Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 8.409
Filtrer
1.
Virol J ; 21(1): 172, 2024 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-39095779

RÉSUMÉ

Human papillomavirus (HPV) 11/16 E6/E7 proteins have been recognized to be pivotal in viral pathogenesis. This study sought to uncover the potential mechanisms of how HPV11/16 E6/E7-transfected keratinocytes inhibit cytokine secretion in peripheral blood mononuclear cells (PBMC). Upon co-culturing HPV11/16 E6/E7-transfected keratinocytes with PBMC in a non-contact manner, we observed a marked decrease in various cytokines secreted by PBMC. To determine if this suppression was mediated by specific common secreted factors, we conducted transcriptomic sequencing on these transfected cells. This analysis identified 53 common differentially secreted genes in all four HPV-transfected cells. Bioinformatics analysis demonstrated these genes were predominantly involved in immune regulation. Results from quantitative PCR (qPCR) and an extensive literature review suggested the downregulation of 12 genes (ACE2, BMP3, BPIFB1, CLU, CST6, CTF1, HMGB2, MMP12, PDGFA, RNASE7, SULF2, TGM2), and upregulation of 7 genes (CCL17, CCL22, FBLN1, PLAU, S100A7, S100A8, S100A9), may be crucial in modulating tumor immunity and combating pathogenic infections, with genes S100A8 and S100A9, and IL-17 signaling pathway being particularly noteworthy. Thus, HPV11/16 E6/E7 proteins may inhibit cytokine secretion of immune cells by altering the expression of host-secreted genes. Further exploration of these genes may yield new insights into the complex dynamics of HPV infection.


Sujet(s)
Cytokines , Agranulocytes , Protéines des oncogènes viraux , Humains , Cytokines/métabolisme , Cytokines/génétique , Agranulocytes/immunologie , Agranulocytes/virologie , Agranulocytes/métabolisme , Protéines des oncogènes viraux/génétique , Protéines des oncogènes viraux/métabolisme , Protéines des oncogènes viraux/immunologie , Kératinocytes/virologie , Kératinocytes/immunologie , Kératinocytes/métabolisme , Papillomavirus humain de type 16/génétique , Papillomavirus humain de type 16/immunologie , Papillomavirus humain de type 11/génétique , Papillomavirus humain de type 11/immunologie , Analyse de profil d'expression de gènes , Infections à papillomavirus/virologie , Infections à papillomavirus/immunologie , Infections à papillomavirus/génétique , Protéines E7 de papillomavirus/génétique , Protéines E7 de papillomavirus/métabolisme , Protéines E7 de papillomavirus/immunologie , Techniques de coculture , Interactions hôte-pathogène/immunologie , Interactions hôte-pathogène/génétique
2.
Front Cell Infect Microbiol ; 14: 1430424, 2024.
Article de Anglais | MEDLINE | ID: mdl-39104853

RÉSUMÉ

Human papillomaviruses (HPVs) account for more than 30% of cancer cases, with definite identification of the oncogenic role of viral E6 and E7 genes. However, the identification of high-risk HPV genotypes has largely relied on lagged biological exploration and clinical observation, with types unclassified and oncogenicity unknown for many HPVs. In the present study, we retrieved and cleaned HPV sequence records with high quality and analyzed their genomic compositional traits of dinucleotide (DNT) and DNT representation (DCR) to overview the distribution difference among various types of HPVs. Then, a deep learning model was built to predict the oncogenic potential of all HPVs based on E6 and E7 genes. Our results showed that the main three groups of Alpha, Beta, and Gamma HPVs were clearly separated between/among types in the DCR trait for either E6 or E7 coding sequence (CDS) and were clustered within the same group. Moreover, the DCR data of either E6 or E7 were learnable with a convolutional neural network (CNN) model. Either CNN classifier predicted accurately the oncogenicity label of high and low oncogenic HPVs. In summary, the compositional traits of HPV oncogenicity-related genes E6 and E7 were much different between the high and low oncogenic HPVs, and the compositional trait of the DCR-based deep learning classifier predicted the oncogenic phenotype accurately of HPVs. The trained predictor in this study will facilitate the identification of HPV oncogenicity, particularly for those HPVs without clear genotype or phenotype.


Sujet(s)
Apprentissage profond , Génome viral , Papillomaviridae , Infections à papillomavirus , Humains , Infections à papillomavirus/virologie , Papillomaviridae/génétique , Génome viral/génétique , Génotype , Protéines des oncogènes viraux/génétique , Protéines E7 de papillomavirus/génétique , Carcinogenèse/génétique
3.
Infect Dis Obstet Gynecol ; 2024: 6651272, 2024.
Article de Anglais | MEDLINE | ID: mdl-39108464

RÉSUMÉ

Objective: The objective of the study is to validate a new human papillomavirus (HPV) L1 high-risk specific serological assay in a case-control study. Methods: Serum samples of 138 patients (cervical intraepithelial neoplasia (CIN) 1, 2, and 3 and cervical cancer), 21 vaccinees, and 246 female controls were tested for the presence of HPV L1 high-risk specific antibodies. Results: HPV L1 high-risk antibodies were detected in 100% of the CIN1 and 2, 86.6% of the CIN3 and 82.4% of the cervical cancer cases, 100% of the vaccinees, and 3.9% of the female controls. Area under the curve (AUC) was calculated with 0.91 for controls versus CIN2+, 0.923 for controls versus CIN1+, and 0.968 for controls versus CIN1/2. Conclusion: The HPV L1 high-risk specific serological lateral flow rapid test shows promising data in the field of early detection of HPV high-risk induced cervical cancer and its precursor lesions. This easy-to-use, robust, and affordable approach could offer a chance to reach women in low- or middle-income countries (LMICs) that could not be reached by HPV molecular testing-based cervical cancer screening programs.


Sujet(s)
Anticorps antiviraux , Dépistage précoce du cancer , Infections à papillomavirus , Sensibilité et spécificité , Dysplasie du col utérin , Tumeurs du col de l'utérus , Humains , Femelle , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/diagnostic , Études cas-témoins , Adulte , Infections à papillomavirus/diagnostic , Infections à papillomavirus/virologie , Anticorps antiviraux/sang , Dysplasie du col utérin/virologie , Dysplasie du col utérin/diagnostic , Adulte d'âge moyen , Dépistage précoce du cancer/méthodes , Protéines des oncogènes viraux/immunologie , Protéines de capside/immunologie , Jeune adulte , Papillomaviridae/immunologie , Papillomaviridae/isolement et purification , Sujet âgé
4.
Arch Dermatol Res ; 316(7): 455, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38967656

RÉSUMÉ

Tirbanibulin 1% ointment is a synthetic antiproliferative agent approved in 2021 by the European Union for treating actinic keratoses (AK). Topical tirbanibulin has clinically resolved HPV-57 ( +) squamous cell carcinoma (SCC), HPV-16 ( +) vulvar high-grade squamous intraepithelial lesion, epidermodysplasia verruciformis, and condyloma. We examined how tirbanibulin might affect HPV oncoprotein expression and affect other cellular pathways involved in cell proliferation and transformation. We treated the HeLa cell line, containing integrated HPV-18, with increasing doses of tirbanibulin to determine the effects on cell proliferation. Immunoblotting was performed with antibodies against the Src canonical pathway, HPV 18 E6 and E7 transcription regulation, apoptosis, and invasion and metastasis pathways. Cell proliferation assays with tirbanibulin determined the half-maximal inhibitory concentration (IC50) of HeLa cells to be 31.49 nmol/L. Increasing concentrations of tirbanibulin downregulates the protein expression of Src (p < 0.001), phospho-Src (p < 0.001), Ras (p < 0.01), c-Raf (p < 0.001), ERK1 (p < 0.001), phospho-ERK1 (p < 0.001), phospho-ERK2 (p < 0.01), phospho-Mnk1 (p < 0.001), eIF4E (p < 0.01), phospho-eIF4E (p < 0.001), E6 (p < 0.01), E7 (p < 0.01), Rb (p < 0.01), phospho-Rb (p < 0.001), MDM2 (p < 0.01), E2F1 (p < 0.001), phospho-FAK (p < 0.001), phospho-p130 Cas (p < 0.001), Mcl-1 (p < 0.01), and Bcl-2 (p < 0.001), but upregulates cPARP (p < 0.001), and cPARP/fPARP (p < 0.001). These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins via the Src- MEK- pathway. Tirbanibulin significantly downregulates oncogenic proteins related to cell cycle regulation and cell proliferation while upregulating apoptosis pathways.


Tirbanibulin is Promising Novel Therapy for Human Papillomavirus (HPV)-associated Diseases.Tirbanibulin 1% ointment is an approved synthetic topical ointment for treating actinic keratoses (AK), a precancer of skin cancer. Topical tirbanibulin has previously been reported to clinically resolve human papillomavirus (HPV)-( +) diseases.In this study, we examine how tirbanibulin may affect the HPV and pathways associated with cancer.We treated the HeLa cell line to determine the effects on HPV cell proliferation. Increasing the concentration of tirbanibulin statistically significantly affected numerous cellular pathways often associated with cancer.These results demonstrate that tirbanibulin may impact expression of HPV oncoproteins and thereby kill cancer cells.


Sujet(s)
Prolifération cellulaire , Régulation négative , Papillomavirus humain de type 18 , Protéines des oncogènes viraux , Humains , Cellules HeLa , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines des oncogènes viraux/métabolisme , Régulation négative/effets des médicaments et des substances chimiques , Infections à papillomavirus/virologie , Infections à papillomavirus/traitement médicamenteux , Protéines E7 de papillomavirus/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Protéines de répression/métabolisme , Protéines de répression/génétique , Transduction du signal/effets des médicaments et des substances chimiques , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , src-Family kinases/métabolisme , src-Family kinases/antagonistes et inhibiteurs , Femelle , Virus des Papillomavirus humains , Protéines de liaison à l'ADN
5.
BMC Med Genomics ; 17(1): 179, 2024 Jul 04.
Article de Anglais | MEDLINE | ID: mdl-38965538

RÉSUMÉ

BACKGROUND: There is a high incidence of cervical cancer in Xinjiang. Genetic variation in human papillomavirus may increase its ability to invade, spread, and escape host immune response. METHODS: HPV16 genome was sequenced for 90 positive samples of HPV16 infection. Sequences of the E4, E5 and L2 genes were analysed to reveal sequence variation of HPV16 in Xinjiang and the distribution of variation among the positive samples of HPV16 infection. RESULTS: Eighty-one of the 90 samples of HPV16 infection showed variation in HPV16 E4 gene with 18 nucleotide variation sites, of which 8 sites were synonymous variations and 11 missense variations. 90 samples of HPV16 infection showed variation in HPV16 E5 and L2 genes with 16 nucleotide variation sites (6 synonymous, 11 missense variations) in the E5 gene and 100 nucleotide variation sites in L2 gene (37 synonymous, 67 missense variations). The frequency of HPV16 L2 gene missense variations G3377A, G3599A, G3703A, and G3757A was higher in the case groups than in the control groups. CONCLUSIONS: Phylogenetic tree analysis showed that 87 samples were European strains, 3 cases were Asian strains, there were no other variations, and G4181A was related to Asian strains. HPV16 L2 gene missense variations G3377A, G3599A, G3703A, and G3757A were significantly more frequent in the case groups than in the control groups.


Sujet(s)
Variation génétique , Papillomavirus humain de type 16 , Protéines des oncogènes viraux , Infections à papillomavirus , Phylogenèse , Humains , Femelle , Chine , Papillomavirus humain de type 16/génétique , Infections à papillomavirus/virologie , Infections à papillomavirus/génétique , Protéines des oncogènes viraux/génétique , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/génétique , Adulte , Adulte d'âge moyen , Mutation faux-sens
6.
Virol J ; 21(1): 152, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970084

RÉSUMÉ

BACKGROUND: High-risk human papillomavirus (HR-HPV) infection is an important factor for the development of cervical cancer. HPV18 is the second most common HR-HPV after HPV16. METHODS: In this study, MEGA11 software was used to analyze the variation and phylogenetic tree of HPV18 E6-E7 and L1 genes. The selective pressure to E6, E7 and L1 genes was estimated using pamlX. In addition, the B cell epitopes of L1 amino acid sequences and T cell epitopes of E6-E7 amino acid sequences in HPV18 were predicted by ABCpred server and IEDB website, respectively. RESULTS: A total of 9 single nucleotide variants were found in E6-E7 sequences, of which 2 were nonsynonymous variants and 7 were synonymous variants. Twenty single nucleotide variants were identified in L1 sequence, including 11 nonsynonymous variants and 9 synonymous variants. Phylogenetic analysis showed that E6-E7 and L1 sequences were all distributed in A lineage. In HPV18 E6, E7 and L1 sequences, no positively selected site was found. The nonconservative substitution R545C in L1 affected hypothetical B cell epitope. Two nonconservative substitutions, S82A in E6, and R53Q in E7, impacted multiple hypothetical T cell epitopes. CONCLUSION: The sequence variation data of HPV18 may lay a foundation for the virus diagnosis, further study of cervical cancer and vaccine design in central China.


Sujet(s)
Variation génétique , Papillomavirus humain de type 18 , Protéines des oncogènes viraux , Protéines E7 de papillomavirus , Phylogenèse , Protéines des oncogènes viraux/génétique , Chine , Humains , Papillomavirus humain de type 18/génétique , Papillomavirus humain de type 18/classification , Protéines E7 de papillomavirus/génétique , Protéines de capside/génétique , Femelle , Déterminants antigéniques des lymphocytes T/génétique , Infections à papillomavirus/virologie , Protéines de répression/génétique , Déterminants antigéniques des lymphocytes B/génétique , Protéines de liaison à l'ADN
7.
Sci Rep ; 14(1): 17182, 2024 07 26.
Article de Anglais | MEDLINE | ID: mdl-39060289

RÉSUMÉ

Despite therapeutic advancements, cervical cancer caused by high-risk subtypes of the human papillomavirus (HPV) remains a leading cause of cancer-related deaths among women worldwide. This study aimed to discover potential drug candidates from the Asian medicinal plant Andrographis paniculata, demonstrating efficacy against the E6 protein of high-risk HPV-16 subtype through an in-silico computational approach. The 3D structures of 32 compounds (selected from 42) derived from A. paniculata, exhibiting higher binding affinity, were obtained from the PubChem database. These structures underwent subsequent analysis and screening based on criteria including binding energy, molecular docking, drug likeness and toxicity prediction using computational techniques. Considering the spectrometry, pharmacokinetic properties, docking results, drug likeliness, and toxicological effects, five compounds-stigmasterol, 1H-Indole-3-carboxylic acid, 5-methoxy-, methyl ester (AP7), andrographolide, apigenin and wogonin-were selected as the potential inhibitors against the E6 protein of HPV-16. We also performed 200 ns molecular dynamics simulations of the compounds to analyze their stability and interactions as protein-ligand complexes using imiquimod (CID-57469) as a control. Screened compounds showed favorable characteristics, including stable root mean square deviation values, minimal root mean square fluctuations and consistent radius of gyration values. Intermolecular interactions, such as hydrogen bonds and hydrophobic contacts, were sustained throughout the simulations. The compounds displayed potential affinity, as indicated by negative binding free energy values. Overall, findings of this study suggest that the selected compounds have the potential to act as inhibitors against the E6 protein of HPV-16, offering promising prospects for the treatment and management of CC.


Sujet(s)
Andrographis , Papillomavirus humain de type 16 , Simulation de docking moléculaire , Protéines des oncogènes viraux , Tumeurs du col de l'utérus , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/virologie , Humains , Femelle , Protéines des oncogènes viraux/métabolisme , Protéines des oncogènes viraux/composition chimique , Andrographis/composition chimique , Papillomavirus humain de type 16/effets des médicaments et des substances chimiques , Protéines de répression/métabolisme , Protéines de répression/antagonistes et inhibiteurs , Protéines de répression/composition chimique , Composés phytochimiques/pharmacologie , Composés phytochimiques/composition chimique , Simulation numérique , Extraits de plantes/composition chimique , Extraits de plantes/pharmacologie , Simulation de dynamique moléculaire , Infections à papillomavirus/traitement médicamenteux , Infections à papillomavirus/virologie , Diterpènes/pharmacologie , Diterpènes/composition chimique , Liaison aux protéines
8.
J Virol ; 98(8): e0100324, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39046232

RÉSUMÉ

Three-stranded DNA-RNA structures known as R-loops that form during papillomavirus transcription can cause transcription-replication conflicts and lead to DNA damage. We found that R-loops accumulated at the viral early promoter in human papillomavirus (HPV) episomal cells but were greatly reduced in cells with integrated HPV genomes. RNA-DNA helicases unwind R-loops and allow for transcription and replication to proceed. Depletion of the RNA-DNA helicase senataxin (SETX) using siRNAs increased the presence of R-loops at the viral early promoter in HPV-31 (CIN612) and HPV-16 (W12) episomal HPV cell lines. Depletion of SETX reduced viral transcripts in episomal HPV cell lines. The viral E2 protein, which binds with high affinity to specific palindromes near the promoter and origin, complexes with SETX, and both SETX and E2 are present at the viral p97 promoter in CIN612 and W12 cells. SETX overexpression increased E2 transcription activity on the p97 promoter. SETX depletion also significantly increased integration of viral genomes in CIN612 cells. Our results demonstrate that SETX resolves viral R-loops to proceed with HPV transcription and prevent genome integration.IMPORTANCEPapillomaviruses contain small circular genomes of approximately 8 kilobase pairs and undergo unidirectional transcription from the sense strand of the viral genome. Co-transcriptional R-loops were recently reported to be present at high levels in cells that maintain episomal HPV and were also detected at the early viral promoter. R-loops can inhibit transcription and DNA replication. The process that removes R-loops from the PV genome and the requisite enzymes are unknown. We propose a model in which the host RNA-DNA helicase senataxin assembles on the HPV genome to resolve R-loops in order to maintain the episomal status of the viral genome.


Sujet(s)
Helicase , Enzymes multifonctionnelles , Régions promotrices (génétique) , Structures en boucle R , RNA helicases , Humains , RNA helicases/génétique , RNA helicases/métabolisme , Enzymes multifonctionnelles/génétique , Enzymes multifonctionnelles/métabolisme , Helicase/métabolisme , Helicase/génétique , Structures en boucle R/génétique , Plasmides/génétique , Réplication virale , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Papillomaviridae/génétique , Génome viral , Protéines des oncogènes viraux/génétique , Protéines des oncogènes viraux/métabolisme , Transcription génétique , Lignée cellulaire , ADN viral/génétique
9.
Front Immunol ; 15: 1405318, 2024.
Article de Anglais | MEDLINE | ID: mdl-39055715

RÉSUMÉ

Human papilloma virus (HPV) is an etiological factor of head and neck squamous cell carcinoma (HNSCC). To investigate the role of HPV antigen in anti-tumor immunity, we established mouse models by expressing HPV16 E6 and E7 in a SCC tumor cell line. We obtained two HPV antigen-expressing clones (C-225 and C-100) transplantable into C57BL/6 recipients. We found that C-225 elicited complete eradication in C57BL/6 mice (eradicated), whereas C-100 grew progressively (growing). We examined immune tumor microenvironment (TME) using flow cytometry and found that eradicated or growing tumors exhibited differential immune profiles that may influence the outcome of anti-tumor immunity. Surprisingly, the percentage of CD8 and CD4 tumor-infiltrating lymphocytes (TILs) was much higher in growing (C-100) than eradicated (C-225) tumor. However, the TILs upregulated PD-1 and LAG-3 more potently and exhibited impaired effector functions in growing tumor compared to their counterparts in eradicated tumor. C-225 TME is highly enriched with myeloid cells, especially polymorphonuclear (PMN) myeloid-derived suppressor cells (MDSC), whereas the percentage of M-MDSC and tumor-associated macrophages (TAMs) was much higher in C-100 TME, especially M2-TAMs (CD206+). The complete eradication of C-225 depended on CD8 T cells and elicited anti-tumor memory responses upon secondary tumor challenge. We employed DNA sequencing to identify differences in the T cell receptor of peripheral blood lymphocytes pre- and post-secondary tumor challenge. Lastly, C-225 and C-100 tumor lines harbored different somatic mutations. Overall, we uncovered differential immune TME that may underlie the divergent outcomes of anti-tumor immunity by establishing two SCC tumor lines, both of which express HPV16 E6 and E7 antigens. Our experimental models may provide a platform for pinpointing tumor-intrinsic versus host-intrinsic differences in orchestrating an immunosuppressive TME in HNSCCs and for identifying new targets that render tumor cells vulnerable to immune attack.


Sujet(s)
Modèles animaux de maladie humaine , Lymphocytes TIL , Souris de lignée C57BL , Protéines des oncogènes viraux , Protéines E7 de papillomavirus , Infections à papillomavirus , Microenvironnement tumoral , Animaux , Microenvironnement tumoral/immunologie , Souris , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme , Lignée cellulaire tumorale , Protéines des oncogènes viraux/immunologie , Protéines des oncogènes viraux/génétique , Protéines E7 de papillomavirus/immunologie , Protéines E7 de papillomavirus/génétique , Infections à papillomavirus/immunologie , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/virologie , Protéines de répression/génétique , Protéine LAG-3 , Humains , Évolution de la maladie , Lymphocytes T CD8+/immunologie , Récepteur-1 de mort cellulaire programmée , Femelle , Papillomavirus humain de type 16/immunologie , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/virologie
10.
Diagn Microbiol Infect Dis ; 110(1): 116440, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39018933

RÉSUMÉ

This study was designed to investigate the expression of HPV16 L1-protein in biopsies of oral mucosa samples. The expression of HPV16 L1 protein was investigated in biopsies taken from oral mucosa from patients who required pathological diagnosis of oral lesions. Seventy-two samples were incubated with anti-L1 protein monoclonal antibodies and protein detection was revealed with diaminobenzidine. Expression of L1 protein was performed by a pathologist blinded for tissue diagnosis under light microscopy. Most of the lesions of oral mucosa were present in lining mucosa (75 %) and the most frequent lesion were mucocele (n = 17, 23.6 %), epithelial hyperplasia (n = 6, 8.33 %), fibroma (n = 5, 6.9 %) and inflammatory hyperplasia (n = 5, 6.9 %). L1 protein expression was observed only in five (6.9 %) samples (two squamous cell carcinomas, two epithelial hyperplasia, and one gingival hyperplasia). We concluded that L1 expression in oral biopsies presented a low frequency in oral mucosal biopsies samples.


Sujet(s)
Protéines de capside , Muqueuse de la bouche , Protéines des oncogènes viraux , Infections à papillomavirus , Humains , Biopsie , Femelle , Muqueuse de la bouche/virologie , Muqueuse de la bouche/anatomopathologie , Infections à papillomavirus/diagnostic , Infections à papillomavirus/virologie , Adulte , Mâle , Protéines des oncogènes viraux/génétique , Adulte d'âge moyen , Équateur/épidémiologie , Protéines de capside/génétique , Protéines de capside/immunologie , Jeune adulte , Adolescent , Sujet âgé , Prévalence , Maladies de la bouche/virologie , Maladies de la bouche/anatomopathologie , Maladies de la bouche/épidémiologie , Papillomavirus humain de type 16/génétique , Immunohistochimie , Tumeurs de la bouche/virologie , Tumeurs de la bouche/anatomopathologie , Tumeurs de la bouche/diagnostic
11.
Anticancer Res ; 44(7): 2921-2931, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38925841

RÉSUMÉ

BACKGROUND/AIM: Human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC) is clinically and immunologically distinct from HPV-negative HNSCC. Herein, we investigated the presence of tumor antigens HPV E6/E7 and wild-type p53-specific T-cell responses, and the impact of immune checkpoint blockade in patients with HPV-positive HNSCC. MATERIALS AND METHODS: Peripheral blood mononuclear cells (PBMCs) from patients with HPV-positive HNSCC were stimulated with HPV E6/E7 or wild-type p53-derived peptide mixture and evaluated using the interferon-γ enzyme-linked immunosorbent spot assay. Flow cytometry was performed to analyze the proportion of T-cell subsets and T cells expressing immune checkpoint molecules. RESULTS: HPV E6/E7-specific T cells were detected in 22 (95.7%) of 23 patients, whereas wild-type p53-specific T cells were detected in 3 (15.0%) of 20 patients. Seven (43.8%) of 16 patients exhibited wild-type p53-specific T-cell responses, as determined using whole proteins instead of peptides. Immune checkpoint blockade enhanced wild-type p53-specific T-cell responses in 9 (45.0%) of 20 patients. Flow cytometric analysis of PBMCs revealed that responders exhibiting enhanced wild-type p53-specific T-cell responses following immune checkpoint blockade had a significantly higher proportion of Ki-67+CD4+ T cells, Ki-67+CD8+ T cells, regulatory T cells, PD-1+CD4+ T cells, and TIM-3+CD4+ T cells than non-responders. CONCLUSION: Our findings indicate that tumor antigen-specific T cells are present in the peripheral blood of patients with HPV-positive HNSCC. Blockade of checkpoint pathways can enhance T-cell responses in certain patients, probably via activated T cells, Tregs, and/or exhausted CD4+ T cells.


Sujet(s)
Tumeurs de la tête et du cou , Inhibiteurs de points de contrôle immunitaires , Infections à papillomavirus , Carcinome épidermoïde de la tête et du cou , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Carcinome épidermoïde de la tête et du cou/immunologie , Carcinome épidermoïde de la tête et du cou/virologie , Carcinome épidermoïde de la tête et du cou/traitement médicamenteux , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Tumeurs de la tête et du cou/immunologie , Tumeurs de la tête et du cou/virologie , Infections à papillomavirus/immunologie , Infections à papillomavirus/virologie , Antigènes néoplasiques/immunologie , Protéines des oncogènes viraux/immunologie , Protéine p53 suppresseur de tumeur/immunologie , Adulte , Agranulocytes/immunologie , Agranulocytes/métabolisme , Papillomaviridae/immunologie , Lymphocytes T/immunologie , Virus des Papillomavirus humains
12.
J Med Virol ; 96(6): e29769, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38932482

RÉSUMÉ

Integration of the human papillomavirus (HPV) genome into the cellular genome is a key event that leads to constitutive expression of viral oncoprotein E6/E7 and drives the progression of cervical cancer. However, HPV integration patterns differ on a case-by-case basis among related malignancies. Next-generation sequencing technologies still face challenges for interrogating HPV integration sites. In this study, utilizing Nanopore long-read sequencing, we identified 452 and 108 potential integration sites from the cervical cancer cell lines (CaSki and HeLa) and five tissue samples, respectively. Based on long Nanopore chimeric reads, we were able to analyze the methylation status of the HPV long control region (LCR), which controls oncogene E6/E7 expression, and to identify transcriptionally-active integrants among the numerous integrants. As a proof of concept, we identified an active HPV integrant in between RUNX2 and CLIC5 on chromosome 6 in the CaSki cell line, which was supported by ATAC-seq, H3K27Ac ChIP-seq, and RNA-seq analysis. Knockout of the active HPV integrant, by the CRISPR/Cas9 system, dramatically crippled cell proliferation and induced cell senescence. In conclusion, identifying transcriptionally-active HPV integrants with Nanopore sequencing can provide viable targets for gene therapy against HPV-associated cancers.


Sujet(s)
Thérapie génétique , Séquençage par nanopores , Infections à papillomavirus , Tumeurs du col de l'utérus , Intégration virale , Humains , Tumeurs du col de l'utérus/virologie , Femelle , Séquençage par nanopores/méthodes , Intégration virale/génétique , Thérapie génétique/méthodes , Infections à papillomavirus/virologie , Lignée cellulaire tumorale , Cellules HeLa , Protéines des oncogènes viraux/génétique , Séquençage nucléotidique à haut débit/méthodes , Papillomaviridae/génétique , Virus des Papillomavirus humains
13.
J Med Virol ; 96(6): e29753, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38895800

RÉSUMÉ

Human papillomavirus (HPV) type 81 has recently become one of the most common low-risk HPV types; however, literature focusing on it is limited. This study aimed to analyze the reasons for the increased detection rate of HPV81 and investigate its evolving pathogenicity. We analyzed the detection rates and trends of HPV81 in 229 061 exfoliated cervical cell samples collected from 2014 to 2023; collected samples of HPV81 single infections from two different time periods; and analyzed the allele frequencies, positive selection, viral load, persistent infection capacity, and pathogenicity of E6 and E7 genotypes. We found that the detection rate of HPV81 ranked first among the low-risk types in exfoliated cervical cells and exhibited a significantly increasing trend (p < 0.001). The frequency of the E6 prototype allele of HPV81 (n = 317) was significantly increased (p = 0.018) and demonstrated the strongest adaptive capacity. The viral load and persistent infection capacity of the E6 prototype were significantly higher than those of the mutants, thus serving as key drivers for increasing the detection rate of HPV81 and enhancing its pathogenicity. The viral load was positively correlated with persistent infection capacity and pathogenicity. Persistent infection was a crucial factor in the pathogenicity of HPV81. Successful adaptive evolution of HPV81 is accompanied by enhanced pathogenicity.


Sujet(s)
Génotype , Infections à papillomavirus , Infection persistante , Polymorphisme génétique , Charge virale , Humains , Infections à papillomavirus/virologie , Femelle , Infection persistante/virologie , Col de l'utérus/virologie , Col de l'utérus/anatomopathologie , Adulte , Papillomaviridae/génétique , Papillomaviridae/pathogénicité , Papillomaviridae/classification , Papillomaviridae/isolement et purification , Fréquence d'allèle , Protéines des oncogènes viraux/génétique , Virulence/génétique , Alphapapillomavirus/génétique , Alphapapillomavirus/pathogénicité , Alphapapillomavirus/classification , Alphapapillomavirus/isolement et purification , Virus des Papillomavirus humains
14.
Mol Biol Rep ; 51(1): 743, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874682

RÉSUMÉ

Human papillomavirus (HPV), a common cause of sexually transmitted diseases, may cause warts and lead to various types of cancers, which makes it important to understand the risk factors associated with it. HPV is the leading risk factor and plays a crucial role in the progression of cervical cancer. Viral oncoproteins E6 and E7 play a pivotal role in this process. Beyond cervical cancer, HPV-associated cancers of the mouth and throat are also increasing. HPV can also contribute to other malignancies like penile, vulvar, and vaginal cancers. Emerging evidence links HPV to these cancers. Research on the oncogenic effect of HPV is still ongoing and explorations of screening techniques, vaccination, immunotherapy and targeted therapeutics are all in progress. The present review offers valuable insight into the current understanding of the role of HPV in cancer and its potential implications for treatment and prevention in the future.


Sujet(s)
Infections à papillomavirus , Tumeurs du col de l'utérus , Humains , Infections à papillomavirus/virologie , Infections à papillomavirus/complications , Femelle , Tumeurs du col de l'utérus/virologie , Tumeurs du col de l'utérus/étiologie , Papillomaviridae/pathogénicité , Tumeurs/virologie , Tumeurs/thérapie , Protéines des oncogènes viraux/métabolisme , Protéines des oncogènes viraux/génétique , Facteurs de risque , Mâle
15.
JCI Insight ; 9(15)2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38916963

RÉSUMÉ

Despite epidermal turnover, the skin is host to a complex array of microbes, including viruses, such as HPV, which must infect and manipulate skin keratinocyte stem cells (KSCs) to survive. This crosstalk between the virome and KSC populations remains largely unknown. Here, we investigated the effect of HPV8 on KSCs using various mouse models. We observed that the HPV8 early region gene E6 specifically caused Lrig1+ hair follicle junctional zone KSC proliferation and expansion, which would facilitate viral transmission. Within Lrig1+ KSCs specifically, HPV8 E6 bound intracellular p300 to phosphorylate the STAT3 transcriptional regulatory node. This induced ΔNp63 expression, resulting in KSC expansion into the overlying epidermis. HPV8 was associated with 70% of human actinic keratoses. Together, these results define the "hit-and-run" mechanism for HPV8 in human actinic keratosis as an expansion of KSCs, which lack melanosome protection and are thus susceptible to sun light-induced malignant transformation.


Sujet(s)
Prolifération cellulaire , Kératinocytes , Kératose actinique , Protéines des oncogènes viraux , Infections à papillomavirus , Facteur de transcription STAT-3 , Cellules souches , Facteur de transcription STAT-3/métabolisme , Kératinocytes/virologie , Kératinocytes/métabolisme , Kératinocytes/anatomopathologie , Humains , Kératose actinique/anatomopathologie , Kératose actinique/métabolisme , Kératose actinique/virologie , Animaux , Souris , Cellules souches/métabolisme , Cellules souches/virologie , Protéines des oncogènes viraux/métabolisme , Protéines des oncogènes viraux/génétique , Infections à papillomavirus/virologie , Infections à papillomavirus/anatomopathologie , Infections à papillomavirus/métabolisme , Infections à papillomavirus/complications , Modèles animaux de maladie humaine , Femelle
16.
PLoS Pathog ; 20(6): e1012289, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38829892

RÉSUMÉ

During entry, human papillomavirus (HPV) traffics from the endosome to the trans Golgi network (TGN) and Golgi and then the nucleus to cause infection. Although dynein is thought to play a role in HPV infection, how this host motor recruits the virus to support infection and which entry step(s) requires dynein are unclear. Here we show that the dynein cargo adaptor BICD2 binds to the HPV L2 capsid protein during entry, recruiting HPV to dynein for transport of the virus along the endosome-TGN/Golgi axis to promote infection. In the absence of BICD2 function, HPV accumulates in the endosome and TGN and infection is inhibited. Cell-based and in vitro binding studies identified a short segment near the C-terminus of L2 that can directly interact with BICD2. Our results reveal the molecular basis by which the dynein motor captures HPV to promote infection and identify this virus as a novel cargo of the BICD2 dynein adaptor.


Sujet(s)
Protéines de capside , Papillomavirus humain de type 16 , Protéines des oncogènes viraux , Infections à papillomavirus , Humains , Protéines de capside/métabolisme , Papillomavirus humain de type 16/métabolisme , Protéines des oncogènes viraux/métabolisme , Infections à papillomavirus/métabolisme , Infections à papillomavirus/virologie , Dynéines/métabolisme , Endosomes/métabolisme , Endosomes/virologie , Réseau trans-golgien/métabolisme , Réseau trans-golgien/virologie , Pénétration virale , Liaison aux protéines , Cellules HeLa , Protéines associées aux microtubules/métabolisme , Dynéines cytoplasmiques/métabolisme
17.
Mol Cells ; 47(7): 100079, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38871298

RÉSUMÉ

The nonsense-mediated mRNA decay (NMD) pathway and the p53 pathway, linked to tumorgenesis, are also promising targets for cancer treatment. NMD plays an important role in RNA quality control, while the p53 pathway is involved in cancer suppression. However, their individual and combined effects on cervical cancer are poorly understood. In this study, we evaluated the impacts of NMD inhibitor, Mouse double minute 2 homolog (MDM2) inhibitor, and their combination on cell apoptosis, cell cycle, and p53 target genes in human papillomavirus-18-positive HeLa cells. Our findings revealed that XR-2 failed to activate p53 or induce apoptosis in HeLa cells, whereas SMG1 (serine/threonine-protein kinase 1) inhibitor repressed cell proliferation at high concentrations. Notably, the combination of these 2 agents significantly inhibited cell proliferation, arrested the cell cycle, and triggered cell apoptosis. Mechanistically, MDM2 inhibitor and NMD inhibitor likely exert a synergistically through the truncated E6 protein. These results underscore the potential of employing a combination of MDM2 inhibitor and NMD inhibitor as a promising candidate for the clinical treatment of human papillomavirus-infected tumors.


Sujet(s)
Apoptose , Prolifération cellulaire , Protéines proto-oncogènes c-mdm2 , Humains , Protéines proto-oncogènes c-mdm2/métabolisme , Protéines proto-oncogènes c-mdm2/antagonistes et inhibiteurs , Apoptose/effets des médicaments et des substances chimiques , Cellules HeLa , Prolifération cellulaire/effets des médicaments et des substances chimiques , Dégradation des ARNm non-sens/effets des médicaments et des substances chimiques , Protéine p53 suppresseur de tumeur/métabolisme , Synergie des médicaments , Protéines des oncogènes viraux/métabolisme , Tumeurs du col de l'utérus/traitement médicamenteux , Tumeurs du col de l'utérus/anatomopathologie , Tumeurs du col de l'utérus/métabolisme , Protéines de répression/métabolisme , Protéines de répression/antagonistes et inhibiteurs , Protéines de répression/génétique , Protéines de liaison à l'ADN
18.
J Virol ; 98(7): e0073524, 2024 Jul 23.
Article de Anglais | MEDLINE | ID: mdl-38874360

RÉSUMÉ

Oncogenic HPV E6 proteins have a PDZ-binding motif (PBM) which plays important roles in both the viral life cycle and tumor development. The PBM confers interaction with a large number of different PDZ domain-containing substrates, one of which is Sorting Nexin 27. This protein is part of the retromer complex and plays an important role in endocytic sorting pathways. It has been shown that at least two SNX27 interacting partners, GLUT1 and TANC2, are aberrantly trafficked due to the E6 PBM-dependent interaction with SNX27. To investigate further which other components of the endocytic trafficking pathway might be affected by the SNX27-HPV E6 interaction, we analyzed the SNX27 proteome interaction profile in a previously described HeLa cell line expressing GFP-SNX27, both in the presence and absence of the HPV-18 E6 oncoprotein. In this study, we identify a novel interacting partner of SNX27, secreted glycoprotein EMILIN2, whose release is blocked by HPV18 E6 in a PBM-dependent manner. Mechanistically, E6 can block EMILIN2 interaction with the WNT1 ligand, thereby enhancing WNT1 signaling and promoting cell proliferation. IMPORTANCE: This study demonstrates that HPV E6 blocks EMILIN2 inhibition of WNT1 signaling, thereby enhancing cell proliferation in HPV-positive tumor cells. This involves a novel mechanism whereby the E6 PBM actually contributes toward enhancing the interaction between SNX27 and EMILIN2, suggesting that the mode of recognition of SNX27 by E6 and EMILIN2 is different. This is the first example of the E6 PBM altering a PDZ domain-containing protein to enhance potential substrate recognition.


Sujet(s)
Papillomavirus humain de type 18 , Protéines des oncogènes viraux , Nexines de tri , Voie de signalisation Wnt , Humains , Protéines de liaison à l'ADN , Cellules HEK293 , Cellules HeLa , Papillomavirus humain de type 18/métabolisme , Papillomavirus humain de type 18/génétique , Protéines des oncogènes viraux/métabolisme , Protéines des oncogènes viraux/génétique , Infections à papillomavirus/virologie , Infections à papillomavirus/métabolisme , Domaines PDZ , Liaison aux protéines , Protéines de répression/métabolisme , Protéines de répression/génétique , Nexines de tri/métabolisme , Nexines de tri/génétique
19.
Hum Vaccin Immunother ; 20(1): 2352908, 2024 Dec 31.
Article de Anglais | MEDLINE | ID: mdl-38780076

RÉSUMÉ

Cervical cancer, among the deadliest cancers affecting women globally, primarily arises from persistent infection with high-risk human papillomavirus (HPV). To effectively combat persistent infection and prevent the progression of precancerous lesions into malignancy, a therapeutic HPV vaccine is under development. This study utilized an immunoinformatics approach to predict epitopes of cytotoxic T lymphocytes (CTLs) and helper T lymphocytes (HTLs) using the E6 and E7 oncoproteins of the HPV16 strain as target antigens. Subsequently, through meticulous selection of T-cell epitopes and other necessary elements, a multi-epitope vaccine was constructed, exhibiting good immunogenic, physicochemical, and structural characteristics. Furthermore, in silico simulations showed that the vaccine not only interacted well with toll-like receptors (TLR2/TLR3/TLR4), but also induced a strong innate and adaptive immune response characterized by elevated Th1-type cytokines, such as interferon-gamma (IFN-γ) and interleukin-2 (IL2). Additionally, our study investigated the effects of different immunization intervals on immune responses, aiming to optimize a time-efficient immunization program. In animal model experiments, the vaccine exhibited robust immunogenic, therapeutic, and prophylactic effects. Administered thrice, it consistently induced the expansion of specific CD4 and CD8 T cells, resulting in substantial cytokines release and increased proliferation of memory T cell subsets in splenic cells. Overall, our findings support the potential of this multi-epitope vaccine in combating HPV16 infection and signify its candidacy for future HPV vaccine development.


Through the stringent selection of T-cell epitopes and other necessary elements, a novel multi-epitope vaccine targeting HPV 16 E6 and E7 oncoproteins was constructed using an immunoinformatics approach.The vaccine designed can induce both cellular and humoral immune responses, encompassing all the required immunogenic, physicochemical, and structural characteristics for an ideal vaccine design. Moreover, it offers decent worldwide coverage.In animal studies, the vaccine demonstrated strong immune responses, including expansion of CD4 and CD8 T cells, cytokine release, and enhanced memory T cell proliferation, resulting in long-term anti-tumor effects, inhibition of tumor growth, and prolonged survival in tumor-bearing mice.The immunological evaluation of the designed vaccine suggests its potential as a novel vaccine candidate against HPV 16.


Sujet(s)
Déterminants antigéniques des lymphocytes T , Papillomavirus humain de type 16 , Protéines des oncogènes viraux , Infections à papillomavirus , Vaccins contre les papillomavirus , Tumeurs du col de l'utérus , Vaccins à ADN , Femelle , Vaccins contre les papillomavirus/immunologie , Vaccins contre les papillomavirus/administration et posologie , Papillomavirus humain de type 16/immunologie , Vaccins à ADN/immunologie , Vaccins à ADN/administration et posologie , Infections à papillomavirus/prévention et contrôle , Infections à papillomavirus/immunologie , Déterminants antigéniques des lymphocytes T/immunologie , Animaux , Protéines des oncogènes viraux/immunologie , Protéines des oncogènes viraux/génétique , Tumeurs du col de l'utérus/prévention et contrôle , Tumeurs du col de l'utérus/immunologie , Tumeurs du col de l'utérus/virologie , Protéines E7 de papillomavirus/immunologie , Souris , Humains , Lymphocytes T cytotoxiques/immunologie , Protéines de répression/immunologie , Lymphocytes T auxiliaires/immunologie , Souris de lignée C57BL , Interféron gamma/métabolisme , Interféron gamma/immunologie
20.
J Med Virol ; 96(6): e29685, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38783790

RÉSUMÉ

Human papillomaviruses (HPVs) are double-stranded DNA (dsDNA) tumor viruses causally associated with 5% of human cancers, comprising both anogenital and upper aerodigestive tract carcinomas. Despite the availability of prophylactic vaccines, HPVs continue to pose a significant global health challenge, primarily due to inadequate vaccine access and coverage. These viruses can establish persistent infections by evading both the intrinsic defenses of infected tissues and the extrinsic defenses provided by professional innate immune cells. Crucial for their evasion strategies is their unique intraepithelial life cycle, which effectively shields them from host detection. Thus, strategies aimed at reactivating the innate immune response within infected or transformed epithelial cells, particularly through the production of type I interferons (IFNs) and lymphocyte-recruiting chemokines, are considered viable solutions to counteract the adverse effects of persistent infections by these oncogenic viruses. This review focuses on the complex interplay between the high-risk HPV oncoproteins E6 and E7 and the innate immune response in epithelial cells and HPV-associated cancers. In particular, it details the molecular mechanisms by which E6 and E7 modulate the innate immune response, highlighting significant progress in our comprehension of these processes. It also examines forward-looking strategies that exploit the innate immune system to ameliorate existing anticancer therapies, thereby providing crucial insights into future therapeutic developments.


Sujet(s)
Échappement immunitaire , Immunité innée , Protéines des oncogènes viraux , Infections à papillomavirus , Humains , Infections à papillomavirus/immunologie , Infections à papillomavirus/virologie , Protéines des oncogènes viraux/immunologie , Protéines E7 de papillomavirus/immunologie , Papillomaviridae/immunologie , Papillomaviridae/pathogénicité , Interactions hôte-pathogène/immunologie , Cellules épithéliales/virologie , Cellules épithéliales/immunologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE