1
|
Ferré VM, Coppée R, Gbeasor-Komlanvi FA, Vacher S, Bridier-Nahmias A, Bucau M, Salou M, Lameiras S, Couvelard A, Dagnra AC, Bieche I, Descamps D, Ekouevi DK, Ghosn J, Charpentier C. Viral whole genome sequencing reveals high variations in APOBEC3 editing between HPV risk categories. J Med Virol 2024; 96:e70002. [PMID: 39400339 DOI: 10.1002/jmv.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/15/2024]
Abstract
High-risk human papillomavirus (HPV) infections are responsible for cervical cancer. However, little is known about the differences between HPV types and risk categories regarding their genetic diversity and particularly APOBEC3-induced mutations - which contribute to the innate immune response to HPV. Using a capture-based next-generation sequencing, 156 HPV whole genome sequences covering 43 HPV types were generated from paired cervical and anal swabs of 30 Togolese female sex workers (FSWs) sampled in 2017. Genetic diversity and APOBEC3-induced mutations were assessed at the viral whole genome and gene levels. Thirty-four pairwise sequence comparisons covering 24 HPV types in cervical and anal swabs revealed identical infections in the two anatomical sites. Differences in genetic diversity among HPV types was observed between patients. The E6 gene was significantly less conserved in low-risk HPVs (lrHPVs) compared to high-risk HPVs (hrHPVs) (p = 0.009). APOBEC3-induced mutations were found to be more common in lrHPVs than in hrHPVs (p = 0.005), supported by our data and by using large HPV sequence collections from the GenBank database. Focusing on the most common lrHPVs 6 and 11 and hrHPVs 16 and 18, APOBEC3-induced mutations were predominantly found in the E4 and E6 genes in lrHPVs, but were almost absent in these genes in hrHPVs. The variable APOBEC3 mutational signatures could contribute to the different oncogenic potentials between HPVs. Further studies are needed to conclusively determine whether APOBEC3 editing levels are associated to the carcinogenic potential of HPVs at the type and sublineage scales.
Collapse
Affiliation(s)
- Valentine Marie Ferré
- Université Paris Cité, Inserm IAME UMR 1137, Paris, F-75018, France
- Service de Virologie, AP-HP, Hôpital Bichat - Claude Bernard, Paris, F-75018, France
| | - Romain Coppée
- Université Paris Cité, Inserm IAME UMR 1137, Paris, F-75018, France
| | - Fifonsi A Gbeasor-Komlanvi
- Département de Santé Publique, Université de Lomé, Faculté des Sciences de la Santé, Lomé, Togo
- Centre Africain de Recherche en Epidémiologie et en Santé Publique (CARESP), Lomé, Togo
| | - Sophie Vacher
- Department of Genetics, Institut Curie, PSL Research University, Paris, France
| | | | - Margot Bucau
- Département de Pathologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, F-75018, France
| | - Mounerou Salou
- Université de Lomé, Centre de Biologie Moléculaire et d'Immunologie, Lomé, Togo
| | - Sonia Lameiras
- Institut Curie, Genomics of Excellence (ICGex) Platform, PSL Research University, Paris, France
| | - Anne Couvelard
- Département de Pathologie, AP-HP, Hôpital Bichat-Claude Bernard, Paris, F-75018, France
- Université de Paris, Centre of Research on Inflammation, Paris, INSERM U1149, France
| | - Anoumou Claver Dagnra
- Université de Lomé, Centre de Biologie Moléculaire et d'Immunologie, Lomé, Togo
- Programme national de lutte contre le sida et les infections sexuellement transmissibles, Lomé, Togo
| | - Ivan Bieche
- Department of Genetics, Institut Curie, PSL Research University, Paris, France
- INSERM U1016, Faculty of Pharmaceutical and Biological Sciences, Paris Cité University, Paris, France
| | - Diane Descamps
- Université Paris Cité, Inserm IAME UMR 1137, Paris, F-75018, France
- Service de Virologie, AP-HP, Hôpital Bichat - Claude Bernard, Paris, F-75018, France
| | - Didier K Ekouevi
- Département de Santé Publique, Université de Lomé, Faculté des Sciences de la Santé, Lomé, Togo
- ISPED, Université de Bordeaux & Centre INSERM U1219 - Bordeaux Population Health, Bordeaux, France
| | - Jade Ghosn
- Université Paris Cité, Inserm IAME UMR 1137, Paris, F-75018, France
- Service de Maladies Infectieuses et Tropicales, AP-HP, Hôpital Bichat-Claude Bernard, Paris, F-75018, France
| | - Charlotte Charpentier
- Université Paris Cité, Inserm IAME UMR 1137, Paris, F-75018, France
- Service de Virologie, AP-HP, Hôpital Bichat - Claude Bernard, Paris, F-75018, France
| |
Collapse
|
2
|
Clarke MA. HPV Testing and its Role in Cervical Cancer Screening. Clin Obstet Gynecol 2023; 66:448-469. [PMID: 37650662 DOI: 10.1097/grf.0000000000000793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
The recognition that persistent infection with carcinogenic human papillomavirus (HPV) is a necessary cause of cervical precancer and cancer has led to the introduction of HPV testing into cervical cancer screening, either as a primary screening test or in conjunction with cervical cytology (i.e., co-testing). HPV testing has much higher sensitivity for detection of cervical precancer and provides greater long-term reassurance if negative compared to cytology. However, most HPV infections are transient, and do not progress to invasive cancer, thus triage tests are required to identify individuals who should be referred to colposcopy for diagnostic evaluation. This chapter begins with a description of the biology, natural history, and epidemiology of HPV as a foundation for understanding the role of HPV in cervical carcinogenesis. This section is followed by a detailed discussion regarding the introduction of HPV-based testing and triage into cervical cancer screening and management. Summarized triage tests include cervical cytology, HPV genotyping, p16/Ki-67 dual stain, and HPV and cellular methylation markers. The final section of this chapter includes an important discussion on cervical cancer disparities, particularly within the United States, followed by concluding remarks.
Collapse
Affiliation(s)
- Megan A Clarke
- Division of Cancer Epidemiology & Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
3
|
Glover A, Zhang Z, Shannon-Lowe C. Deciphering the roles of myeloid derived suppressor cells in viral oncogenesis. Front Immunol 2023; 14:1161848. [PMID: 37033972 PMCID: PMC10076641 DOI: 10.3389/fimmu.2023.1161848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/10/2023] [Indexed: 04/11/2023] Open
Abstract
Myeloid derived suppressor cells (MDSCs) are a heterogenous population of myeloid cells derived from monocyte and granulocyte precursors. They are pathologically expanded in conditions of ongoing inflammation where they function to suppress both innate and adaptive immunity. They are subdivided into three distinct subsets: monocytic (M-) MDSC, polymorphonuclear (or neutrophilic) (PMN-) MDSC and early-stage (e-) MDSC that may exhibit differential function in different pathological scenarios. However, in cancer they are associated with inhibition of the anti-tumour immune response and are universally associated with a poor prognosis. Seven human viruses classified as Group I carcinogenic agents are jointly responsible for nearly one fifth of all human cancers. These viruses represent a large diversity of species, including DNA, RNA and retroviridae. They include the human gammaherpesviruses (Epstein Barr virus (EBV) and Kaposi's Sarcoma-Associated Herpesvirus (KSHV), members of the high-risk human papillomaviruses (HPVs), hepatitis B and C (HBV, HCV), Human T cell leukaemia virus (HTLV-1) and Merkel cell polyomavirus (MCPyV). Each of these viruses encode an array of different oncogenes that perturb numerous cellular pathways that ultimately, over time, lead to cancer. A prerequisite for oncogenesis is therefore establishment of chronic infection whereby the virus persists in the host cells without being eradicated by the antiviral immune response. Although some of the viruses can directly modulate the immune response to enable persistence, a growing body of evidence suggests the immune microenvironment is modulated by expansions of MDSCs, driven by viral persistence and oncogenesis. It is likely these MDSCs play a role in loss of immune recognition and function and it is therefore essential to understand their phenotype and function, particularly given the increasing importance of immunotherapy in the modern arsenal of anti-cancer therapies. This review will discuss the role of MDSCs in viral oncogenesis. In particular we will focus upon the mechanisms thought to drive the MDSC expansions, the subsets expanded and their impact upon the immune microenvironment. Importantly we will explore how MDSCs may modulate current immunotherapies and their impact upon the success of future immune-based therapies.
Collapse
|
4
|
Haręża DA, Wilczyński JR, Paradowska E. Human Papillomaviruses as Infectious Agents in Gynecological Cancers. Oncogenic Properties of Viral Proteins. Int J Mol Sci 2022; 23:1818. [PMID: 35163748 PMCID: PMC8836588 DOI: 10.3390/ijms23031818] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/29/2022] [Accepted: 02/03/2022] [Indexed: 01/25/2023] Open
Abstract
Human papillomaviruses (HPVs), which belong to the Papillomaviridae family, constitute a group of small nonenveloped double-stranded DNA viruses. HPV has a small genome that only encodes a few proteins, and it is also responsible for 5% of all human cancers, including cervical, vaginal, vulvar, penile, anal, and oropharyngeal cancers. HPV types may be classified as high- and low-risk genotypes (HR-HPVs and LR-HPVs, respectively) according to their oncogenic potential. HR-HPV 16 and 18 are the most common types worldwide and are the primary types that are responsible for most HPV-related cancers. The activity of the viral E6 and E7 oncoproteins, which interfere with critical cell cycle points such as suppressive tumor protein p53 (p53) and retinoblastoma protein (pRB), is the major contributor to HPV-induced neoplastic initiation and progression of carcinogenesis. In addition, the E5 protein might also play a significant role in tumorigenesis. The role of HPV in the pathogenesis of gynecological cancers is still not fully understood, which indicates a wide spectrum of potential research areas. This review focuses on HPV biology, the distribution of HPVs in gynecological cancers, the properties of viral oncoproteins, and the molecular mechanisms of carcinogenesis.
Collapse
Affiliation(s)
- Daria A. Haręża
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 93-232 Lodz, Poland;
- BioMedChem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences, 90-237 Lodz, Poland
| | - Jacek R. Wilczyński
- Department of Surgical and Oncological Gynecology, Medical University of Lodz, 90-419 Lodz, Poland;
| | - Edyta Paradowska
- Laboratory of Virology, Institute of Medical Biology of the Polish Academy of Sciences, 93-232 Lodz, Poland;
| |
Collapse
|
5
|
Stanley M. Host defence and persistent human papillomavirus infection. Curr Opin Virol 2021; 51:106-110. [PMID: 34628358 DOI: 10.1016/j.coviro.2021.09.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/17/2021] [Accepted: 09/14/2021] [Indexed: 10/20/2022]
Abstract
The ability to establish long term persistent infection is a feature of human papillomaviruses. The available evidence is that this ability is a consequence of a complex local immune milieu whereby innate immune receptors and signalling pathway cascades are inhibited by HPV early proteins resulting in failure of dendritic cell maturation, antigen processing and presentation and activation of cytotoxic antigen specific T cell responses. The development of cutaneous and mucosal infection models with the mouse papillomavirus MmuPV1 and the access to multiple gene deficient strains is providing the frame work to dissect the mechanisms underlying these complex host virus interactions.
Collapse
|
6
|
Cytokine response following perturbation of the cervicovaginal milieu during HPV genital infection. Immunol Res 2021; 69:255-263. [PMID: 33939124 DOI: 10.1007/s12026-021-09196-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
Human papillomaviruses (HPVs) are oncogenic viruses causing most cervical cancers. Highly prevalent in young, sexually active women, only a minority of HPV infections persist. To better characterize the immuno-modulatory impact of early HPV infections, we measured changes in a panel of 20 cytokines in cervicovaginal samples collected from young women who were tested for HPV and self-reported for genital inflammation and infection symptoms. Multi-factor statistical analyses revealed that increased IL-1Alpha and IL-12/IL-23p40 concentrations were associated with HPV infection and that macrophage inflammatory proteins were associated in particular with high-risk HPV infections. ClinicalTrials.gov identifier NCT02946346.
Collapse
|
7
|
Della Fera AN, Warburton A, Coursey TL, Khurana S, McBride AA. Persistent Human Papillomavirus Infection. Viruses 2021; 13:v13020321. [PMID: 33672465 PMCID: PMC7923415 DOI: 10.3390/v13020321] [Citation(s) in RCA: 91] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/16/2021] [Accepted: 02/17/2021] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The success of HPV as an infectious agent lies not within its ability to cause disease, but rather in the adeptness of the virus to establish long-term persistent infection. The ability of HPV to replicate and maintain its genome in a stratified epithelium is contingent on the manipulation of many host pathways. HPVs must abrogate host anti-viral defense programs, perturb the balance of cellular proliferation and differentiation, and hijack DNA damage signaling and repair pathways to replicate viral DNA in a stratified epithelium. Together, these characteristics contribute to the ability of HPV to achieve long-term and persistent infection and to its evolutionary success as an infectious agent. Abstract Persistent infection with oncogenic human papillomavirus (HPV) types is responsible for ~5% of human cancers. The HPV infectious cycle can sustain long-term infection in stratified epithelia because viral DNA is maintained as low copy number extrachromosomal plasmids in the dividing basal cells of a lesion, while progeny viral genomes are amplified to large numbers in differentiated superficial cells. The viral E1 and E2 proteins initiate viral DNA replication and maintain and partition viral genomes, in concert with the cellular replication machinery. Additionally, the E5, E6, and E7 proteins are required to evade host immune responses and to produce a cellular environment that supports viral DNA replication. An unfortunate consequence of the manipulation of cellular proliferation and differentiation is that cells become at high risk for carcinogenesis.
Collapse
|
8
|
Britto AMA, Goes LR, Sivro A, Policarpo C, Meirelles ÂR, Furtado Y, Almeida G, Arthos J, Cicala C, Soares MA, Machado ES, Giannini ALM. HPV Induces Changes in Innate Immune and Adhesion Molecule Markers in Cervical Mucosa With Potential Impact on HIV Infection. Front Immunol 2020; 11:2078. [PMID: 33013878 PMCID: PMC7494736 DOI: 10.3389/fimmu.2020.02078] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 07/30/2020] [Indexed: 12/03/2022] Open
Abstract
While most HPV infections are asymptomatic and clear spontaneously, persistent infection with high-risk HPVs is associated with cervical cancer and with increased risk of HIV acquisition. Although several hypotheses have been proposed to explain this phenomenon, none has been confirmed. Our aim was to investigate the expression of host factors involved in the susceptibility to HIV infection among HPV-infected women. Cervical samples were collected to characterize the expression levels of HIV susceptibility markers in the mucosa of HPV-infected compared with HPV-uninfected women. No differences in the frequency of CCR5+, integrin α4β7+, activated and memory CD4+ T-cell were detected between the groups. We additionally evaluated the expression levels of genes involved in innate immune responses and in cell adhesion. HPV infected patients expressed higher levels of TLR9 and lower levels of pattern recognition receptors that recognize RNA (TLR3, TLR7, and MDA5/IFIH1). We also detected an impaired IFN pathway, with an increased Type I IFN and a decreased IFNα2 receptor expression. HPV+ samples displayed reduced expression of genes for adherens and tight junctions. Taken together, these results suggest that although HPV infection does not result in the recruitment/activation of susceptible CD4+ T-cell in the female genital tract, it leads to changes in the innate antiviral immune responses and in cell adhesion that are likely to favor HIV infection.
Collapse
Affiliation(s)
- Alan Messala A Britto
- Programa de Oncovirologia, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil.,Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Livia R Goes
- Programa de Oncovirologia, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil.,Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Aida Sivro
- Centre for the AIDS Programme of Research in South Africa (CAPRISA), Durban, South Africa.,Department of Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada.,Department of Medical Microbiology, University of KwaZulu-Natal, Durban, South Africa
| | - Cintia Policarpo
- Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ângela R Meirelles
- Instituto de Ginecologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yara Furtado
- Instituto de Ginecologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gutemberg Almeida
- Instituto de Ginecologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Marcelo A Soares
- Programa de Oncovirologia, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | - Elizabeth S Machado
- Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ana Lúcia M Giannini
- Laboratório de Genômica Funcional e Transdução de Sinal, Departamento de Genética, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
9
|
Hua C, Zhu J, Zhang B, Sun S, Song Y, van der Veen S, Cheng H. Digital RNA Sequencing of Human Epidermal Keratinocytes Carrying Human Papillomavirus Type 16 E7. Front Genet 2020; 11:819. [PMID: 32849815 PMCID: PMC7419603 DOI: 10.3389/fgene.2020.00819] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/08/2020] [Indexed: 12/13/2022] Open
Abstract
High-risk human papillomavirus (HPV) infections are the predominant cause of cervical cancer and its early gene E7 plays an important role in cellular proliferation and cell-cycle progression. While tremendous progress has been made in exploring the molecular mechanisms in late tumorigenesis, many pathways showing how HPV deregulates host gene expression in early inapparent infections and early tumorigenesis still remain undefined. Digital RNA sequencing was performed and a total of 195 differentially expressed genes were identified between the HPV16 E7-transfected NHEKs and control cells (p < 0.05, fold-change > 2). GO enrichment showed that HPV16 E7 primarily affected processes involved in anti-viral and immune responses, while KEGG pathway analysis showed enrichment of gene clusters of associated with HPV infection and MAPK signaling. Of the differentially expressed genes, IFI6, SLC39A9 and ZNF185 showed a strong correlation with tumor progression and patient survival in the OncoLnc database while roles for AKAP12 and DUSP5 in carcinogenesis and poor prognosis have previously been established for other cancer types. Our study identified several novel HPV16 E7-regulated candidate genes with putative functions in tumorigenesis, thus providing new insights into HPV persistence in keratinocytes and early onset of tumorigenesis.
Collapse
Affiliation(s)
- Chunting Hua
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiang Zhu
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Boya Zhang
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Siyuan Sun
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yinjing Song
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Stijn van der Veen
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Microbiology and Parasitology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hao Cheng
- Department of Dermatology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
Bolt R, Foran B, Murdoch C, Lambert DW, Thomas S, Hunter KD. HPV-negative, but not HPV-positive, oropharyngeal carcinomas induce fibroblasts to support tumour invasion through micro-environmental release of HGF and IL-6. Carcinogenesis 2018; 39:170-179. [PMID: 29140428 DOI: 10.1093/carcin/bgx130] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 11/07/2017] [Indexed: 01/02/2023] Open
Abstract
Human papillomavirus (HPV) infection is causally related to a subset of oropharyngeal carcinomas (OPC) and is linked to a more favourable prognosis compared to HPV-negative OPC. The mechanisms underlying this effect on prognosis are not fully understood, but interactions with the tumour microenvironment may be pivotal. Here, we investigated the role of the tumour microenvironment in HPV-positive compared to HPV-negative cancer using 2D and 3D modelling of OPC interactions with stromal fibroblasts. HPV-negative, but not HPV-positive, OPC-derived cell lines induced a rapid fibroblast secretory response that supported 2D cancer cell migration and invasion in vitro. Array profiling of this HPV-negative induced fibroblast secretome identified hepatocyte growth factor (HGF) as the principal secreted factor that promoted cancer cell migration. The interaction between HPV-negative cell lines and fibroblasts in 2D was prevented using c-Met (HGF receptor) inhibitors, which further restricted both HPV-negative and positive cell invasion in 3D co-culture models. Furthermore, we discovered a synergistic relationship between HGF and IL-6 in the support of migration that relates JAK activation to HGF responsiveness in HPV-negative lines. In summary, our data show significant differences in the interactions between HPV-positive and HPV-negative OPC cells and stromal fibroblasts. In addition, we, provide in vitro evidence to support the clinical application of c-MET inhibitors in the control of early HPV-negative OPC.
Collapse
Affiliation(s)
- Robert Bolt
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Bernadette Foran
- Department of Oncology, Weston Park Hospital, Sheffield, South Yorkshire, UK
| | - Craig Murdoch
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Daniel W Lambert
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Sally Thomas
- Department of Biomedical Sciences, University of Sheffield, Sheffield, South Yorkshire, UK
| | - Keith D Hunter
- School of Clinical Dentistry, University of Sheffield, Sheffield, South Yorkshire, UK
| |
Collapse
|
11
|
Abstract
Human papillomaviruses (HPVs) are an ancient group of viruses with small, double-stranded DNA circular genomes. They are species-specific and have a strict tropism for mucosal and cutaneous stratified squamous epithelial surfaces of the host. A subset of these viruses has been demonstrated to be the causative agent of several human cancers. Here, we review the biology, natural history, evolution and cancer association of the oncogenic HPVs.This article is part of the themed issue 'Human oncogenic viruses'.
Collapse
Affiliation(s)
- Alison A McBride
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
12
|
Chen L, Luan S, Xia B, Liu Y, Gao Y, Yu H, Mu Q, Zhang P, Zhang W, Zhang S, Wei G, Yang M, Li K. Integrated analysis of HPV-mediated immune alterations in cervical cancer. Gynecol Oncol 2018; 149:248-255. [PMID: 29572030 DOI: 10.1016/j.ygyno.2018.01.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 01/25/2018] [Accepted: 01/28/2018] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Human papillomavirus (HPV) infection is the primary cause of cervical cancer. HPV-mediated immune alterations are known to play crucial roles in determining viral persistence and host cell transformation. We sought to thoroughly understand HPV-directed immune alterations in cervical cancer by exploring publically available datasets. METHODS 130 HPV positive and 7 HPV negative cervical cancer cases from The Cancer Genome Atlas were compared for differences in gene expression levels and functional enrichment. Analyses for copy number variation (CNV) and genetic mutation were conducted for differentially expressed immune genes. Kaplan-Meier analysis was performed to assess survival and relapse differences across cases with or without alterations of the identified immune signature genes. RESULTS Genes up-regulated in HPV positive cervical cancer were enriched for various gene ontology terms of immune processes (P=1.05E-14~1.00E-05). Integrated analysis of the differentially expressed immune genes identified 9 genes that displayed either CNV, genetic mutation and/or gene expression changes in at least 10% of the cases of HPV positive cervical cancer. Genomic amplification may cause elevated levels of these genes in some HPV positive cases. Finally, patients with alterations in at least one of the nine signature genes overall had earlier relapse compared to those without any alterations. The altered expression of either TFRC or MMP13 may indicate poor survival for a subset of cervical cancer patients (P=1.07E-07). CONCLUSIONS We identified a novel immune gene signature for HPV positive cervical cancer that is potentially associated with early relapse of cervical cancer.
Collapse
Affiliation(s)
- Long Chen
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China.
| | - Shaohong Luan
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Baoguo Xia
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Yansheng Liu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Yuan Gao
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Hongyan Yu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Qingling Mu
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Ping Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Weina Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Shengmiao Zhang
- Department of Gynecology, Qingdao Municipal Hospital, Qingdao 266011, PR China
| | - Guopeng Wei
- Gezhi Research Lab, Building T1, No.722 Yizhou Avenue, Chengdu 610000, PR China
| | - Min Yang
- Gezhi Research Lab, Building T1, No.722 Yizhou Avenue, Chengdu 610000, PR China
| | - Ke Li
- Gezhi Research Lab, Building T1, No.722 Yizhou Avenue, Chengdu 610000, PR China
| |
Collapse
|
13
|
McCormack SE, Cruz CRY, Wright KE, Powell AB, Lang H, Trimble C, Keller MD, Fuchs E, Bollard CM. Human papilloma virus-specific T cells can be generated from naïve T cells for use as an immunotherapeutic strategy for immunocompromised patients. Cytotherapy 2018; 20:385-393. [PMID: 29331266 DOI: 10.1016/j.jcyt.2017.11.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 11/21/2017] [Accepted: 11/24/2017] [Indexed: 01/31/2023]
Abstract
Human papilloma virus (HPV) is a known cause of cervical cancer, squamous cell carcinoma and laryngeal cancer. Although treatments exist for HPV-associated malignancies, patients unresponsive to these therapies have a poor prognosis. Recent findings from vaccine studies suggest that T-cell immunity is essential for disease control. Because Epstein-Barr Virus (EBV)-specific T cells have been highly successful in treating or preventing EBV-associated tumors, we hypothesized that the development of a manufacturing platform for HPV-specific T cells from healthy donors could be used in a third-party setting to treat patients with high-risk/relapsed HPV-associated cancers. Most protocols for generating virus-specific T cells require prior exposure of the donor to the targeted virus and, because the seroprevalence of high-risk HPV types varies greatly by age and ethnicity, manufacturing of donor-derived HPV-specific T cells has proven challenging. We, therefore, made systematic changes to our current Good Manufacturing Practice (GMP)-compliant protocols to improve antigen presentation, priming and expansion for the manufacture of high-efficacy HPV-specific T cells. Like others, we found that current methodologies fail to expand HPV-specific T cells from most healthy donors. By optimizing dendritic cell maturation and function with lipopolysaccharide (LPS) and interferon (IFN)γ, adding interleukin (IL)-21 during priming and depleting memory T cells, we achieved reliable expansion of T cells specific for oncoproteins E6 and E7 to clinically relevant amounts (mean, 578-fold expansion; n = 10), which were polyfunctional based on cytokine multiplex analysis. In the third-party setting, such HPV-specific T-cell products might serve as a potent salvage therapy for patients with HPV-associated diseases.
Collapse
Affiliation(s)
- Sarah E McCormack
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA
| | - Conrad Russell Y Cruz
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA; Sheikh Zayed Institute, Children's National Medical Center, Washington, DC, USA
| | - Kaylor E Wright
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA; Sheikh Zayed Institute, Children's National Medical Center, Washington, DC, USA
| | - Allison B Powell
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA; Sheikh Zayed Institute, Children's National Medical Center, Washington, DC, USA
| | - Haili Lang
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA
| | - Cornelia Trimble
- Department of Gynecology and Obstetrics, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Michael D Keller
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA; Division of Allergy and Immunology, Children's National Medical Center, Washington, DC, USA
| | - Ephraim Fuchs
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Catherine M Bollard
- Program for Cell Enhancement and Technologies for Immunotherapy, Children's National Medical Center, Washington, DC, USA; Sheikh Zayed Institute, Children's National Medical Center, Washington, DC, USA; Division of Allergy and Immunology, Children's National Medical Center, Washington, DC, USA; Division of Blood and Marrow Transplantation, Children's National Medical Center, Washington, DC, USA.
| |
Collapse
|
14
|
Behdari A, Saburi E. Successful treatment of common warts with alum. Anc Sci Life 2018. [DOI: 10.4103/asl.asl_79_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15
|
Levan J, Vliet-Gregg PA, Robinson KL, Katzenellenbogen RA. Human papillomavirus type 16 E6 and NFX1-123 mislocalize immune signaling proteins and downregulate immune gene expression in keratinocytes. PLoS One 2017; 12:e0187514. [PMID: 29117186 PMCID: PMC5695606 DOI: 10.1371/journal.pone.0187514] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/22/2017] [Indexed: 12/16/2022] Open
Abstract
Human papillomavirus (HPV) is the most prevalent sexually transmitted infection, affecting an estimated 11% of the world's population. The high-risk HPV types (HR HPV) account for approximately 5% of the global burden of cancer and thus cause high morbidity and mortality. Although it is known that persistent infection with HR HPV is the greatest risk factor for developing HPV-associated cancer, and that the HPV early proteins E6 and E7 dysregulate immune detection by its host cells, the mechanisms of immune evasion by HR HPV are not well understood. Previous work in the laboratory identified the endogenous cytoplasmic host protein NFX1-123 as a binding partner of the HR HPV type 16 oncoprotein E6 (16E6). Together NFX1-123 and 16E6 affect cellular growth, differentiation, and immortalization genes and pathways. In a whole genome microarray, human foreskin keratinocytes (HFKs) stably expressing 16E6 and overexpressing NFX1-123 showed a diverse set of innate immune genes downregulated two-fold or more when compared to 16E6 cells with endogenous NFX1-123. We demonstrated that 16E6 and NFX1-123 decreased expression of pro-inflammatory cytokines and interferon-stimulated genes (ISGs) in 16E6 HFKs at the mRNA and protein level. Knock down of NFX1-123 in 16E6 HFKs resulted in a derepression of innate immune genes, pointing to the requirement of NFX1-123 for immune regulation in the context of 16E6. Studies using immunofluorescent microscopy revealed that 16E6 and NFX1-123 disturbed the normal localization of signaling proteins involved in initiating the immune response. This study identifies NFX1-123 as a critical host protein partner through which 16E6 is able to subvert the immune response and in turn permit a long-lived HR HPV infection.
Collapse
Affiliation(s)
- Justine Levan
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Pathobiology Interdisciplinary Program, Department of Global Health, University of Washington, Seattle, Washington, United States of America
| | - Portia A. Vliet-Gregg
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Kristin L. Robinson
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
| | - Rachel A. Katzenellenbogen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, Washington, United States of America
- Pathobiology Interdisciplinary Program, Department of Global Health, University of Washington, Seattle, Washington, United States of America
- Department of Pediatrics, Division of Adolescent Medicine, University of Washington, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
16
|
HPV11 E6 mutation by overexpression of APOBEC3A and effects of interferon-ω on APOBEC3s and HPV11 E6 expression in HPV11.HaCaT cells. Virol J 2017; 14:211. [PMID: 29100527 PMCID: PMC5670706 DOI: 10.1186/s12985-017-0878-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 10/27/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Condyloma acuminatum, infected by low-risk human papillomaviruses (e.g., HPV6 and HPV11), is one of the most widespread sexually transmitted diseases. Apolipoprotein B mRNA-editing enzyme catalytic polypeptide-like 3 proteins (APOBEC3s, A3s) are cellular cytidine deaminases acting as antiviral factors through hypermutation of viral genome. However, it remains unknown whether A3s results in HPV11 gene mutations and interferon-ω (IFN-ω) exhibits antiviral activities through the A3s system. Here we investigated whether enhanced APOBEC3A (A3A) resulted in the E6 gene mutations and explore the effects of recombinant human interferon-ω (rhIFN-ω) on A3s/E6 expression in HaCaT keratinocytes containing the genome of HPV 11 (HPV11.HaCaT cells). METHODS A3A-overexpressed HPV11.HaCaT (A3A-HPV11.HaCaT) cells were established by lentiviral infection and verified by immunofluorescence and western-blotting. Cell cycle, E6 gene mutations, APOBEC3s/E6 gene expression and subcellular localization were detected by FACS, 3D-PCR and sequencing, qRT-PCR and immunofluorescence respectively. RESULTS The results suggested that A3A-HPV11.HaCaT cells were successfully established. Enhanced A3A induced S-phase arrest, G > A/C > T mutations and obvious reduction of E6 mRNA expression. A3A/A3B mRNA expression was up-regulated at 6 h and 12 h and obvious A3A staining existed throughout HPV11.HaCaT cells after rhIFN-ω treatment. RhIFN-ω could also inhibit mRNA expression of HPV11 E6 significantly. CONCLUSIONS Enhanced A3A repressed HPV11 E6 expression through gene hypermutation, and rhIFN-ω might be an effective agent against HPV11 infection by up-regulation of A3A.
Collapse
|
17
|
Hu J, Cladel NM, Budgeon LR, Balogh KK, Christensen ND. The Mouse Papillomavirus Infection Model. Viruses 2017; 9:v9090246. [PMID: 28867783 PMCID: PMC5618012 DOI: 10.3390/v9090246] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/28/2022] Open
Abstract
The mouse papillomavirus (MmuPV1) was first reported in 2011 and has since become a powerful research tool. Through collective efforts from different groups, significant progress has been made in the understanding of molecular, virological, and immunological mechanisms of MmuPV1 infections in both immunocompromised and immunocompetent hosts. This mouse papillomavirus provides, for the first time, the opportunity to study papillomavirus infections in the context of a small common laboratory animal for which abundant reagents are available and for which many strains exist. The model is a major step forward in the study of papillomavirus disease and pathology. In this review, we summarize studies using MmuPV1 over the past six years and share our perspectives on the value of this unique model system. Specifically, we discuss viral pathogenesis in cutaneous and mucosal tissues as well as in different mouse strains, immune responses to the virus, and local host-restricted factors that may be involved in MmuPV1 infections and associated disease progression.
Collapse
Affiliation(s)
- Jiafen Hu
- The Jake Gittlen Laboratories for Cancer Research, Hershey, PA 17033, USA.
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Nancy M Cladel
- The Jake Gittlen Laboratories for Cancer Research, Hershey, PA 17033, USA.
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Lynn R Budgeon
- The Jake Gittlen Laboratories for Cancer Research, Hershey, PA 17033, USA.
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Karla K Balogh
- The Jake Gittlen Laboratories for Cancer Research, Hershey, PA 17033, USA.
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Neil D Christensen
- The Jake Gittlen Laboratories for Cancer Research, Hershey, PA 17033, USA.
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
18
|
Cardoso MDFS, Castelletti CHM, Lima-Filho JLD, Martins DBG, Teixeira JAC. Putative biomarkers for cervical cancer: SNVs, methylation and expression profiles. MUTATION RESEARCH-REVIEWS IN MUTATION RESEARCH 2017; 773:161-173. [PMID: 28927526 DOI: 10.1016/j.mrrev.2017.06.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 02/08/2023]
Abstract
Cervical cancer is primarily caused by Human papillomavirus (HPV) infection, but other factors such as smoking habits, co-infections and genetic background, can also contribute to its development. Although this cancer is avoidable, it is the fourth most frequent type of cancer in females worldwide and can only be treated with chemotherapy and radical surgery. There is a need for biomarkers that will enable early diagnosis and targeted therapy for this type of cancer. Therefore, a systems biology pipeline was applied in order to identify potential biomarkers for cervical cancer, which show significant reports in three molecular aspects: DNA sequence variants, DNA methylation pattern and alterations in mRNA/protein expression levels. CDH1, CDKN2A, RB1 and TP53 genes were selected as putative biomarkers, being involved in metastasis, cell cycle regulation and tumour suppression. Other ten genes (CDH13, FHIT, PTEN, MLH1, TP73, CDKN1A, CACNA2D2, TERT, WIF1, APC) seemed to play a role in cervical cancer, but the lack of studies prevented their inclusion as possible biomarkers. Our results highlight the importance of these genes. However, further studies should be performed to elucidate the impact of DNA sequence variants and/or epigenetic deregulation and altered expression of these genes in cervical carcinogenesis and their potential as biomarkers for cervical cancer diagnosis and prognosis.
Collapse
Affiliation(s)
- Maria de Fátima Senra Cardoso
- Molecular Prospection and Bioinformatics Group (ProspecMol), Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil.
| | - Carlos Henrique Madeiros Castelletti
- Molecular Prospection and Bioinformatics Group (ProspecMol), Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil; Agronomic Institute of Pernambuco (IPA), Av. General San Martin 1371, Bongi, Recife - PE, 50761-000, Brazil
| | - José Luiz de Lima-Filho
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil; Biochemistry Department, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil
| | - Danyelly Bruneska Gondim Martins
- Molecular Prospection and Bioinformatics Group (ProspecMol), Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil; Biochemistry Department, Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil
| | - José António Couto Teixeira
- Laboratory of Immunopathology Keizo Asami (LIKA), Federal University of Pernambuco (UFPE), Av. Prof. Moraes Rego s/n, Recife - PE, 50670-901, Brazil; Department of Biological Engineering, University of Minho (UM), Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
19
|
Ma W, Melief CJ, van der Burg SH. Control of immune escaped human papilloma virus is regained after therapeutic vaccination. Curr Opin Virol 2017; 23:16-22. [PMID: 28282583 DOI: 10.1016/j.coviro.2017.02.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 02/20/2017] [Indexed: 11/15/2022]
Abstract
High-risk human papillomaviruses infect the basal cells of human epithelia. There it deploys several mechanisms to suppress pathogen receptor recognition signalling, impeding the immune system to control viral infection. Furthermore, infected cells become more resistant to type I and II interferon, tumour necrosis factor-α and CD40 activation, via interference with downstream programs halting viral replication or regulating the proliferation and cell death. Consequently, some infected individuals fail to raise early protein-specific T-cell responses that are strong enough to protect against virus-induced premalignant disease and ultimately cancer. Therapeutic vaccines triggering a strong T-cell response against the early proteins can successfully be used to treat patients at the premalignant stage but combinations of different treatment modalities are required for cancer therapy.
Collapse
Affiliation(s)
- Wenbo Ma
- Department of Medical Oncology, Building 1, C7-141, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands
| | | | - Sjoerd H van der Burg
- Department of Medical Oncology, Building 1, C7-141, Leiden University Medical Center, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| |
Collapse
|
20
|
Porter SS, Stepp WH, Stamos JD, McBride AA. Host cell restriction factors that limit transcription and replication of human papillomavirus. Virus Res 2017; 231:10-20. [PMID: 27863967 PMCID: PMC5325803 DOI: 10.1016/j.virusres.2016.11.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 02/08/2023]
Abstract
The life cycle of human papillomaviruses (HPV) is tightly regulated by the differentiation state of mucosal and cutaneous keratinocytes. To counteract viral infection, constitutively expressed cellular factors, which are defined herein as restriction factors, directly mitigate viral gene expression and replication. In turn, some HPV gene products target these restriction factors and abrogate their anti-viral effects to establish efficient gene expression and replication programs. Ironically, in certain circumstances, this delicate counterbalance between viral gene products and restriction factors facilitates persistent infection by HPVs. This review serves to recapitulate the current knowledge of nuclear restriction factors that directly affect the HPV infectious cycle.
Collapse
Affiliation(s)
- Samuel S Porter
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, 33 North Drive, MSC3209, Bethesda, MD 20892, USA; Biological Sciences Graduate Program, University of Maryland, University of Maryland, 4066 Campus Drive, College Park, MD 20742, USA
| | - Wesley H Stepp
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, 33 North Drive, MSC3209, Bethesda, MD 20892, USA
| | - James D Stamos
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, 33 North Drive, MSC3209, Bethesda, MD 20892, USA
| | - Alison A McBride
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Disease, National Institutes of Health, 33 North Drive, MSC3209, Bethesda, MD 20892, USA.
| |
Collapse
|
21
|
Egawa N, Doorbar J. The low-risk papillomaviruses. Virus Res 2016; 231:119-127. [PMID: 28040475 DOI: 10.1016/j.virusres.2016.12.017] [Citation(s) in RCA: 155] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Revised: 12/20/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Abstract
Human Papillomavirus (HPV) research has been dominated by the study of a subset of Alpha papillomaviruses that together cause almost 5% of human cancers worldwide, with the focus being on the two most prominent of these (HPV16 and 18). These viruses are referred to as 'high-risk' (hrHPV), to distinguish them from the over 200 prevalent HPV types that more commonly cause only benign epithelial lesions. The 'low-risk' (lrHPV) term used to describe this group belies their cumulative morbidity. Persistent laryngeal papillomas, which occur rarely in children and adults, require regular surgical de-bulking to allow breathing. Such infections are not curable, and despite being caused by HPV11 (a lrHPV) are associated with 1-3% risk of cancer progression if not resolved. Similarly, the ubiquitous Beta HPV types, which commonly cause asymptomatic infections at cutaneous sites, can sometimes cause debilitating papillomatosis with associated cancer risk. Recalcitrant genital warts, which affect 1 in 200 young adults in the general population, and even the ubiquitous common warts and verrucas that most of us at some time experience, cannot be reliably eradicated, with treatment strategies advancing little over the last 100 years. The review highlights molecular similarities between high and low-risk HPV types, and focuses on the different pathways that the two groups use to ensure persistent infection and adequate virus shedding from the epithelial surface. Understanding the normal patterns of viral gene expression that underlie lesion formation, and which also prevent loss of the infected basal cells in established lesions, are particularly important when considering new treatment options. Finally, the common requirement for deregulated viral gene expression and genome persistence in development of cancers, unites both high and low-risk HPV types, and when considered alongside viral protein functions, provides us with a working understanding of the mechanisms that underlie HPV-associated pathology.
Collapse
Affiliation(s)
- Nagayasu Egawa
- Department of Pathology, Tennis Court Road, University of Cambridge, Cambridge, UK
| | - John Doorbar
- Department of Pathology, Tennis Court Road, University of Cambridge, Cambridge, UK.
| |
Collapse
|
22
|
Schiffman M, Doorbar J, Wentzensen N, de Sanjosé S, Fakhry C, Monk BJ, Stanley MA, Franceschi S. Carcinogenic human papillomavirus infection. Nat Rev Dis Primers 2016; 2:16086. [PMID: 27905473 DOI: 10.1038/nrdp.2016.86] [Citation(s) in RCA: 587] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Infections with human papillomavirus (HPV) are common and transmitted by direct contact. Although the great majority of infections resolve within 2 years, 13 phylogenetically related, sexually transmitted HPV genotypes, notably HPV16, cause - if not controlled immunologically or by screening - virtually all cervical cancers worldwide, a large fraction of other anogenital cancers and an increasing proportion of oropharyngeal cancers. The carcinogenicity of these HPV types results primarily from the activity of the oncoproteins E6 and E7, which impair growth regulatory pathways. Persistent high-risk HPVs can transition from a productive (virion-producing) to an abortive or transforming infection, after which cancer can result after typically slow accumulation of host genetic mutations. However, which precancerous lesions progress and which do not is unclear; the majority of screening-detected precancers are treated, leading to overtreatment. The discovery of HPV as a carcinogen led to the development of effective preventive vaccines and sensitive HPV DNA and RNA tests. Together, vaccination programmes (the ultimate long-term preventive strategy) and screening using HPV tests could dramatically alter the landscape of HPV-related cancers. HPV testing will probably replace cytology-based cervical screening owing to greater reassurance when the test is negative. However, the effective implementation of HPV vaccination and screening globally remains a challenge.
Collapse
Affiliation(s)
- Mark Schiffman
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Room 6E544, 9609 Medical Center Drive, Rockville, Maryland 20850, USA
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nicolas Wentzensen
- National Cancer Institute, Division of Cancer Epidemiology and Genetics, Room 6E544, 9609 Medical Center Drive, Rockville, Maryland 20850, USA
| | - Silvia de Sanjosé
- Catalan Institute of Oncology, IDIBELL, Cancer Epidemiology Research Programme and CIBER Epidemiologia Y Salud Publica, Barcelona, Spain
| | - Carole Fakhry
- Department of Otolaryngology, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Bradley J Monk
- Division of Gynecologic Oncology, US Oncology Network, University of Arizona-Phoenix, Phoenix, Arizona, USA
| | | | - Silvia Franceschi
- International Agency for Research on Cancer, Infections and Cancer Epidemiology Group, Lyon, France
| |
Collapse
|
23
|
Ma W, Tummers B, van Esch EMG, Goedemans R, Melief CJM, Meyers C, Boer JM, van der Burg SH. Human Papillomavirus Downregulates the Expression of IFITM1 and RIPK3 to Escape from IFNγ- and TNFα-Mediated Antiproliferative Effects and Necroptosis. Front Immunol 2016; 7:496. [PMID: 27920775 PMCID: PMC5118436 DOI: 10.3389/fimmu.2016.00496] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/26/2016] [Indexed: 01/29/2023] Open
Abstract
The clearance of a high-risk human papillomavirus (hrHPV) infection takes time and requires the local presence of a strong type 1 cytokine T cell response, suggesting that hrHPV has evolved mechanisms to resist this immune attack. Using an unique system for non, newly, and persistent hrHPV infection, we show that hrHPV infection renders keratinocytes (KCs) resistant to the antiproliferative- and necroptosis-inducing effects of IFNγ and TNFα. HrHPV-impaired necroptosis was associated with the upregulation of several methyltransferases, including EZH2, and the downregulation of RIPK3 expression. Restoration of RIPK3 expression using the global histone methyltransferase inhibitor 3-deazaneplanocin increased necroptosis in hrHPV-positive KCs. Simultaneously, hrHPV effectively inhibited IFNγ/TNFα-mediated arrest of cell growth at the S-phase by downregulating IFITM1 already at 48 h after hrHPV infection, followed by an impaired increase in the expression of the antiproliferative gene RARRES1 and a decrease of the proliferative gene PCNA. Knockdown of IFITM1 in uninfected KCs confirmed its role on RARRES1 and its antiproliferative effects. Thus, our study reveals how hrHPV deregulates two pathways involved in cell death and growth regulation to withstand immune-mediated control of hrHPV-infected cells.
Collapse
Affiliation(s)
- Wenbo Ma
- Department of Medical Oncology, Leiden University Medical Center , Leiden , Netherlands
| | - Bart Tummers
- Department of Medical Oncology, Leiden University Medical Center , Leiden , Netherlands
| | - Edith M G van Esch
- Department of Gynaecology, Leiden University Medical Center , Leiden , Netherlands
| | - Renske Goedemans
- Department of Medical Oncology, Leiden University Medical Center , Leiden , Netherlands
| | - Cornelis J M Melief
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Center , Leiden , Netherlands
| | - Craig Meyers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine , Hershey, PA , USA
| | - Judith M Boer
- Human Genetics, Leiden University Medical Center , Leiden , Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
24
|
Guven-Maiorov E, Keskin O, Gursoy A, VanWaes C, Chen Z, Tsai CJ, Nussinov R. TRAF3 signaling: Competitive binding and evolvability of adaptive viral molecular mimicry. Biochim Biophys Acta Gen Subj 2016; 1860:2646-55. [PMID: 27208423 PMCID: PMC7117012 DOI: 10.1016/j.bbagen.2016.05.021] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 04/18/2016] [Accepted: 05/08/2016] [Indexed: 12/16/2022]
Abstract
BACKGROUND The tumor necrosis factor receptor (TNFR) associated factor 3 (TRAF3) is a key node in innate and adaptive immune signaling pathways. TRAF3 negatively regulates the activation of the canonical and non-canonical NF-κB pathways and is one of the key proteins in antiviral immunity. SCOPE OF REVIEW Here we provide a structural overview of TRAF3 signaling in terms of its competitive binding and consequences to the cellular network. For completion, we also include molecular mimicry of TRAF3 physiological partners by some viral proteins. MAJOR CONCLUSIONS By out-competing host partners, viral proteins aim to subvert TRAF3 antiviral action. Mechanistically, dynamic, competitive binding by the organism's own proteins and same-site adaptive pathogen mimicry follow the same conformational selection principles. GENERAL SIGNIFICANCE Our premise is that irrespective of the eliciting event - physiological or acquired pathogenic trait - pathway activation (or suppression) may embrace similar conformational principles. However, even though here we largely focus on competitive binding at a shared site, similar to physiological signaling other pathogen subversion mechanisms can also be at play. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
Affiliation(s)
- Emine Guven-Maiorov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702,USA.
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc University, Istanbul, Turkey; Center for Computational Biology and Bioinformatics, Koc University, Istanbul, Turkey.
| | - Attila Gursoy
- Center for Computational Biology and Bioinformatics, Koc University, Istanbul, Turkey; Department of Computer Engineering, Koc University, Istanbul, Turkey.
| | - Carter VanWaes
- Clinical Genomic Unit, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, NIH, Bethesda, MD 20892, USA.
| | - Zhong Chen
- Clinical Genomic Unit, Head and Neck Surgery Branch, National Institute on Deafness and Communication Disorders, NIH, Bethesda, MD 20892, USA.
| | - Chung-Jung Tsai
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702,USA.
| | - Ruth Nussinov
- Cancer and Inflammation Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21702,USA; Sackler Inst. of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
25
|
Guven-Maiorov E, Tsai CJ, Nussinov R. Pathogen mimicry of host protein-protein interfaces modulates immunity. Semin Cell Dev Biol 2016; 58:136-45. [DOI: 10.1016/j.semcdb.2016.06.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 06/02/2016] [Accepted: 06/06/2016] [Indexed: 12/21/2022]
|
26
|
Friedenson B. Comment on 'The incidence of leukaemia in women with BRCA1 and BRCA2 mutations: an International Prospective Cohort Study'. Br J Cancer 2016; 115:e2. [PMID: 27459694 PMCID: PMC4997539 DOI: 10.1038/bjc.2016.192] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Bernard Friedenson
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| |
Collapse
|
27
|
Chuerduangphui J, Pientong C, Chaiyarit P, Patarapadungkit N, Chotiyano A, Kongyingyoes B, Promthet S, Swangphon P, Wongjampa W, Ekalaksananan T. Effect of human papillomavirus 16 oncoproteins on oncostatin M upregulation in oral squamous cell carcinoma. Med Oncol 2016; 33:83. [PMID: 27349249 DOI: 10.1007/s12032-016-0800-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Accepted: 06/21/2016] [Indexed: 01/17/2023]
Abstract
Human papillomavirus (HPV) infection modulates several host cytokines contributing to cancer development. Oncostatin M (OSM), an IL-6 family cytokine, acts to promote cell senescence and inhibit growth. Its dysregulation promotes cell survival, cell proliferation and metastasis in various malignancies. The effect of HPV on OSM dysregulation has not been investigated. To elucidate this, immunohistochemistry was used on formalin-fixed, paraffin-embedded oral squamous cell carcinoma (OSCC) tissues: HPV-positive (50) and HPV-negative (50) cases. Immortalized human cervical keratinocytes expressing HPV16E6 (HCK1T, Tet-On system) were used to demonstrate the role of HPV16E6 in OSM expression. In addition, a vector containing HPV16E6/E7 was transiently transfected into oral cancer cell lines. Cell viability, cell-cycle progression and cell migration were evaluated using flow cytometry and a wound healing assay, respectively. The results showed various intensities of OSM expression in OSCC. Interestingly, the median percentages of strongly stained cells were significantly higher in HPV-positive OSCCs than in HPV-negative OSCCs. To explore the role of HPV oncoproteins on OSM expression, the expression of HPV16E6 in the HCK1T Tet-On condition was induced by doxycycline and HPV16E6 was found to significantly upregulate levels of OSM mRNA and protein, with concomitant upregulation of c-Myc. In addition, the levels of OSM mRNA and protein in E6/E7 transiently transfected oral cancer cells also gradually increased in a time-dependent manner and these transfected cells showed greater viability and higher migration rates and cell-cycle progression than controls. This result demonstrates that HPV16 oncoproteins upregulate OSM and play an important role to promote OSCC development.
Collapse
Affiliation(s)
- Jureeporn Chuerduangphui
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Ponlatham Chaiyarit
- Department of Oral Diagnosis, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand.,Research Group of Chronic Inflammatory Oral Diseases and Systemic Diseases Associated with Oral Health, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Natcha Patarapadungkit
- Department of Pathology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Apinya Chotiyano
- Anatomical Pathology Unit, Khon Kaen Hospital, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Bunkerd Kongyingyoes
- Department of Pharmacology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Supannee Promthet
- Department of Epidemiology, Faculty of Public Health, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Piyawut Swangphon
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Weerayut Wongjampa
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand. .,HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
28
|
Poltorak A, Kurmyshkina O, Volkova T. Stimulator of interferon genes (STING): A "new chapter" in virus-associated cancer research. Lessons from wild-derived mouse models of innate immunity. Cytokine Growth Factor Rev 2016; 29:83-91. [PMID: 26980676 DOI: 10.1016/j.cytogfr.2016.02.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/27/2016] [Indexed: 12/19/2022]
Abstract
Thanks to the numerous studies that have been carried out recently in the field of cytosolic DNA sensing, STING (Stimulator of Interferon Genes) is now recognized as a key mediator of innate immune signaling. A substantial body of evidence derived from in vivo mouse models demonstrates that STING-regulated pathways underlie the pathogenesis of many diseases including infectious diseases and cancers. It has also become evident from these studies that STING is a promising therapeutic target for the treatment of cancer. However, mouse strains commonly used for modelling innate immune response against infections or tumors do not allow investigators to accurately reproduce certain specific characteristics of immune response observed in human cells. In this review, we will discuss recent data demonstrating that the use of wild-derived genetically distinct inbred mice as a model for investigation into the innate immunity signaling networks may provide valuable insight into the STING-regulated pathways specific for human cells. The maximum complexity of STING-mediated mechanisms can probably be seen in case of DNA virus-induced carcinogenesis in which STING may perform unexpected biological activities. Therefore, in another part of this review we will summarize emerging data on the role of STING in human DNA virus-related oncopathologies, with particular attention to HPV-associated cervical cancer, aiming to demonstrate that STING indeed "starts a new chapter" in research on this issue and that wild-derived mouse models of STING-mediated response to infections will probably be helpful in finding out molecular basis for clinical observations.
Collapse
Affiliation(s)
- Alexander Poltorak
- Department of Integrative Physiology and Pathobiology, Tufts University School of Medicine, 136 Harrison Ave., Boston, MA 02111, United States; Institute of High-Tech Biomedicine, Petrozavodsk State University, 33 Lenin str., Petrozavodsk 185910, Russian Federation.
| | - Olga Kurmyshkina
- Institute of High-Tech Biomedicine, Petrozavodsk State University, 33 Lenin str., Petrozavodsk 185910, Russian Federation.
| | - Tatyana Volkova
- Institute of High-Tech Biomedicine, Petrozavodsk State University, 33 Lenin str., Petrozavodsk 185910, Russian Federation.
| |
Collapse
|
29
|
Nakahara T, Kiyono T. Interplay between NF-κB/interferon signaling and the genome replication of HPV. Future Virol 2016. [DOI: 10.2217/fvl.16.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
HPV infection can persist within the infected epithelium for years. The viral persistence is primarily attributed to the ability of the virus to maintain its genome as nuclear episomes in the basal cells. Recent studies have revealed that HPV induces DNA damage response to facilitate productive amplification of the viral genome. DNA damage response comprises a part of the cellular defense mechanism against viral infection and its activation can result in induction of innate immune responses. The activation of NF-κB and interferon (IFN) signals has been shown to suppress the genome replication of HPV while the viral proteins inhibit NF-κB/IFN signaling. This review intends to focus on illustrating the interplay between NFκB/IFN signaling and HPV genome replication in the HPV life cycle.
Collapse
Affiliation(s)
- Tomomi Nakahara
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tokyo, Japan, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Tohru Kiyono
- Division of Carcinogenesis and Cancer Prevention, National Cancer Center Research Institute, Tokyo, Japan, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| |
Collapse
|
30
|
|