ABSTRACT
In 2017, cervical cancer screening in the Netherlands switched from cytology to human papillomavirus (HPV) testing using the validated PCR-based cobas 4800. Women could order and subsequently received a free self-sampling kit (Evalyn Brush) at their home address instead of clinician sampling. In the laboratory, the shipped brush was placed into 20 mL of PreservCyt fluid, before testing. In the first 2 years of the new program, only 7% of screening tests were performed on a self-sample. Those who chose self-sampling versus clinician sampling were more likely to have never been screened previously and differed also with respect to sociodemographic factors. Subsequent more active promotion and increasing the ease to obtain kits increased the proportion opting for self-sampling (16% in 2020). HPV positivity and detection rate of precancer (CIN3+) were lower in the self-sampling compared with the clinician-sampling group (adjusted ORs of 0.65 and 0.86, respectively). Although population differences may partially explain these results, self-samples may have been too dilute, thereby reducing the analytic and clinical sensitivity. The Dutch findings demonstrate the importance of optimizing outreach, specimen handling and testing protocols for self-samples to effectively screen the target population and reach in particular the women at highest risk for cervical cancer. See related article by Aitken et al., p. 183.
Subject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/virology , Early Detection of Cancer/methods , Netherlands/epidemiology , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , Specimen Handling/methods , Mass Screening/methods , Papillomaviridae/geneticsABSTRACT
The expression of human papillomavirus (HPV) oncoproteins perturbed multiple cellular events of the host cells, leading to the formation of cancer phenotypes. Our current and previous studies indicated that Aurora kinase A (AurA), a mitotic regulator that is often aberrantly expressed in human cancers, is preferentially bound to E6-encoded by cancer-causing HPV. AurA is believed to be important for the proliferation and survival of HPV-positive cells. Nonetheless, the interaction between AurA and E6, and the mechanism of how this association is involved in carcinogenesis, have not been elucidated clearly. Hence, we performed a series of biochemical assays to characterize the AurA-E6 association and complex formation. We found the C-terminus of E6, upstream of the PDZ binding motif of E6, is important to forming the AurA-E6 complex in the nucleus. We also showed that the expression level of E6 corresponded positively with AurA expression. Meanwhile, the functional consequences of the AurA-E6 association to AurA kinase function and host cellular events were also delineated. Intriguingly, we revealed that AurA-E6 association regulated the expression of cyclin E and phosphor-Histone H3, which are involved in G1/S and mitotic phases of the cell cycle, respectively. Depletion of AurA also reduced the invasive ability of HPV-positive cells. AurA inhibition may not be sufficient to reduce the oncogenic potential exerted by E6. Altogether, our study unleashed the mechanism of how HPVE6 deploy AurA to promote cancer phenotypes, particularly through dysregulation of cell cycle checkpoints and suggests that the AurA-E6 complex possesses a therapeutic value. IMPORTANCE We unveiled the mechanism of how HPV employs Aurora kinase A (AurA) of host cells to exert its oncogenic capability synergistically. We systematically characterized the mode of interaction between E6-encoded by cancer-causing HPV and AurA. Then, we delineated the consequences of AurA-E6 complex formation on AurA kinase function and changes to cellular events at molecular levels. Using a cell-based approach, we unleashed that disruption of AurA-E6 association can halt cancer phenotype exhibited by HPV-positive cancer cells. Our findings are vital for the designing of state-of-the-art therapies for HPV-associated cancers.
Subject(s)
Aurora Kinase A , Human Papillomavirus Viruses , Neoplasms , Papillomavirus Infections , Viral Envelope Proteins , Humans , Aurora Kinase A/genetics , Aurora Kinase A/metabolism , Carcinogenesis/pathology , Human Papillomavirus Viruses/genetics , Human Papillomavirus Viruses/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Viral Envelope Proteins/metabolism , Gene Expression Regulation, Viral , Neoplasms/etiology , Neoplasms/physiopathology , Neoplasms/virologyABSTRACT
The causal relationship between HPV and cervical cancer in association with the high prevalence of high risk HPV genotypes led to the design of HPV vaccines based on the major capsid L1 protein. In recent years, capsid protein L2 has also become a focal point in the field of vaccine research. The present review focuses on the variability of HPV16 L1 and L2 genes, emphasizing the distribution of specific amino acid changes in the epitopes of capsid proteins. Moreover, a substantial bioinformatics analysis was conducted to describe the worldwide distribution of amino acid substitutions throughout HPV16 L1, L2 proteins. Five amino acid changes (T176N, N181T; EF loop), (T266A; FG loop), (T353P, T389S; HI loop) are frequently observed in the L1 hypervariable surface loops, while two amino acid substitutions (D43E, S122P) are adjacent to L2 specific epitopes. These changes have a high prevalence in certain geographic regions. The present review suggests that the extensive analysis of the amino acid substitutions in the HPV16 L1 immunodominant loops may provide insights concerning the ability of the virus in evading host immune response in certain populations. The genetic variability of the HPV16 L1 and L2 epitopes should be extensively analyzed in a given population.
Subject(s)
Capsid Proteins , Oncogene Proteins, Viral , Humans , Amino Acids/genetics , Antibodies, Viral , Capsid Proteins/genetics , Epitopes , Human papillomavirus 16/genetics , Mutation , Oncogene Proteins, Viral/genetics , Papillomavirus Infections/virologyABSTRACT
During entry, human papillomavirus (HPV) traffics from the cell surface to the endosome and then to the trans-Golgi network (TGN) and Golgi apparatus. HPV must transit across the TGN/Golgi and exit these compartments to reach the nucleus to cause infection, although how these steps are accomplished is unclear. Combining cellular fractionation, unbiased proteomics, and gene knockdown strategies, we identified the coat protein complex I (COPI), a highly conserved protein complex that facilitates retrograde trafficking of cellular cargos, as a host factor required for HPV infection. Upon TGN/Golgi arrival, the cytoplasmic segment of HPV L2 binds directly to COPI. COPI depletion causes the accumulation of HPV in the TGN/Golgi, resembling the fate of a COPI binding-defective L2 mutant. We propose that the L2-COPI interaction drives HPV trafficking through the TGN and Golgi stacks during virus entry. This shows that an incoming virus is a cargo of the COPI complex.
Subject(s)
Coat Protein Complex I , Human Papillomavirus Viruses , Papillomavirus Infections , Virus Internalization , Humans , Coat Protein Complex I/genetics , Coat Protein Complex I/metabolism , Human Papillomavirus Viruses/physiology , Papillomavirus Infections/virology , Protein TransportABSTRACT
High-risk-human papillomavirus (HR-HPV)-induced cervical cancer is the second most common cause of death among females worldwide. HPV16 is the most prevalent HR-HPV infection worldwide. This study found the genotypic distribution of HR-HPV in the local population and investigated the sequence variations among the E6 and E7 oncogenes of the local HPV16 genotype to the E6 and E7 oncogenes of the foreign HPV16 genotypes and constructed a phylogenetic relationship based on nucleotide sequence comparison among the variants identified in our study along with previously reported isolates that were obtained from different regions of the world. The samples were collected from patients with cervical cancer. Genomic DNA was extracted, and HR-HPV genotypes were determined using real-time PCR. The HPV16 E6 and E7 genes were amplified and sequenced. A HPV16 phylogenetic tree was constructed using the maximum likelihood method with MEGA 7. HPV16 was the most prevalent human papillomavirus (HPV) type identified in the present study. HPV16 isolates belonged to the A1 sublineage of the European branch. Twenty-one nucleotide sequences were included in this analysis. The first, second, and third codon positions are also included. The final dataset included 776 positions.
Subject(s)
Human Papillomavirus Viruses , Oncogene Proteins, Viral , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Genotype , Human papillomavirus 16/genetics , Human Papillomavirus Viruses/genetics , Oncogene Proteins, Viral/genetics , Pakistan/epidemiology , Papillomavirus E7 Proteins/genetics , Papillomavirus Infections/epidemiology , Papillomavirus Infections/virology , PhylogenyABSTRACT
SUMMARY: Chronic infection by several "high-risk" human papillomavirus (HPV) types has been causally implicated in several forms of anogenital and oropharyngeal cancers. Now, HPV42, which is usually classified as a "low-risk" type, can be listed as the main cause of digital papillary adenocarcinoma, an uncommon malignant tumor of the fingers and toes. See related article by Leiendecker et al., p. 70 (3).
Subject(s)
Adenocarcinoma, Papillary , Bone Neoplasms , Breast Neoplasms , Oropharyngeal Neoplasms , Papillomavirus Infections , Humans , Female , Human Papillomavirus Viruses , Papillomavirus Infections/complications , Papillomavirus Infections/virology , Oropharyngeal Neoplasms/virology , Germ Cells/pathologyABSTRACT
BACKGROUND: Cervical cancer is a preventable disease. This study aimed to share the results of the national cervical cancer screening program performed in primary health care institutions in Samsun between 2015 and 2019. METHODS: Women aged 30-65 years who were screened for cervical cancer in screening centers of Samsun between January 01, 2015, and December 31, 2019, were included in this descriptive study. The data were obtained from the automation program of the "National Human Papilloma Virus (HPV) Laboratory Application" used by the Provincial Directorate of Health Cancer Unit through filtering the completion time of the tests, and all results were evaluated without sampling. Thus, data were presented using descriptive statistics. RESULTS: The mean age of 89,302 women included in the cervical cancer screening program was 45.9 ± 9.0 years. Of the samples obtained from the participants, 1.0% were determined as insufficient material, 94.1% as HPV-negative, and 4.9% as HPV-positive. The most common HPV genotypes were 16, 51, 31, and 52. Of the 4337 HPV-positive women, 74.7% of the pap smear results were negative (including infection, 36.5%), and the most common premalignant lesions were atypical squamous cells of undetermined significance in 7.1% and low-grade squamous intraepithelial lesions in 6.9%. HPV 16/18 was also observed in 31.7% of HPV-positive women. Seven hundred ninety-five women were referred to a specialist physician for further examination and treatment within the scope of the screening algorithm. CONCLUSION: Detecting HPV-positivity by reaching more women within the national cervical cancer screening program's scope is vital in fighting against this disease. The effectiveness of cancer screening programs should be increased by ensuring community participation through awareness activities.
Subject(s)
Early Detection of Cancer , Papillomaviridae , Uterine Cervical Neoplasms , Adult , Aged , Early Detection of Cancer/methods , Female , Human papillomavirus 16/isolation & purification , Human papillomavirus 18/isolation & purification , Humans , Middle Aged , National Health Programs , Papanicolaou Test , Papillomaviridae/genetics , Papillomaviridae/isolation & purification , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , Program Evaluation , Turkey , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/prevention & control , Vaginal SmearsABSTRACT
Human papillomavirus 16 (HPV16) E2 is a DNA-binding protein that regulates transcription, replication and potentially, segregation of the HPV16 genome during the viral life cycle. In the segregation model, E2 simultaneously binds to viral and host chromatin, acting as a bridge to ensure that viral genomes reside in daughter nuclei following cell division. The host chromatin receptor for E2 mediating this function is unknown. Recently, we demonstrated that CK2 phosphorylation of E2 on serine 23 (S23) is required for interaction with TopBP1, and that this interaction promotes E2 and TopBP1 recruitment to mitotic chromatin. Here, we demonstrate that in U2OS cells expressing wild-type E2 and a non-TopBP1-binding mutant (S23A, serine 23 mutated to alanine), interaction with TopBP1 is essential for E2 recruitment of plasmids to mitotic chromatin. Using novel quantitative segregation assays, we demonstrate that interaction with TopBP1 is required for E2 plasmid segregation function in U2OS and N/Tert-1 cells. Small interfering RNA (siRNA) knockdown of TopBP1 or CK2 enzyme components disrupts E2 segregation/retention function. The interaction of E2 with TopBP1 promotes increased levels of E2 protein during mitosis in U2OS and N/Tert-1 cells, as well as in human foreskin keratinocytes (HFK) immortalized by the HPV16 genome. Overall, our results demonstrate that E2 has plasmid segregation activity, and that the E2-TopBP1 interaction is essential for this E2 function. IMPORTANCE HPV16 causes 3% to 4% of all human cancers. It is proposed that during the viral life cycle, the viral genome is actively segregated into daughter nuclei, ensuring viral replication in the subsequent S phase. The E2 protein potentially bridges the viral and host genomes during mitosis to mediate segregation of the circular viral plasmid. Here, we demonstrate that E2 has the ability to mediate plasmid segregation, and that this function is dependent upon interaction with the host protein TopBP1. Additionally, we demonstrate that the E2-TopBP1 interaction promotes enhanced E2 expression during mitosis, which likely promotes the plasmid segregation function of E2. Overall, our results present a mechanism of how HPV16 can segregate its viral genome during an active infection, a critical aspect of the viral life cycle.
Subject(s)
DNA-Binding Proteins/metabolism , Human papillomavirus 16/physiology , Mitosis , Nuclear Proteins/metabolism , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/pathology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chromatin/metabolism , DNA-Binding Proteins/genetics , Genome, Viral , Humans , Papillomavirus Infections/metabolism , Papillomavirus Infections/virology , Plasmids/geneticsABSTRACT
Lack of prospectively planned follow-up and minimal characterization of the patient population studied complicate interpretation of circulating human papillomavirus (HPV) DNA as a prognostic biomarker for patients with HPV-associated oropharyngeal carcinoma treated with curative intent. See related article by Berger et al., p. 4292.
Subject(s)
Carcinoma, Squamous Cell , Oropharyngeal Neoplasms , Papillomavirus Infections , Biomarkers , Carcinoma, Squamous Cell/pathology , DNA , Humans , Oropharyngeal Neoplasms/diagnosis , Oropharyngeal Neoplasms/pathology , Papillomavirus Infections/complications , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , PrognosisABSTRACT
Human papillomaviruses (HPVs) infect the basal proliferating cells of the stratified epithelium, but the productive phase of the life cycle (consisting of viral genome amplification, late gene expression, and virion assembly) is restricted to the highly differentiated suprabasal cells. While much is known regarding the mechanisms that HPVs use to block activation of an innate immune response in undifferentiated cells, little is known concerning how HPV prevents an interferon (IFN) response upon differentiation. Here, we demonstrate that high-risk HPVs hijack a natural function of apoptotic caspases to suppress an IFN response in differentiating epithelial cells. We show that caspase inhibition results in the secretion of type I and type III IFNs that can act in a paracrine manner to induce expression of interferon-stimulated genes (ISGs) and block productive replication of HPV31. Importantly, we demonstrate that the expression of IFNs is triggered by the melanoma differentiation-associated gene 5 (MDA5)-mitochondrial antiviral-signaling protein (MAVS)-TBK1 (TANK-binding kinase 1) pathway, signifying a response to double-stranded RNA (dsRNA). Additionally, we identify a role for MDA5 and MAVS in restricting productive viral replication during the normal HPV life cycle. This study identifies a mechanism by which HPV reprograms the cellular environment of differentiating cells through caspase activation, co-opting a nondeath function of proteins normally involved in apoptosis to block antiviral signaling and promote viral replication.
Subject(s)
Caspases , Human papillomavirus 31 , Interferon-Induced Helicase, IFIH1 , Interferons , Papillomavirus Infections , Virus Replication , Caspases/metabolism , Human papillomavirus 31/physiology , Humans , Interferon-Induced Helicase, IFIH1/metabolism , Interferons/metabolism , Papillomavirus Infections/immunology , Papillomavirus Infections/virologyABSTRACT
BACKGROUND: To investigate the differences in HPV genotypes and clinical indicators between cervical squamous cell carcinoma and adenocarcinoma and to identify independent predictors for differentiating cervical squamous cell carcinoma and adenocarcinoma. METHODS: A total of 319 patients with cervical cancer, including 238 patients with squamous cell carcinoma and 81 patients with adenocarcinoma, were retrospectively analysed. The clinical characteristics and laboratory indicators, including HPV genotypes, SCCAg, CA125, CA19-9, CYFRA 21-1 and parity, were analysed by univariate and multivariate analyses, and a classification model for cervical squamous cell carcinoma and adenocarcinoma was established. The model was validated in 96 patients with cervical cancer. RESULTS: There were significant differences in SCCAg, CA125, CA19-9, CYFRA 21-1, HPV genotypes and clinical symptoms between cervical squamous cell carcinoma and adenocarcinoma (P < 0.05). Logistic regression analysis showed that SCCAg and HPV genotypes (high risk) were independent predictors for differentiating cervical squamous cell carcinoma from adenocarcinoma. The AUC value of the established classification model was 0.854 (95% CI: 0.804-0.904). The accuracy, sensitivity and specificity of the model were 0.846, 0.691 and 0.899, respectively. The classification accuracy was 0.823 when the model was verified. CONCLUSION: The histological type of cervical cancer patients with persistent infection of high-risk HPV subtypes and low serum SCCAg levels was more prone to being adenocarcinoma. When the above independent predictors occur, the occurrence and development of cervical adenocarcinoma should be anticipated, and early active intervention treatment should be used to improve the prognosis and survival of patients.
Subject(s)
Adenocarcinoma , Carcinoma, Squamous Cell , Papillomaviridae , Papillomavirus Infections , Serpins , Uterine Cervical Neoplasms , Antigens, Neoplasm , CA-19-9 Antigen , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/virology , Female , Genotype , Humans , Keratin-19 , Papillomaviridae/genetics , Papillomavirus Infections/virology , Retrospective Studies , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/virologyABSTRACT
OBJECTIVES: To investigate prognostic-related gene signature based on DNA damage repair and tumor microenvironment statue in human papillomavirus 16 negative (HPV16-) head and neck squamous cell carcinoma (HNSCC). METHODS: For the RNA-sequence matrix in HPV16- HNSCC in the Cancer Genome Atlas (TCGA) cohort, the DNA damage response (DDR) and tumor microenvironment (TM) status of each patient sample was estimated by using the ssGSEA algorithm. Through bioinformatics analysis in DDR_high/TM_high (n = 311) and DDR_high/TM_low (n = 53) groups, a survival-related gene signature was selected in the TCGA cohort. Two independent external validation cohorts (GSE65858 (n = 210) and GSE41613 (n = 97)) with HPV16- HNSCC patients validated the gene signature. Correlations among the clinical-related hub differentially expressed genes (DEGs) and infiltrated immunocytes were explored with the TIMER2.0 server. Drug screening based on hub DEGs was performed using the CellMiner and GSCALite databases. The loss-of-function studies were used to evaluate the effect of screened survival-related gene on the motility of HPV- HNSCC cells in vitro. RESULTS: A high DDR level (P = 0.025) and low TM score (P = 0.012) were independent risk factors for HPV16- HNSCC. Downregulated expression of ALOX12B or SPRR1A was associated with poor survival rate and advanced cancer stages. The pathway enrichment analysis showed the DDR_high/TM_low samples were enriched in glycosphingolipid biosynthesis-lacto and neolacto series, glutathione metabolism, platinum drug resistance, and ferroptosis pathways, while the DDR_high/TM_low samples were enriched in Th17 cell differentiation, Neutrophil extracellular trap formation, PD - L1 expression and PD - 1 checkpoint pathway in cancer. Notably, the expression of ALOX12B and SPRR1A were negatively correlated with cancer-associated fibroblasts (CAFs) infiltration and CAFs downstream effectors. Sensitivity to specific chemotherapy regimens can be derived from gene expressions. In addition, ALOX12B and SPRR1A expression was associated with the mRNA expression of insulin like growth factor 1 receptor (IGF1R), AKT serine/threonine kinase 1 (AKT1), mammalian target of rapamycin (MTOR), and eukaryotic translation initiation factor 4E binding protein 1 (EIF4EBP1) in HPV negative HNSCC. Down-regulation of ALOX12B promoted HPV- HNSCC cells migration and invasion in vitro. CONCLUSIONS: ALOX12B and SPRR1A served as a gene signature for overall survival in HPV16- HNSCC patients, and correlated with the amount of infiltrated CAFs. The specific drug pattern was determined by the gene signature.
Subject(s)
Arachidonate 12-Lipoxygenase , Cornified Envelope Proline-Rich Proteins , DNA Repair , Head and Neck Neoplasms , Human papillomavirus 16 , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Arachidonate 12-Lipoxygenase/genetics , Arachidonate 12-Lipoxygenase/metabolism , Cornified Envelope Proline-Rich Proteins/metabolism , DNA Damage , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/virology , Human papillomavirus 16/isolation & purification , Humans , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/virology , Tumor Microenvironment/geneticsABSTRACT
The natural history of cervical cancer is strongly related to the presence of human papillomavirus (HPV) infection, with its relationship with cervical cancer being a matter of concern. It is estimated that 70% of all cervical cancers worldwide are caused by HPV 16 and 18. Accordingly, the present study aimed to contribute to the identification of HPV subtypes circulating in a group of women of Manaus-Brazil. Cervical samples were collected from 49 women, following the eligibility criteria of the study, and DNA was then extracted from the samples, which were analyzed for the presence of the virus in the genetic material through the polymerase chain reaction (PCR) using generic primers (GP05/06). Finally, identification of the viral subtypes was performed using specific primers for the detection of the main subtypes already examined (16 and 18). Positive HPV DNA was detected in 100% of the samples included in the study. Human papillomavirus 16 was the most prevalent subtype in the majority of lesions, accounting for 29 (59.2%) of the positive cases, and HPV 18 was detected in four (8.2%) women. In these 4 cases there was co-infection, with the presence of both HPV 18 and HPV 16. Therefore, 40.8% (20 cases) in which HPV DNA was detected presented infection with other subtypes of HPV not included in the study. This data has clinical implications related to cervical cancer prevention, as the current prophylactic HPV vaccines are only effective against high-risk HPV 16 and 18 subtypes.
Subject(s)
Humans , Female , Adult , Middle Aged , Uterine Cervical Neoplasms/diagnosis , Papillomavirus Infections/diagnosis , Papillomavirus Infections/virology , Women , Colposcopy/instrumentation , Human papillomavirus 16/growth & development , Human papillomavirus 18/growth & development , Papanicolaou Test/instrumentationSubject(s)
Papillomavirus Infections , Uterine Cervical Neoplasms , Early Detection of Cancer , Female , Humans , Papillomavirus Infections/diagnosis , Papillomavirus Infections/prevention & control , Papillomavirus Infections/virology , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , VaccinationABSTRACT
SignificanceHigh-risk (HR) human papillomaviruses (HPV) from the genus alpha cause anogenital and oropharyngeal cancers, whereas the contribution of HPV from the genus beta to the development of cutaneous squamous cell cancer is still under debate. HR-HPV genomes display potent immortalizing activity in human keratinocytes, the natural target cell for HPV. This paper shows that immortalization of keratinocytes by the beta-HPV49 genome requires the inactivation of the viral E8^E2 repressor protein and the presence of the E6 and E7 oncoproteins but also of the E1 and E2 replication proteins. This reveals important differences in the carcinogenic properties of HR-HPV and beta-HPV but also warrants further investigations on the distribution and mutation frequencies of beta-HPV in human cancers.
Subject(s)
Betapapillomavirus/physiology , Gene Expression Regulation, Viral , Host-Pathogen Interactions , Keratinocytes/virology , Papillomavirus Infections/virology , Virus Replication , Cell Line, Transformed , Genome, Viral , Humans , Keratinocytes/metabolism , Oncogene Proteins, Viral/genetics , RNA, ViralABSTRACT
The equine sarcoid is one of the most common neoplasias in the Equidae family. Despite the association of this tumor with the presence of bovine papillomavirus (BPV), the molecular mechanism of this lesion has not been fully understood. The transgenization of equine adult cutaneous fibroblast cells (ACFCs) was accomplished by nucleofection, followed by detection of molecular modifications using high-throughput NGS transcriptome sequencing. The results of the present study confirm that BPV-E4- and BPV-E1^E4-mediated nucleofection strategy significantly affected the transcriptomic alterations, leading to sarcoid-like neoplastic transformation of equine ACFCs. Furthermore, the results of the current investigation might contribute to the creation of in vitro biomedical models suitable for estimating the fates of molecular dedifferentiability and the epigenomic reprogrammability of BPV-E4 and BPV-E4^E1 transgenic equine ACFC-derived sarcoid-like cell nuclei in equine somatic cell-cloned embryos. Additionally, these in vitro models seem to be reliable for thoroughly recognizing molecular mechanisms that underlie not only oncogenic alterations in transcriptomic signatures, but also the etiopathogenesis of epidermal and dermal sarcoid-dependent neoplastic transformations in horses and other equids. For those reasons, the aforementioned transgenic models might be useful for devising clinical treatments in horses afflicted with sarcoid-related neoplasia of cutaneous and subcutaneous tissues.
Subject(s)
Fibroblasts/virology , Horse Diseases/virology , Horses/virology , Neoplasms/virology , Papillomaviridae/genetics , Sarcoidosis/virology , Skin Diseases/virology , Animals , Animals, Genetically Modified/virology , Equidae/virology , Papillomavirus Infections/virology , Skin/virology , Transcriptome/geneticsABSTRACT
Human Papillomaviruses have co-evolved with their human host, with each of the over 200 known HPV types infecting distinct epithelial niches to cause diverse disease pathologies. Despite the success of prophylactic vaccines in preventing high-risk HPV infection, the development of HPV anti-viral therapies has been hampered by the lack of enzymatic viral functions, and by difficulties in translating the results of in vitro experiments into clinically useful treatment regimes. In this review, we discuss recent advances in anti-HPV drug development, and highlight the importance of understanding persistent HPV infections for future anti-viral design. In the infected epithelial basal layer, HPV genomes are maintained at a very low copy number, with only limited viral gene expression; factors which allow them to hide from the host immune system. However, HPV gene expression confers an elevated proliferative potential, a delayed commitment to differentiation, and preferential persistence of the infected cell in the epithelial basal layer, when compared to their uninfected neighbours. To a large extent, this is driven by the viral E6 protein, which functions in the HPV life cycle as a modulator of epithelial homeostasis. By targeting HPV gene products involved in the maintenance of the viral reservoir, there appears to be new opportunities for the control or elimination of chronic HPV infections.
Subject(s)
Alphapapillomavirus/drug effects , Antiviral Agents/therapeutic use , Papillomavirus Infections/drug therapy , Persistent Infection/drug therapy , Antiviral Agents/pharmacology , Drug Development , Epithelium/drug effects , Epithelium/pathology , Epithelium/virology , Homeostasis/drug effects , Humans , Oncogene Proteins, Viral/antagonists & inhibitors , Oncogene Proteins, Viral/metabolism , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , Persistent Infection/pathology , Persistent Infection/virologyABSTRACT
Our fundamental understanding of papillomaviruses and their interactions with their host, including their role in cancer and how the immune system responds to them, has made the elimination of cervical cancer a realistic global health goal [...].
Subject(s)
Papillomaviridae , Papillomavirus Infections , Papillomavirus Vaccines , Carcinogenesis , Humans , Papillomaviridae/physiology , Papillomavirus Infections/physiopathology , Papillomavirus Infections/therapy , Papillomavirus Infections/virologyABSTRACT
This study aimed to explore the changes of the vaginal microbiota and enzymes in the women with high-risk human papillomavirus (HR-HPV) infection and cervical lesions. A total of 448 participants were carried out HPV genotyping, cytology tests, and microecology tests, and 28 participants were treated as sub-samples, in which vaginal samples were characterized by sequencing the bacterial 16S V4 ribosomal RNA (rRNA) gene region. The study found the prevalence of HR-HPV was higher in patients with BV (P = 0.036). The HR-HPV infection rate was 72.73% in G. vaginalis women, which was significantly higher than that of women with lactobacillus as the dominant microbiota (44.72%) (P = 0.04). The positive rate of sialidase (SNA) was higher in women with HR-HPV infection (P = 0.004) and women diagnosed with cervical intraepithelial neoplasia (CIN) (P = 0.041). In HPV (+) women, the α-diversity was significantly higher than that in HPV (-) women. The 16S rRNA gene-based amplicon sequencing results showed that Lactobacillus was the dominant bacteria in the normal vaginal microbiota. However, the proportion of Gardnerella and Prevotella were markedly increased in HPV (+) patients. Gardnerella and Prevotella are the most high-risk combination for the development of HPV (+) women. The SNA secreted by Gardnerella and Prevotella may play a significant role in HPV infection progress to cervical lesions.
Subject(s)
Microbiota/genetics , Papillomavirus Infections/microbiology , Uterine Cervical Dysplasia/microbiology , Vagina/microbiology , Alphapapillomavirus/genetics , Alphapapillomavirus/pathogenicity , Bacteria/classification , Bacteria/genetics , Female , Humans , Lactobacillus/genetics , Neuraminidase/genetics , Papillomavirus Infections/genetics , Papillomavirus Infections/pathology , Papillomavirus Infections/virology , RNA, Ribosomal, 16S/genetics , Vagina/virology , Uterine Cervical Dysplasia/genetics , Uterine Cervical Dysplasia/pathology , Uterine Cervical Dysplasia/virologyABSTRACT
Despite the well-established pathogenic effect of high-risk human papillomavirus (hrHPV) genotypes on endocervical adenocarcinomas (ECAs), the prognostic values of hrHPV genotypes and their association with other prognostic variables have not been established. We categorized 120 usual-type human papillomavirus-associated (HPVA) ECA cases into 3 species groups (HPV16+, HPV18/45+, and other genotypes+) based on the hrHPV status. The clinical-stage, invasion patterns (Silva), and programmed death ligand-1 (PD-L1) expression were compared among genotype groups. In addition, log-rank test and Kaplan-Meier survival curves were used to compare progression-free survival (PFS) among different patient groups. A total of 120 ECA cases with positive hrHPV tests were included in this study. Among them, 51 (42.5%) were positive for HPV16, 50 (41.7%) were positive for HPV18 or 18/45, 9 (7.5%) were positive for other hrHPV genotypes (not including HPV16/18/45). Our data showed patients had no significant difference in clinical stages (P=0.51), invasion patterns (P=0.55), and PFS (P=0.59) across genotype groups. Overall, a relatively high prevalence of PD-L1 expression was observed in HPVA ECAs (25% by tumor proportion score [TPS] and 55% by a combined positive score [CPS]). Using TPS, 19.6% (10/51) HPV16+ cases, 32.0% (16/50) cases of HPV18 or 18/45+ cases, and 22.2% (2/9) cases of other genotypes+ cases demonstrated PD-L1 positivity. No significant difference in PD-L1 expression was seen across genotype groups (P=0.35). PD-L1 expression in tumors with patterns B and C was significantly higher than in those with pattern A (P=0.00002). Patients with PD-L1-positive tumors by either CPS or TPS showed significantly poorer PFS than those with PD-L1-negative tumors (CPS, P=0.025; TPS, P=0.001). Our data support that HPV genotypes have no prognostic value in HPVA ECAs, while PD-L1 expression serves as a negative prognostic marker in HPVA ECAs and implies an unfavorable outcome.