Genetic characterization of variants of HPV‑16, HPV‑18 and HPV‑52 circulating in Italy among general and high‑risk populations

  • Authors:
    • Elena Rosanna Frati
    • Silvia Bianchi
    • Antonella Amendola
    • Daniela Colzani
    • Fabio Petrelli
    • Gianguglielmo Zehender
    • Elisabetta Tanzi
  • View Affiliations

  • Published online on: November 25, 2019     https://doi.org/10.3892/mmr.2019.10847
  • Pages: 894-902
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Viral factors, such as high‑risk human papillomavirus variants, can increase the risk of viral persistence and influence the progression to cancer. In the present study, the long control region (LCR) of human papillomavirus (HPV)‑16 and HPV‑52, and the L1 region of HPV‑16 and HPV‑18, identified from subjects belonging to both general and high‑risk populations (migrants, HIV+ subjects and adolescent/young people) residing in Italy, were characterized using molecular and phylogenetic techniques. To the best of our knowledge, this is the first Italian study to analyze a large number of sequences (n=458) and report phylogenetic data on the HPV‑52 variants. The phylogenetic analysis showed that 90% of the LCR variants of HPV‑16 and HPV‑52 clustered within lineage A (European lineage) and only sequences identified from subjects belonging to high‑risk populations fell into the non‑European lineages. Analysis of the LCRs revealed a high genomic diversity with a large number of changes. Several mutations in the binding sites for viral and cellular transcription factors characterized the HPV‑16 LCR variants belonging to the African lineages B and C, were observed in subjects with cytological abnormalities (high squamous intraepithelial lesions). The HPV‑16 and HPV‑18 L1 molecular characterization identified 30% of changes in the immune‑dominant epitope loops. These data give a clear picture of the situation in Italy, and a starting point for understanding the molecular pathogenesis and developing molecular diagnostics for HPV, vaccines and other therapeutic approaches in order to control and/or eliminate virus‑induced diseases.

Introduction

Human papillomavirus (HPV) infection is one of the main causes of infection-related cancer in both men and women (1). Among >200 types of HPVs, high-risk HPV (HR-HPV-16, −18, −31, −33, −35, −39, −45, −51, −52, −56, −58 and −59) infections are responsible for almost all cervical cancer cases and there is growing evidence that they can cause other anogenital cancers, including anal, vulval, vaginal and penile, and head and neck cancers (1,2). However, only a relatively small number of lesions associated with HR-HPV infections evolve into high-grade lesions or cancer. In addition to genetic and immunological factors, viral factors, such as HR-HPV variants, viral load and viral integration, can increase the risk of viral persistence and influence progression to cancer (3,4). In particular, it is important to monitor the genetic variability of HR-HPVs over time in order to estimate the clinical course of HR-HPV infections, predict the prognosis of benign and malignant lesions and define treatment strategies, as genetic diversity can influence the long-term efficacy of current HPV vaccines.

The α-HPV types have a circular, double-stranded DNA genome of ~7,900 bp consisting of eight protein-coding genes (L1, L2, E1, E2, E4, E5, E6 and E7), a non-coding region (NCR) and a long control region (LCR) (5). The LCR of HPVs contain the highest degree of genomic diversity and is usually used to classify HPV variants into lineages and sublineages, previously described as geographic origin lineages (4,68). In particular, HPV-16 LCR variants were grouped into four major lineages and nine sublineages: i) Lineage A, including A1, A2, A3 (previously known as European) and A4 (Asian) sublineages; ii) lineage B, including B1 (African-1a) and B2 (African-1b) sublineages; iii) lineage C (African-2); and iv) lineage D, including D1 (North American, NA1), D2 (Asian-American, AA2) and D3 (Asian-American, AA1) sublineages (4,7). HPV-52 variants were grouped into four major lineages and 6 sublineages: i) Lineage A, including A1 and A2 (European) sublineages; ii) lineage B, including B1 (African-1a) and B2 (African-1b) sublineages; iii) lineage C, including C1 (African-2a) and C2 (African-2b) sublineages; and iv) lineage D (Asian-American) (4). Moreover, the LCR contains the early promoter and various transcriptional regulatory sites for both viral and cellular proteins, such as E2, yin-yang 1 (YY1), activator protein 1 (AP-1), octamer 1 (Oct-1), nuclear factor 1 (NF-1) and transcriptional enhancer factor 1 (TEF-1) (9). The L1 gene, encoding the major capsid protein, is a conserved region that is used to classify HPVs into species and types. Furthermore, the L1 protein can self-assemble into virus-like particles (VLPs), which are used for producing prophylactic vaccines (10). In total, five hypervariable immune-dominant regions (BC, DE, EF, FG and HI) show high levels of polymorphism in and among HPV types, resulting in the generation of neutralizing antibodies of different binding affinities (11). The phylogenetic analysis of the LCR and L1 regions, and the study of single nucleotide polymorphisms (SNPs) and amino acid mutations allows the identification of the HR-HPV variants circulating in the population. The description and understanding of HR-HPV genetic variants is an important area for molecular pathogenesis, and for the development of molecular diagnostics for HPV, vaccines and other therapeutic approaches aimed at controlling and/or eliminating virus-induced diseases.

Among HR-HPVs, HPV-16 and HPV-18 are responsible for approximately 70% of cervical cancers worldwide (1). The HPV-16 and HPV-18 L1 regions are the targets of the HPV vaccine and, therefore, the study of their variants is a high priority. HPV-16 is the most common HPV type worldwide, while other HR-HPVs, such as HPV-52 in HIV-positive subjects (12), are particularly prevalent among high-risk populations. These types can develop persistent HPV infections and put the patients at risk of progression to cancer, and so these patients should be closely monitored.

The aim of the present study was to retrospectively describe the genetic variability of the LCR region in the HPV-16 and HPV-52 types, and of the L1 region of the HPV-16 and HPV-18 types. In the present study, HPV sequences identified from subjects enrolled in previous studies (1317) were analyzed.

Materials and methods

Study sequences

The LCR sequences of HPV-16 and HPV-52 (n=221 and n=41, respectively), and L1 sequences of HPV-16 and HPV-18 (n=148 and n=48, respectively) were analyzed. The sequences were obtained from 375 cervical and 83 anal swabs positive for HPV-16, HPV-18 and HPV-52 in our previous studies (1317). No patients were enrolled in the present study.

In total, 95 of the 458 sequences analyzed were identified from the general population [54 women; median age, 34 years; interquartile range (IQR)=29–41], whereas 363 were identified in high-risk groups for the acquisition of HPV infection, as adolescents/young people (36 girls; median age, 22 years; IQR=21–25), HIV positive subjects (114 women, median age 43 years, IQR=37–47; 69 men, median age 35 years, IQR=30–42) and migrants (37 women, median age 29 years, IQR=25–40). Upon informed consent of the participants, all the samples were collected at the clinical centers that collaborated in the previous studies (1317). The approval of the ethics committee was obtained for each of the previous studies (1317).

Nucleic acid extraction and sequencing amplification

Nucleic acids were extracted from the biological samples using the NucliSENS® easyMAG™ automated platform (BioMérieux Benelux B.V.), according to the off-board lysis protocol (https://www.biomerieux-diagnostics.com/nuclisensr-easymagr). HPV-16 LCR fragments were obtained using a previously described in-house PCR (17) and HPV-52 LCR was amplified using nested-PCR, as previously described (18). L1 genetic characterization was performed by sequence analysis of a 1,488 bp L1 gene amplicon for HPV-16 and a 1,489 bp L1 gene amplicon for HPV-18 obtained by two partially overlapping fragments amplified using degenerate primers (19). The primer sequences used in the amplification protocols are presented in Table SI.

Each PCR run included both negative (water) and positive controls (DNA extracted from HPV-16, HPV-18 and HPV-52 positive samples); each sample was tested three times to confirm the mutations detected.

Following PCR amplification, the amplicons were purified using the NucleoSpin® Extract II purification kit (Macherey-Nagel GmbH) and the nucleotide sequences were obtained using automated DNA sequencing with an ABI PRISM 3100 genetic analyzer (Applied Biosystem; Thermo Fisher Scientific, Inc.).

Sequence analysis

Multiple nucleotide sequences were aligned using ClustalX version 2.0 (20). SNPs and amino acid changes were determined by examining the sequence chromatograms using the MEGA 6.0 software package (21). BioEdit version 7.2.5 (22) was used to assess the effects of LCR and L1 variations on the binding sites for cellular transcription factors and immune-dominant epitopes, respectively. Site-specific entropy was estimated using BioEdit to evaluate genetic diversity (22). A value of zero indicated site-specific conservation and higher values indicated increasing degrees of site-specific variation.

Phylogenetic analysis

Phylogenetic trees were constructed using the Neighbor-Joining method (23), the Kimura 2-Parameter model (24) and the MEGA 6.0 software package (21). A bootstrap re-sampling analysis was performed (1,000 replicates) to test tree robustness. The reference viral strains used for constructing the phylogenetic trees, selected according to the classification used by Burk et al (4), were obtained from the NCBI GenBank Database (https://www.ncbi.nlm.nih.gov/nucleotide/).

LCR sequences and cytology

In total, 117 (52.9%) of the 221 HPV-16 LCR sequences were obtained from cervical/anal samples with cytological abnormalities, 78 of which were low-grade squamous intraepithelial lesions (LSIL) and 39 were high-grade squamous intraepithelial lesions (HSIL). With regard to the 41 HPV-52 LCR sequences, 17 (41.5%) were obtained from samples with cytological abnormalities (13 LSIL and 4 HSIL).

L1 sequences and selective pressure

The partitioning approach to robust interference of selection method, available on the DataMonkey 2.0 server (www.datamonkey.org) (25), was used to detect whether a proportion of sites in the alignment of HPV-16 and HPV-18 L1 gene sequences evolved with a positive selective pressure, shown by a dN/dS>1, which expresses the ratio of non-synonymous to synonymous substitutions. The integrative analyses of four different codon-based maximum likelihood methods, single likelihood ancestor counting (SLAC), fixed effects likelihood (FEL), random effects likelihood (REL) and mixed effect model of evolution (MEME), all incorporating the HKY85 substitution model with phylogenetic trees, inferred using the Neighbor-Joining method, were carried out in order to estimate the dN/dS ratio for all codon alignments.

Results

HPV-16 and HPV-52 LCR variants
HPV-16
Nucleotide polymorphisms analysis

A total of 221 HPV-16 partial LCRs were successfully sequenced and analyzed. In total 80 SNPs were identified in 73 nucleotide sites of the 735 bases of HPV-16 LCR (10.9% variable nucleotide positions), resulting in 62 unique variants identified in one or more of the study samples (variant IDs 1 to 62; Table I). On comparing the sequences analyzed with the reference sequence (accession number, K02718 A1), it was observed that >40 SNPs had previously been identified and reported, while 40 SNPs (40/80; 50%; Table SII) were, to the best of our knowledge, identified for the first time in the present study.

Table I.

Frequencies and GenBank accession numbers of analyzed HPV-16, HPV-18 and HPV-52 variants.

Table I.

Frequencies and GenBank accession numbers of analyzed HPV-16, HPV-18 and HPV-52 variants.

TargetVariant nameFrequencyGenBank accession number
LCR HPV-16Variant 12EU650455
Variant 262EU650473
Variant 33EU650433
Variant 41MH028439
Variant 51EU650463
Variant 62MH028440
Variant 71MH028441
Variant 81MH028442
Variant 91MH028443
Variant 101MH028444
Variant 111MH028445
Variant 124EU650446
Variant 131MH028446
Variant 142MH028447
Variant 152MH028448
Variant 161MH028449
Variant 171MH028450
Variant 181MH028451
Variant 191MH028452
Variant 203MH028453
Variant 211MH028454
Variant 221MH028455
Variant 231MH028456
Variant 246EU650442
Variant 254MH028457
Variant 2631EU650481
Variant 271MH028458
Variant 281MH028459
Variant 292MH028460
Variant 303EU650465
Variant 311MH028461
Variant 321MH028462
Variant 331MH028463
Variant 342MH028464
Variant 351MH028465
Variant 361MH028466
Variant 371MH028467
Variant 381MH028468
Variant 3938EU650484
Variant 403MH028469
Variant 411MH028470
Variant 421MH028471
Variant 432MH028472
Variant 441MH028473
Variant 451MH028474
Variant 461MH028475
Variant 471MH028476
Variant 481MH028477
Variant 491EU650448
Variant 501MH028478
Variant 511MH028479
Variant 521MH028480
Variant 531MH028481
Variant 542EU650474
Variant 552MH028482
Variant 561MH028483
Variant 571MH028484
Variant 581MH028485
Variant 591MH028486
Variant 601MH028487
Variant 611MH028488
Variant 623MH028489
L1 HPV-16Variant 11JF728181
Variant 22JF728156
Variant 31JF728169
Variant 43JF728175
Variant 52JF728173
Variant 63JF728162
Variant 75JF728179
Variant 81JF728168
Variant 91JF728165
Variant 1021EU650438
Variant 112JF728176
Variant 121JF728167
Variant 132JF728163
Variant 141JF728172
Variant 1576JF728182
Variant 161JF728161
Variant 171EU650439
Variant 181JF728180
Variant 191JF728159
Variant 201JF728170
Variant 211JF728158
Variant 221JF728164
Variant 238JF728171
Variant 241JF728177
Variant 251JF728155
Variant 261JF728166
Variant 271JF728160
Variant 281EU650483
Variant 291JF728174
Variant 302JF728183
Variant 311JF728157
L1 HPV-18Variant 11MH028419
Variant 21MH028420
Variant 38MH028421
Variant 41MH028422
Variant 53MH028423
Variant 61MH028424
Variant 74MH028425
Variant 81MH028426
Variant 91JF728186
Variant 102JF728184
Variant 111MH028427
Variant 121MH028428
Variant 1310MH028429
Variant 141MH028430
Variant 151JF728185
Variant 161MH028431
Variant 171MH028432
Variant 182MH028433
Variant 191MH028434
Variant 202MH028435
Variant 211MH028436
Variant 221MH028430
Variant 231MH028437
Variant 241MH028438
LCR HPV-52Variant 12MH028409
Variant 220MH028410
Variant 32MH028411
Variant 41MH028412
Variant 52MH028413
Variant 65MH028414
Variant 73MH028415
Variant 82MH028416
Variant 91MH028417
Variant 103MH028418

[i] HPV, human papillomavirus; LCR, long control region.

The site-specific variations in the LCR fragment varied from 0 (no variation) to 0.63. The most common LCR changes with entropy scores>0.4 were as follows: G7521A (168/221; 76%), C7764T (22/221; 9.9%), G7489A (21/221; 9.5%), C7786T (21/221; 9.5%), A7485C (20/221; 9%), C7669T (19/221; 8.6%), C7689A (19/221; 8.6%), G7834T (14/221; 6.3%), C14T/G (12/221; 5.4%), A7729C (11/221; 5.0%), C32T (10/221; 4.5%), A7837C (9/221; 4.1%), A7839G (9/221; 4.1%; Fig. S1). No insertion or deletion mutation sites were identified.

Phylogenetic analysis

Phylogenetic analysis showed that the 62 variants clustered into four main groups, corresponding to lineages A, B, C and D (Fig. 1). In particular, 45 of the variants identified in 200 of the 221 HPV-16 partial LCRs analyzed (200/221; 90.5%) clustered within lineage A (European lineage), 1 variant (1/221; 0.4%) within lineage B (African 1 lineage), 8 variants (9/221; 4.1%) within lineage C (African 2 lineage) and 8 variants (11/221; 4.9%) within lineage D (Asian-American lineage). With regards to the samples in lineage D, 36.3% (4/11) belonged to the sub-lineage D1 (North-American sub-lineages), 27.3% (3/11) to D2 (Asian-American 2 sub-lineages) and 36.3% (4/11) to D3 (Asian-American 1 sub-lineages). All of the 17 variants belonging to lineages B, C and D (Non-European) were detected in the groups of people at higher risk for acquiring HPV infection, such as migrants, HIV+ subjects and adolescent/young people.

SNPs in the transcription factor binding sites

Analysis of the SNPs revealed that 19 fell into the HPV-16 LCR binding sites of transcription factors. In total, three SNPs were located in the TEF-1 binding site, four SNPs were located in the NF-1 binding site, six were located in the YY1 binding site, one SNP fell into the AP-1 binding site, one SNP fell into the Oct-1 binding site and four SNPs were located in the E2 binding site (Table II).

Table II.

Single nucleotide polymorphisms in the human papillomavirus-16 long control region binding sites of transcription factors.

Table II.

Single nucleotide polymorphisms in the human papillomavirus-16 long control region binding sites of transcription factors.

Binding sitesVariant IDLineage
TEF-1 T7469A41A
TEF-1 C7689A46, 47–53, 55–62B, C, D
TEF-1 G7826A32, 33, 47–54A, C
NF-1 T7475G59D
NF-1 G7478A17A
NF-1 G7677A18, 19A
NF-1 C7748A59D
YY1 A7485C47-54, 55–62C, D
YY1 G7521A1-32, 35, 45, 46, 47–54, 55–62A, B, C, D
YY1 C7786T46, 47–54, 55–62B, C, D
YY1 G7826A32-33, 47–54A, C
YY1 A7837C47–54C
YY1 A7839G47–54C
AP-1 T7637A59D
Oct-1 A7839G47–54C
E2 G7869A25, 46, 51–53A, B, C
E2 G7869C50C
E2 A35C6A
E2 C37T47, 48, 53C

[i] TEF1, transcriptional enhancer factor 1; NF-1, nuclear factor 1; YY1, yin-yang 1; AP-1, activator protein 1; Oct-1, octamer 1.

LCR variants and cytology

Overall, the 117 LCR sequences detected from subjects with cytological abnormalities clustered into the four main lineage groups A, B, C and D.

In total, 36.3% (4/11) of the LCR sequences belonging to lineage D (Asian-American lineage) were identified in subjects with cytological abnormalities, all classified as LSIL. With respect to lineage A (European lineage), 51.5% (104/200) of the LCR sequences were from subjects with cytological lesions, 74 of which were LSIL and 30 were HSIL.

Almost all (9/10; 90%) of the African variants (lineages B and C) were detected in subjects with HSIL. The eight sequences belonging to lineage C were characterized by nine mutations in the transcription factor binding sites (one in the TEF-1 site, six in YY1 sites, one in Oct-1 and one in E2), whereas the single sequence clustering within lineage B showed four mutations in the transcription factor binding sites (one in the TEF-1 site, two in YY1 sites and one in E2).

HPV-52
Nucleotide polymorphisms analysis

In total, 41 HPV-52 partial LCR regions were successfully sequenced. On comparing the analyzed sequences with the reference sequence (accession number, X74481 A1), 21 SNPs were identified in the 19 nucleotide sites among the 726 bases of the HPV-52 LCR (2.9% variable nucleotide positions), resulting in 10 unique variants identified in one or more of the study samples (variant IDs 1 to 10; Table I). In total, 13 SNPs had already been identified and reported, while 6 SNPs (6/19; 31.6%, Table SIII) were identified, to the best of our knowledge, for the first time in the present study.

Site-specific variation in the LCR fragment ranged from 0 (no variation) to 1.0 (Fig. S2). The most common LCR changes, with entropy scores >0.4, were C7188A (4/41; 9.8%), G7354T (4/41; 9.8%), G7605A (9/41; 22%), T7607C (16/41; 39%), A7640C (4/41; 9.8%), T7642C (4/41; 9.8%), G7695C (4/41; 9.8%), C7726T (10/41; 24.4%), T7727C (5/41; 12.2%), G7844A (4/41; 9.8%), A7848G (4/41; 9.8%). On comparing the analyzed sequences with the reference sequence (accession number, X74481 A1), four sequences with deletions were found. All of the variants showed deletion sites in position 7370–7374; variant ID 9 presented a deletion site in position 7173–7180, variant IDs 7 and 8 were characterized by a deletion in the nucleotide 7626, and variants IDs 7–9 were characterized by deletion sites at position 7681–7683. No insertion mutation sites were identified.

Phylogenetic analysis

Phylogenetic analysis showed that the 10 variants clustered into two main groups, corresponding to lineages A and B (Fig. 2). In total, eight variants identified in 37 of the 41 HPV-52 partial LCRs analyzed (37/41; 90.2%) clustered within lineage A (formerly European lineage) and 2 variants (4/41; 9.6%) clustered within lineage B (Asian-American lineage).

SNPs in the transcription factor binding sites

On analyzing the SNPs, it was observed that none of them fell within the HPV-52 LCR binding sites of transcription factors.

LCR variants and cytology

The 17 LCR sequences isolated from subjects with cytological abnormalities, 13 LSIL and 4 HSIL, clustered into lineage A.

HPV-16 and HPV-18 L1
HPV-18 L1 variants

In total, 48 HPV-18 partial L1 regions were successfully sequenced. The phylogenetic analysis showed that 24 different variants were identified in one or more of the study samples (variant IDs 1 to 24; Table I).

These variants clustered into three main groups, corresponding to lineages A, B, and C (Fig. 3). More specifically, 19 variants, representing 43 samples (43/48; 89.6%), clustered into lineage A (European lineage), while four variants (4/48; 8.3%) clustered into lineage B and one variant (1/48; 2.1%) clustered into lineage C (both African lineage). All except one of these non-European HPV-18 variants were detected in HIV+ subjects; the exception was detected in a subject in the adolescent/young people age group.

Amino acid changes in the immune-dominant epitopes

A total of 148 HPV-16 partial L1 open reading frames were successfully sequenced. On comparing the sequences analyzed with the reference sequence (accession number, K02718 A1), 33 amino acidic changes were identified among the 330 amino acids of the HPV-16 L1 protein (Table SIV), resulting in 31 unique variants identified in one or more of the study samples (variant IDs 1 to 31; Table I). In total, 10 of these amino acid changes (10/33; 30.3%) occurred in sequences encoding immune-dominant loops. More specifically, the H76Y mutation occurred in the BC loop, and the amino acid mutations A139E, T176N and N177T occurred in the DE loop, H202D and Q214H fell into the EF loop, while mutations A287T, P293S, T294S and Q314R were observed in the FG loop.

In total, 96.7% (30/31) of the identified variants (140 sequences) showed >1 amino acid substitution in an immune-dominant loop. In particular, 7 (variant IDs 2, 8, 11, 16, 19 and 29) and 5 (variant IDs 4–7 and 9) were characterized by two and three amino acid substitutions in the immune-dominant loop, respectively.

On comparing the analyzed sequences with the reference sequence (accession number, AY262282 A1), a total of 15 amino acidic substitutions were identified among the 441 amino acids of the HPV-18 L1 protein (Table SV). No insertion or deletion mutation sites were found. In total, four (4/15; 26.7%) of these amino acid substitutions occurred in sequences encoding immune-dominant loops. More specifically, the R112K mutation occurred in the BC loop, while the amino acid mutations Q334P, I338L and R344P occurred in the FG loop.

In total, 29.2% (7/24) of the variants identified (14 sequences) showed at>1 amino acid substitution in an immune-dominant loop.

Selective pressure analysis

MEME found evidence of episodic positive selection at one site of HPV-16 L1 (P<0.1), while there was no evidence for positive selection in the analyzed sequence alignment of HPV-18. The integrative selection analysis (SLAC, P=0.1; FEL, P=0.1 and REL Bayes factor=50) identified eight negatively selected codons in HPV-16 L1 sequences (55, 93, 109, 130, 145, 216, 308 and 351), none of which fell into the immune-dominant loops, and 23 negatively selected codons in HPV-18 L1 sequences (85, 100, 126, 135, 144, 165, 168, 171, 191, 196, 201, 234, 239, 252, 296, 324, 369, 399, 405, 416, 430, 500 and 519), nine of which fell into four different immune-dominant loops (loop BC, 126; loop DE, 171, 191, 196, 201; loop EF, 234 and 239; loop HI, 416) but none caused amino acid changes.

Discussion

The present study identified and analyzed a large number of HPV-16, HPV-18 and HPV-52 sequences (n=458) obtained from subjects belonging to both general and high-risk populations in Italy. Furthermore, to the best of our knowledge, this is the first Italian study reporting phylogenetic data on HPV-52 variants.

The phylogenetic analysis showed that ~90% of HPV-16 (90.5%), HPV-18 (89.6%) and HPV-52 (90.2%) variants clustered into lineage A, previously defined as the European lineage, as expected for the geographical area under study.

Non-European variants (belonging to lineages B, C and D) were only detected in populations at higher risk of HPV infection, such as migrants, HIV+ subjects and adolescent/young people. In particular, 53% of the non-European HPV-16 sequences were detected in migrant women, 29% in HIV+ subjects and 18% in adolescent/young people. With regards to the five non-European HPV-18 sequences, four were detected in HIV+ subjects, while one was found in an adolescent. Non-European HPV-52 sequences were all identified in HIV+ subjects. The identification of non-European variants in populations engaging in risky sexual behavior is associated with a high risk of contracting several HPV infections supported by different types/variants (1315,17,18,26).

The LCR region of HPV-16 and HPV-52 types showed high levels of genetic diversity (10.9 and 2.9%, respectively) and a large number of new non-lineage-specific SNPs (50 and 31.6%, respectively) were identified in the sequences analyzed. Furthermore, HPV-52 LCR sequences were characterized by various sequence deletion sites. However, at present, these SNPs and deletions have not shown evidence of determining phylogenetic groups, therefore, functional studies are required to clarify whether these changes can affect any HPV variant phenotype.

With regards to the HPV-16 LCR sequences, 23.7% (n=19) of the identified SNPs fell within the binding sites for cellular and viral transcriptional factors, such as E2, YY1, AP-1, Oct-1, NF-1, and TEF-1. Sequence changes in these sites can alter carcinogenic potential by modulating virus replication and transcription (9,27). In particular, SNPs at E2 transcription factor sites may affect the repression of E6/E7 oncoproteins, known as landmarks in cancer progression, and multiple disruptions to YY1 binding sites, such as the six SNPs identified in the sequences analyzed in the present study, are required to significantly upregulate E6 promoter activity by three- to six-fold (27). The results of the present study showed that several mutations in transcription factor sites characterized variants belonging to the African lineages B and C isolated from subjects with HSIL. However, it is important to carry out a thorough transcriptional analysis of the HPV-16 LCR in order to investigate the association between LCR SNPs and the carcinogenicity of HPV-16 variants.

A sequence analysis of the HPV-16 and HPV-18 L1 proteins determined that ~30% of the amino acid mutations fell within the immune-dominant epitope loops (30.3 and 26.7%, respectively). Although these mutations were neutral and not under positive selection, HR-HPVs use this strategy to evade recognition by neutralizing antibodies. In fact, with respect to HPV-16, six out of 10 identified amino acid mutations (A139E, N181S, A266T, G28 1W, S282P and T353P) were involved in the loss of reactivity of a specific monoclonal antibody (MAb), due to the loss of direct interactions with the MAb and for the conformational changes that indirectly disrupt interactions with the Mab (28). Therefore, the real-time monitoring of mutations is essential in this post-vaccination era and it is important to highlight their ability to fix in the viral population. In fact, L1 proteins self-assemble into VLPs, which are components of prophylactic vaccines, and amino acid mutations in the L1 protein may affect viral antigenicity and limit vaccine efficacy (3).

A limitation of the present study was that the HPV-16, HPV-18 and HPV-52 genomes were only partially analyzed. Complete sequencing of the genome would provide better insights into the different HPV types, thus enhancing the phylogenetic classification of HPV in relation to oncogenic risk (29). However, the present study provided, to the best of our knowledge, the first data on the circulation and characterization of HPV-52 variants in Italy. Due to the ongoing implementation of vaccination programs with the 9vHPV vaccine, which also include the HPV-52 type, it is important to monitor all HR-HPV variants, especially in vaccinated populations. In fact, awareness of the viruses currently circulating in the population will facilitate the medium- to long-term monitoring of genetic viral evolution, thus enabling predictions about the potential of evading the vaccine-induced immune response.

As the study of HR-HPV variants is important for understanding the pathogenic role of the virus in malignant lesions, well-designed and extensive epidemiological and clinical studies are required in order to better determine the strength of the correlation between the cytology and the identified SNPs, and between the amino acid mutations and the oncogenic risks.

Supplementary Material

Supporting Data
Supporting Data
Supporting Data
Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

The sequence datasets analyzed in the present study are available at the NCBI GenBank database (https://www.ncbi.nlm.nih.gov/nucleotide/).

Authors' contributions

ERF, AA and ET conceived and designed the experiments and wrote the manuscript. SB and DC performed the experiments. SB, ERF, FP and GZ analyzed the data. GZ supervised the findings. ET supervised the study. SB and FP critically evaluated the literature and revised the manuscript. All of the authors reviewed and approved the final manuscript.

Ethics approval and consent to participate

The approval from the ethical committee was obtained for each of the previous studies. No subjects were enrolled in the present study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Serrano B, Brotons M, Bosch FX and Bruni L: Epidemiology and burden of HPV-related disease. Best Pract Res Clin Obstet Gynaecol. 47:14–26. 2018. View Article : Google Scholar : PubMed/NCBI

2 

De Martel C, Plummer M, Vignat J and Franceschi S: Worldwide burden of cancer attributable to HPV by site, country and HPV type. Int J Cancer. 141:664–670. 2017. View Article : Google Scholar : PubMed/NCBI

3 

Westrich JA, Warren CJ and Pyeon D: Evasion of the host immune defences by human papillomavirus. Virus Res. 231:21–33. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Burk RD, Harari A and Chen Z: Human papillomavirus genome variants. Virology. 445:232–243. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Smith B, Chen Z, Reimers L, van Doorslaer K, Schiffman M, Desalle R, Herrero R, Yu K, Wacholder S, Wang T and Burk RD: Sequence imputation of HPV16 genomes for genetic association studies. PLoS One. 6:e213752011. View Article : Google Scholar : PubMed/NCBI

6 

Mammas IN, Spandidos DA and Sourvinos G: Genomic diversity of human papillomaviruses (HPV) and clinical implications: An overview in adulthood and childhood. Infect Genet Evol. 21:220–226. 2014. View Article : Google Scholar : PubMed/NCBI

7 

Cornet I, Gheit T, Franceschi S, Vignat J, Burk RD, Sylla BS, Tommasino M and Clifford GM; IARC HPV Variant Study Group, : Human papillomavirus type 16 genetic variants: Phylogeny and classification based on E6 and LCR. J Virol. 86:6855–6861. 2012. View Article : Google Scholar : PubMed/NCBI

8 

Chen AA, Gheit T, Franceschi S, Tommasino M and Clifford GM: Human papillomavirus 18 genetic variation and cervical cancer risk worldwide. J Virol. 89:10680–10687. 2015. View Article : Google Scholar : PubMed/NCBI

9 

O'Connor M, Chan SY and Bernard HU: Transcription factor binding sites in the long control region of genital HPVs. Human papillomaviruses. A compilation and analysis of nucleic acid and amino acid sequences Los Alamos, New Mexico: Los Alamos National Laboratory; pp. III21–III41. 1995

10 

Kirnbauer R, Booy F, Cheng N, Lowy DR and Schiller JT: Papillomavirus L1 major capsid protein self-assembles into virus-like particles that are highly immunogenic. Proc Natl Acad Sci USA. 89:12180–12184. 1992. View Article : Google Scholar : PubMed/NCBI

11 

Christensen ND, Dillner J, Eklund C, Carter JJ, Wipf GC, Reed CA, Cladel NM and Galloway DA: Surface conformational and linear epitopes on HPV-16 and HPV-18 L1 virus-like particles as defined by monoclonal antibodies. Virology. 223:174–184. 1996. View Article : Google Scholar : PubMed/NCBI

12 

Martins AE, Lucena-Silva N, Garcia RG, Welkovic S, Barboza A, Menezes ML, Maruza M, Tenorio T and Ximenes RA: Prevalence of human papillomavirus infection, distribution of viral types and risk factors in cervical samples from human immunodeficiency virus-positive women attending three human immunodeficiency virus-acquired immune deficiency syndrome reference centres in northeastern Brazil. Mem Inst Oswaldo Cruz. 109:738–747. 2014. View Article : Google Scholar : PubMed/NCBI

13 

Bianchi S, Boveri S, Igidbashian S, Amendola A, Urbinati AM, Frati ER, Bottari F, Colzani D, Landoni F, Tanzi E, et al: Chlamydia trachomatis infection and HPV/Chlamydia trachomatis co-infection among HPV-vaccinated young women at the beginning of their sexual activity. Arch Gynecol Obstet. 294:1227–1233. 2016. View Article : Google Scholar : PubMed/NCBI

14 

Panatto D, Amicizia D, Bianchi S, Frati ER, Zotti CM, Lai PL, Domnich A, Colzani D, Gasparini R and Tanzi E: Chlamydia trachomatis prevalence and chlamydial/HPV co-infection among HPV-unvaccinated young Italian females with normal cytology. Hum Vaccin Immunother. 11:270–276. 2015. View Article : Google Scholar : PubMed/NCBI

15 

Orlando G, Fasolo M, Mazza F, Ricci E, Esposito S, Frati E, Zuccotti GV, Cetin I, Gramegna M, Rizzardini G, et al: Risk of cervical HPV infection and prevalence of vaccine-type and other high-risk HPV types among sexually active teens and young women (13–26 years) enrolled in the VALHIDATE study. Hum Vaccin Immunother. 10:986–994. 2014. View Article : Google Scholar : PubMed/NCBI

16 

Orlando G, Tanzi E, Rizzardini G, Chatenoud L, Zanchetta N, Esposito S, Tisi G, Fasolo M, Bosari S, Boero V, et al: Modifiable and Non-modifiable factors related to HPV infection and cervical abnormalities in women at high risk: A cross-sectional analysis from the VALHIDATE Study. Ann Virol Res. 2:10132016.

17 

Tanzi E, Amendola A, Bianchi S, Fasolo MM, Beretta R, Pariani E, Zappa A, Frati E and Orlando G: Human papillomavirus genotypes and phylogenetic analysis of HPV-16 variants in HIV-1 infected subjects in Italy. Vaccine. 27 (Suppl 1):A17–A23. 2009. View Article : Google Scholar : PubMed/NCBI

18 

Zhang C, Park JS, Grce M, Hibbitts S, Palefsky JM, Konno R, Smith-McCune KK, Giovannelli L, Chu TY, Picconi MA, et al: Geographical distribution and risk association of human papillomavirus genotype 52-variant lineages. J Infect Dis. 210:1600–1604. 2014. View Article : Google Scholar : PubMed/NCBI

19 

Frati ER, Bianchi S, Colzani D, Zappa A, Orlando G and Tanzi E: Genetic variability in the major capsid L1 protein of human papillomavirus type 16 (HPV-16) and 18 (HPV-18). Infect Genet Evol. 11:2119–2124. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Larkin MA, Blackshields G, Brown NP, Chenna R, McGettigan PA, McWilliam H, Valentin F, Wallace IM, Wilm A, Lopez R, et al: Clustal W and Clustal X version 2.0. Bioinformatics. 23:2947–2948. 2007. View Article : Google Scholar : PubMed/NCBI

21 

Tamura K, Stecher G, Peterson D, Filipski A and Kumar S: MEGA6: Molecular evolutionary genetics analysis version 6.0. Mol Biol Evol. 30:2725–2729. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Hall TA: BioEdit: A user-friendly biological sequence alignment editor and analysis program for Windows 95/98/NT. Nucl Acids Symp. 41:95–98. 1999.

23 

Saitou N and Nei M: The neighbor-joining method: A new method for reconstructing phylogenetic trees. Mol Biol Evol. 4:406–425. 1987.PubMed/NCBI

24 

Kimura M: A simple method for estimating evolutionary rate of base substitutions through comparative studies of nucleotide sequences. J Mol Evol. 16:111–120. 1980. View Article : Google Scholar : PubMed/NCBI

25 

Weaver S, Shank SD, Spielman SJ, Li M, Muse SV and Kosakovsky Pond SL: Datamonkey 2.0: A modern web application for characterizing selective and other evolutionary processes. Mol Biol Evol. Jan 2–2018.doi: 10.1093/molbev/msx335 (Epub ahead of print). View Article : Google Scholar

26 

Frati ER, Fasoli E, Martinelli M, Colzani D, Bianchi S, Carnelli L, Amendola A, Olivani P and Tanzi E: Sexually transmitted infections: A novel screening strategy for improving women's health in vulnerable populations. Int J Mol Sci. 18:E13112017. View Article : Google Scholar : PubMed/NCBI

27 

Marongiu L, Godi A, Parry JV and Beddows S: Human papillomavirus type 16 long control region and E6 variants stratified by cervical disease stage. Infect Genet Evol. 26:8–13. 2014. View Article : Google Scholar : PubMed/NCBI

28 

Ning T, Wolfe A, Nie J, Huang W, Chen XS and Wang Y: Naturally occurring single amino acid substitution in the L1 major capsid protein of human papillomavirus type 16: Alteration of susceptibility to antibody-mediated neutralization. J Infect Dis. 216:867–876. 2017. View Article : Google Scholar : PubMed/NCBI

29 

Van der Weele P, Meijer CJLM and King AJ: Whole-genome sequencing and variant analysis of human papillomavirus 16 infections. J Virol. 91(pii): e00844–17. 2017.PubMed/NCBI

Related Articles

Journal Cover

February-2020
Volume 21 Issue 2

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Frati ER, Bianchi S, Amendola A, Colzani D, Petrelli F, Zehender G and Tanzi E: Genetic characterization of variants of HPV‑16, HPV‑18 and HPV‑52 circulating in Italy among general and high‑risk populations. Mol Med Rep 21: 894-902, 2020
APA
Frati, E.R., Bianchi, S., Amendola, A., Colzani, D., Petrelli, F., Zehender, G., & Tanzi, E. (2020). Genetic characterization of variants of HPV‑16, HPV‑18 and HPV‑52 circulating in Italy among general and high‑risk populations. Molecular Medicine Reports, 21, 894-902. https://doi.org/10.3892/mmr.2019.10847
MLA
Frati, E. R., Bianchi, S., Amendola, A., Colzani, D., Petrelli, F., Zehender, G., Tanzi, E."Genetic characterization of variants of HPV‑16, HPV‑18 and HPV‑52 circulating in Italy among general and high‑risk populations". Molecular Medicine Reports 21.2 (2020): 894-902.
Chicago
Frati, E. R., Bianchi, S., Amendola, A., Colzani, D., Petrelli, F., Zehender, G., Tanzi, E."Genetic characterization of variants of HPV‑16, HPV‑18 and HPV‑52 circulating in Italy among general and high‑risk populations". Molecular Medicine Reports 21, no. 2 (2020): 894-902. https://doi.org/10.3892/mmr.2019.10847