 and the transcriptional activator E2 is required for replicator specific DNA replication of bovine papillomavirus in vivo and in vitro. EMBO J 14(24), 6218-6228.
  134. 134. SedmanJ.StenlundA. 1998 The papillomavirus E1 protein forms a DNA-dependent hexameric complex with ATPase and DNA helicase activities. J Virol 72 68936897 .
  135. 135. SedmanT.SedmanJ.StenlundA. 1997 Binding of the E1 and E2 proteins to the origin of replication of bovine papillomavirus. J Virol 71(4), 2887-2896.
  136. 136. ShermanL.SchlegelR. 1996 Serum- and calcium-induced differentiation of human keratinocytes is inhibited by the E6 oncoprotein of human papillomavirus type 16. J Virol 70(5), 3269-3279.
  137. 137. SillaT.HaalI.GeimanenJ.JaniksonK.AbroiA.UstavE.UstavM. 2005 Episomal maintenance of plasmids with hybrid origins in mouse cells. J Virol 79(24), 15277-15288.
  138. 138. SillaT.MannikA.UstavM. 2010 Effective formation of the segregation-competent complex determines successful partitioning of the bovine papillomavirus genome during cell division. J Virol 84(21), 11175-11188.
  139. 139. SkiadopoulosM.Mc BrideA. 1998 Bovine papillomavirus type 1 genomes and the E2 transactivator protein are closely associated with mitotic chromatin. J Virol 72(3), 2079-2088.
  140. 140. SmithJ. A.WhiteE. A.SowaM. E.PowellM. L.OttingerM.HarperJ. W.HowleyP. M. 2010 Genome-wide siRNA screen identifies SMCX, EP400, and Brd4 as E2 -dependent regulators of human papillomavirus oncogene expression. Proc Natl Acad Sci U S A 107(8), 3752-3757.
  141. 141. SoedaE.FerranM. C.BakerC. C.Mc BrideA. A. 2006 Repression of HPV16 early region transcription by the E2 protein. Virology 351(1), 29-41.
  142. 142. SpalholzB.YangY.HowleyP. 1985 Transactivation of a bovine papilloma virus transcriptional regulatory element by the E2 gene product. Cell 42(1), 183-191.
  143. 143. SzymanskiP.StenlundA. 1991 Regulation of early gene expression from the bovine papillomavirus genome in transiently transfected C127 cells. J Virol 65(11), 5710-5720.
  144. 144. StanleyM. A.BrowneH. M.ApplebyM.MinsonA. C. 1989 Properties of a non-tumorigenic human cervical keratinocyte cell line. Int J Cancer 43(4), 672-676.
  145. 145. StenlundA. 2003a E1 initiator DNA binding specificity is unmasked by selective inhibition of non-specific DNA binding. Embo J 22(4), 954-963.
  146. 146. StenlundA. 2003b Initiation of DNA replication: lessons from viral initiator proteins. Nature Reviews 4 777785 .
  147. 147. StopplerH.HartmannD. P.ShermanL.SchlegelR. 1997 The human papillomavirus type 16 E6 and E7 oncoproteins dissociate cellular telomerase activity from the maintenance of telomere length. J Biol Chem 272(20), 13332-13337.
  148. 148. StubenrauchF.ColbertA. M.LaiminsL. A. 1998 Transactivation by the E2 protein of oncogenic human papillomavirus type 31 is not essential for early and late viral functions. J Virol 72(10), 8115-8123.
  149. 149. StubenrauchF.HummelM.IftnerT.LaiminsL. A. 2000 The E8E2C protein, a negative regulator of viral transcription and replication, is required for extrachromosomal maintenance of human papillomavirus type 31 in keratinocytes. J Virol 74(3), 1178-1186.
  150. 150. StubenrauchF.LimH. B.LaiminsL. A. 1998 Differential requirements for conserved E2 binding sites in the life cycle of oncogenic human papillomavirus type 31. J Virol 72(2), 1071-1077.
  151. 151. zurHausen. H. 2000 Papillomaviruses causing cancer: evasion from host-cell control in early events in carcinogenesis. J Natl Cancer Inst 92(9), 690-698.
  152. 152. ThainA.JenkinsO.ClarkeA.GastonK. 1996 CpG methylation directly inhibits binding of the human papillomavirus type 16 E2 protein to specific DNA sequences. J Virol 70(10), 7233-7235.
  153. 153. ThierryF. 2009 Transcriptional regulation of the papillomavirus oncogenes by cellular and viral transcription factors in cervical carcinoma. Virology 384(2), 375-379.
  154. 154. ThierryF.BenotmaneM. A.DemeretC.MoriM.TeissierS.DesaintesC. 2004 A genomic approach reveals a novel mitotic pathway in papillomavirus carcinogenesis. Cancer Res 64(3), 895-903.
  155. 155. ThierryF.SpyrouG.YanivM.HowleyP. 1992 Two AP1 sites binding JunB are essential for human papillomavirus type 18 transcription in keratinocytes. J Virol 66(6), 3740-3748.
  156. 156. ThierryF.YanivM. 1987 The BPV1 -E2 trans-acting protein can be either an activator or a repressor of the HPV18 regulatory region. Embo J 6(11), 3391-3397.
  157. 157. ThomasJ. T.HubertW. G.RueschM. N.LaiminsL. A. 1999 Human papillomavirus type 31 oncoproteins E6 and E7 are required for the maintenance of episomes during the viral life cycle in normal human keratinocytes. Proc Natl Acad Sci U S A 96(15), 8449-8454.
  158. 158. UstavE.UstavM.SzymanskiP.StenlundA. 1993 The bovine papillomavirus origin of replication requires a binding site for the E2 transcriptional activator. Proc Natl Acad Sci U S A 90(3), 898-902.
  159. 159. UstavM.StenlundA. 1991 Transient replication of BPV-1 requires two viral polypeptides encoded by the E1 and E2 open reading frames. EMBO J 10(2), 449-457.
  160. 160. UstavM.UstavE.SzymanskiP.StenlundA. 1991 Identification of the origin of replication of bovine papillomavirus and characterization of the viral origin recognition factor E1. EMBO J 10(13), 4321-4329.
  161. 161. Van TineB.DaoL.WuS.Y.SonbuchnerT.LinB.ZouN.ChiangC.M.BrokerT.ChowL. 2004 Human papillomavirus (HPV) origin-binding protein associates with mitotic spindles to enable viral DNA partitioning. PNAS 101 40304035 .
  162. 162. WangH. K.DuffyA. A.BrokerT. R.ChowL. T. 2009a Robust production and passaging of infectious HPV in squamous epithelium of primary human keratinocytes. Genes Dev 23(2), 181-194.
  163. 163. WangX.NaiduS. R.SverdrupF.AndrophyE. J. 2009b Tax1BP1 interacts with papillomavirus E2 and regulates E2 -dependent transcription and stability. J Virol 83(5), 2274-2284.
  164. 164. WellsS. I.FrancisD. A.KarpovaA. Y.DowhanickJ. J.BensonJ. D.HowleyP. M. 2000 Papillomavirus E2 induces senescence in HPV-positive cells via pRB- and 21 CIP)-dependent pathways. Embo J 19(21), 5762-5771.
  165. 165. WernessB. A.LevineA. J.HowleyP. M. 1990 Association of human papillomavirus types 16 and 18 E6 proteins with 53 Science 248(4951), 76-79.
  166. 166. WuS. Y.LeeA. Y.HouS. Y.KemperJ. K.Erdjument-BromageH.TempstP.ChiangC. M. 2006 Brd4 links chromatin targeting to HPV transcriptional silencing. Genes Dev 20(17), 2383-2396.
  167. 167. YangL.LiR.MohrI.ClarkR.BotchanM. 1991a Activation of BPV-1 replication in vitro by the transcription factor E2. Nature 353(6345), 628-632.
  168. 168. YangL.MohrI.FoutsE.LimD.NohaileM.BotchanM. 1993 The E1 protein of bovine papilloma virus 1 is an ATP-dependent DNA helicase. Proc Natl Acad Sci U S A 90(11), 5086-5090
  169. 169. YangL.MohrI.LiR.NottoliT.SunS.BotchanM. 1991b Transcription factor E2 regulates BPV-1 DNA replication in vitro by direct protein-protein interaction. Cold Spring Harb Symp Quant Biol 56 335346 .
  170. 170. YouJ. 2010 Papillomavirus interaction with cellular chromatin. Biochim Biophys Acta 1799(3-4), 192-199.
  171. 171. YouJ.CroyleJ.NishimuraA.OzatoK.HowleyP. 2004 Interaction of the bovine papillomavirus E2 protein with brd4 tethers the viral DNA to host mitotic chromosomes. Cell 117 349360 .
  172. 172. YuT.PengY. C.AndrophyE. J. 2007 Mitotic kinesin-like protein 2 binds and colocalizes with papillomavirus E2 during mitosis. J Virol 81(4), 1736-1745.

Written By

Reet Kurg

Submitted: 04 November 2010 Published: 01 August 2011