1
|
Irina P, Alena V, Arsene MMJ, Milana D, Alla P, Lyudmila K, Boris E. Comparison of Vaginal microbiota in HPV-negative and HPV-positive pregnant women using a culture-based approach. Diagn Microbiol Infect Dis 2024; 110:116419. [PMID: 39116654 DOI: 10.1016/j.diagmicrobio.2024.116419] [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: 04/04/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024]
Abstract
The purpose of this study was to conduct a comparative analysis of the composition of the dominant groups of vaginal microorganisms in healthy pregnant women and pregnant women infected with HPV using a microbiological culture-based method. The MALDI TOF MS method and 16S rRNA gene fragment sequencing were used to identify microorganisms isolated from healthy pregnant women (n=32) and pregnant women infected with HPV (n=24). It was found that vaginal secretion samples from both groups contained bacteria of 4 phyla: Bacillota, Actinomycetota, Pseudomonadota, Bacteroidota, and Ascomycota fungi. The most common microbial community in healthy pregnant women being CST I (p=0.0007), and CST V in pregnant women infected with HPV (p=0.0001). At the genus level, a total of 25 taxa were found in all samples, with Lactobacillus being the dominant genus overall. Escherichia (p<0.0001) and Prevotella (p=0.0001) concentrations were higher in HPV infected patients. When calculating the Pearson correlation coefficient for the phyla, it was found that Bacillota correlated negatively with HPV genotypes 16 and 51 (p≤0.05), but positively with HPV genotype 59 (p≤0.05), just like Actinomycetota (p≤0.05). Bacteroidota correlated positively with HPV genotype 56 (0.001
Collapse
Affiliation(s)
- Podoprigora Irina
- Department of Microbiology named after V.S. Kiktenko, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia.
| | - Vasina Alena
- Mytishchi Regional Clinical Hospital, Mytishchi, Russia
| | - Mbarga Manga Joseph Arsene
- Department of Microbiology named after V.S. Kiktenko, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia.
| | - Das Milana
- Department of Microbiology named after V.S. Kiktenko, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia; Research Institute of Molecular and Cellular Medicine, Medical Institute, RUDN University named after Patrice Lumumba, Moscow, Russia
| | - Pikina Alla
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Russia
| | - Kafarskaya Lyudmila
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Russia
| | - Efimov Boris
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Russia
| |
Collapse
|
2
|
Trejo-Cerro O, Basukala O, Myers MP, Banks L. HPV16 E7 modulates the cell surface expression of MET and CD109 via the AP2 complex. Tumour Virus Res 2024; 17:200279. [PMID: 38485055 PMCID: PMC10958106 DOI: 10.1016/j.tvr.2024.200279] [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: 01/23/2024] [Revised: 03/11/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024] Open
Abstract
Multiple cellular pathways are affected by HPV E6 and E7 oncoproteins, including endocytic and cellular trafficking. HPV-16 E7 can target the adaptor protein (AP) complex, which contains proteins important during endocytosis transport. To further investigate the role of HPV E7 during this process, we analysed the expression of cell surface proteins in NIKS cells expressing HPV-16 E7. We show that different cell surface proteins are regulated by HPV-16 E7 via interaction with AP2. We observed that the expression of MET and CD109 membrane protein seems to be upregulated in cells expressing E7. Moreover, the interaction of MET and CD109 with AP2 proteins is disrupted by HPV-16 E7. In addition, in the absence of HPV-16 E7, there is a downregulation of the cell membrane expression of MET and CD109 in HPV-positive cell lines. These results expand our knowledge of the functions of E7 and open new potential cellular pathways affected by this oncoprotein.
Collapse
Affiliation(s)
- Oscar Trejo-Cerro
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy.
| | - Om Basukala
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy; Dana-Farber Cancer Institute, 450 Brookline Avenue, Mayer 440, Boston, MA, 02215, USA
| | - Michael P Myers
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy
| | - Lawrence Banks
- International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149, Trieste, Italy
| |
Collapse
|
3
|
Ferrarone JR, Thomas J, Unni AM, Zheng Y, Nagiec MJ, Gardner EE, Mashadova O, Li K, Koundouros N, Montalbano A, Mustafa M, Cantley LC, Blenis J, Sanjana NE, Varmus H. Genome-wide CRISPR screens in spheroid culture reveal that the tumor suppressor LKB1 inhibits growth via the PIKFYVE lipid kinase. Proc Natl Acad Sci U S A 2024; 121:e2403685121. [PMID: 38743625 PMCID: PMC11127050 DOI: 10.1073/pnas.2403685121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024] Open
Abstract
The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor-suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1-mediated growth suppression, we developed a spheroid-based cell culture assay to study LKB1-dependent growth. We then performed genome-wide CRISPR screens in spheroidal culture and found that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase. Finally, we used chemical inhibitors and a pH-sensitive reporter to determine that LKB1 impairs growth by promoting the internalization of wild-type EGFR in a PIKFYVE-dependent manner.
Collapse
Affiliation(s)
- John R. Ferrarone
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Cornell Medicine, New York, NY10021
| | - Jerin Thomas
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Arun M. Unni
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Yuxiang Zheng
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Michal J. Nagiec
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Eric E. Gardner
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | | | - Kate Li
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
| | - Nikos Koundouros
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Antonino Montalbano
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Meer Mustafa
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Lewis C. Cantley
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Medicine, Weill Cornell Medicine, New York, NY10021
| | - John Blenis
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Pharmacology, Weill Cornell Medicine, New York, NY10021
| | - Neville E. Sanjana
- New York Genome Center, New York, NY10013
- Department of Biology, New York University, New York, NY10003
| | - Harold Varmus
- Meyer Cancer Center, Weill Cornell Medicine, New York, NY10021
- Department of Medicine, Weill Cornell Medicine, New York, NY10021
| |
Collapse
|
4
|
Trammel J, Amusan O, Hultgren A, Raikhy G, Bodily JM. Epidermal growth factor receptor-dependent stimulation of differentiation by human papillomavirus type 16 E5. Virology 2024; 590:109952. [PMID: 38103269 PMCID: PMC10842332 DOI: 10.1016/j.virol.2023.109952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/14/2023] [Accepted: 11/22/2023] [Indexed: 12/19/2023]
Abstract
Human papillomaviruses (HPVs) infect keratinocytes of stratified squamous epithelia, and persistent infection with high-risk HPV types, such as HPV16, may lead to the development of malignancies. HPV evades host immunity in part by linking its gene expression to the host differentiation program, and therefore relies on differentiation to complete its life cycle. Based on previous reports indicating that the HPV16 protein E5 is important in the late stages of the differentiation-dependent life cycle, we found that organotypic cultures harboring HPV16 genomes lacking E5 showed reduced markers of terminal differentiation compared to wild type HPV16-containing cultures. We found that epidermal growth factor receptor (EGFR) levels and activation were increased in an E5-depdendent manner in these tissues, and that EGFR promoted terminal differentiation and expression of the HPV16 L1 gene. These findings suggest a function for E5 in preserving the ability of HPV16 containing keratinocytes to differentiate, thus facilitating the production of new virus progeny.
Collapse
Affiliation(s)
- Jessica Trammel
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Oluwamuyiwa Amusan
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Allison Hultgren
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA; School of Medicine, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Gaurav Raikhy
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA
| | - Jason M Bodily
- Department of Microbiology and Immunology, Louisiana State University Health Sciences Center - Shreveport, Shreveport, LA, 71103, USA.
| |
Collapse
|
5
|
Chen B, Wang Y, Wu Y, Xu T. Effect of HPV Oncoprotein on Carbohydrate and Lipid Metabolism in Tumor Cells. Curr Cancer Drug Targets 2024; 24:987-1004. [PMID: 38284713 DOI: 10.2174/0115680096266981231215111109] [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: 07/09/2023] [Revised: 09/29/2023] [Accepted: 11/13/2023] [Indexed: 01/30/2024]
Abstract
High-risk HPV infection accounts for 99.7% of cervical cancer, over 90% of anal cancer, 50% of head and neck cancers, 40% of vulvar cancer, and some cases of vaginal and penile cancer, contributing to approximately 5% of cancers worldwide. The development of cancer is a complex, multi-step process characterized by dysregulation of signaling pathways and alterations in metabolic pathways. Extensive research has demonstrated that metabolic reprogramming plays a key role in the progression of various cancers, such as cervical, head and neck, bladder, and prostate cancers, providing the material and energy foundation for rapid proliferation and migration of cancer cells. Metabolic reprogramming of tumor cells allows for the rapid generation of ATP, aiding in meeting the high energy demands of HPV-related cancer cell proliferation. The interaction between Human Papillomavirus (HPV) and its associated cancers has become a recent focus of investigation. The impact of HPV on cellular metabolism has emerged as an emerging research topic. A significant body of research has shown that HPV influences relevant metabolic signaling pathways, leading to cellular metabolic alterations. Exploring the underlying mechanisms may facilitate the discovery of biomarkers for diagnosis and treatment of HPV-associated diseases. In this review, we introduced the molecular structure of HPV and its replication process, discussed the diseases associated with HPV infection, described the energy metabolism of normal cells, highlighted the metabolic features of tumor cells, and provided an overview of recent advances in potential therapeutic targets that act on cellular metabolism. We discussed the potential mechanisms underlying these changes. This article aims to elucidate the role of Human Papillomavirus (HPV) in reshaping cellular metabolism and the application of metabolic changes in the research of related diseases. Targeting cancer metabolism may serve as an effective strategy to support traditional cancer treatments, as metabolic reprogramming is crucial for malignant transformation in cancer.
Collapse
Affiliation(s)
- Biqing Chen
- The Second Hospital of Jilin University, Changchun, China
| | - Yichao Wang
- The Second Hospital of Jilin University, Changchun, China
| | - Yishi Wu
- The Second Hospital of Jilin University, Changchun, China
| | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Ferrarone JR, Thomas J, Unni AM, Zheng Y, Nagiec MJ, Gardner EE, Mashadova O, Li K, Koundouros N, Montalbano A, Mustafa M, Cantley LC, Blenis J, Sanjana NE, Varmus H. LKB1 suppresses growth and promotes the internalization of EGFR through the PIKFYVE lipid kinase. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563158. [PMID: 37904985 PMCID: PMC10614957 DOI: 10.1101/2023.10.19.563158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The tumor suppressor LKB1 is a serine/threonine protein kinase that is frequently mutated in human lung adenocarcinoma (LUAD). LKB1 regulates a complex signaling network that is known to control cell polarity and metabolism; however, the pathways that mediate the tumor suppressive activity of LKB1 are incompletely defined. To identify mechanisms of LKB1- mediated growth suppression we developed a spheroid-based cell culture assay to study LKB1- dependent growth. Using this assay, along with genome-wide CRISPR screens and validation with orthogonal methods, we discovered that LKB1 suppresses growth, in part, by activating the PIKFYVE lipid kinase, which promotes the internalization of wild-type EGFR. Our findings reveal a new mechanism of regulation of EGFR, which may have implications for the treatment of LKB1 -mutant LUAD.
Collapse
|
7
|
Chen B, Zhao L, Yang R, Xu T. Advances in molecular mechanism of HPV16 E5 oncoprotein carcinogenesis. Arch Biochem Biophys 2023; 745:109716. [PMID: 37553047 DOI: 10.1016/j.abb.2023.109716] [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: 05/16/2023] [Revised: 08/02/2023] [Accepted: 08/05/2023] [Indexed: 08/10/2023]
Abstract
For a considerable duration, cervical cancer has posed a significant risk to the well-being and survival of women. The emergence and progression of cervical cancer have garnered extensive attention, with prolonged chronic infection of HPV serving as a crucial etiological factor. Consequently, investigating the molecular mechanism underlying HPV-induced cervical cancer has become a prominent research area. The HPV molecule is composed of a long control region (LCR), an early coding region and a late coding region.The early coding region encompasses E1, E2, E4, E5, E6, E7, while the late coding region comprises L1 and L2 ORF.The investigation into the molecular structure and function of HPV has garnered significant attention, with the aim of elucidating the carcinogenic mechanism of HPV and identifying potential targets for the treatment of cervical cancer. Research has demonstrated that the HPV gene and its encoded protein play a crucial role in the invasion and malignant transformation of host cells. Consequently, understanding the function of HPV oncoprotein is of paramount importance in comprehending the pathogenesis of cervical cancer. E6 and E7, the primary HPV oncogenic proteins, have been the subject of extensive study. Moreover, a number of contemporary investigations have demonstrated the significant involvement of HPV16 E5 oncoprotein in the malignant conversion of healthy cells through its regulation of cell proliferation, differentiation, and apoptosis via diverse pathways, albeit the precise molecular mechanism remains unclear. This manuscript aims to provide a comprehensive account of the molecular structure and life cycle of HPV.The HPV E5 oncoprotein mechanism modulates cellular processes such as proliferation, differentiation, apoptosis, and energy metabolism through its interaction with cell growth factor receptors and other cellular proteins. This mechanism is crucial for the survival, adhesion, migration, and invasion of tumor cells in the early stages of carcinogenesis. Recent studies have identified the HPV E5 oncoprotein as a promising therapeutic target for early-stage cervical cancer, thus offering a novel approach for treatment.
Collapse
Affiliation(s)
- Biqing Chen
- The Second Hospital of Jilin University, Changchun, China
| | - Liping Zhao
- The Second Hospital of Jilin University, Changchun, China
| | - Rulin Yang
- The Second Hospital of Jilin University, Changchun, China
| | - Tianmin Xu
- The Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Isono T, Hirayama S, Domon H, Maekawa T, Tamura H, Hiyoshi T, Sirisereephap K, Takenaka S, Noiri Y, Terao Y. Degradation of EGFR on lung epithelial cells by neutrophil elastase contributes to the aggravation of pneumococcal pneumonia. J Biol Chem 2023; 299:104760. [PMID: 37119853 DOI: 10.1016/j.jbc.2023.104760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/19/2023] [Accepted: 04/20/2023] [Indexed: 05/01/2023] Open
Abstract
Streptococcus pneumoniae is the main cause of bacterial pneumonia. S. pneumoniae infection has been shown to cause elastase, an intracellular host defense factor, to leak from neutrophils. However, when neutrophil elastase (NE) leaks into the extracellular environment, it can degrade host cell surface proteins such as epidermal growth factor receptor (EGFR) and potentially disrupt the alveolar epithelial barrier. In this study, we hypothesized that NE degrades the extracellular domain of EGFR in alveolar epithelial cells and inhibits alveolar epithelial repair. Using SDS-PAGE, we showed that NE degraded the recombinant EGFR extracellular domain and its ligand EGF, and that the degradation of these proteins was counteracted by NE inhibitors. Furthermore, we confirmed the degradation by NE of EGFR expressed in alveolar epithelial cells in vitro. We show intracellular uptake of EGF and EGFR signaling were downregulated in alveolar epithelial cells exposed to NE, and found cell proliferation was inhibited in these cells These negative effects of NE on cell proliferation were abolished by NE inhibitors. Finally, we confirmed the degradation of EGFR by NE in vivo. Fragments of the extracellular domain of EGFR were detected in bronchoalveolar lavage fluid from pneumococcal pneumonia mice, and the percentage of cells positive for a cell proliferation marker Ki67 in lung tissue was reduced. In contrast, administration of an NE inhibitor decreased EGFR fragments in bronchoalveolar lavage fluid and increased the percentage of Ki67-positive cells. These findings suggest that degradation of EGFR by NE could inhibit the repair of alveolar epithelium and cause severe pneumonia.
Collapse
Affiliation(s)
- Toshihito Isono
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Satoru Hirayama
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hisanori Domon
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Tomoki Maekawa
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hikaru Tamura
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Takumi Hiyoshi
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Kridtapat Sirisereephap
- Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Division of Periodontology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Shoji Takenaka
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yuichiro Noiri
- Division of Cariology, Operative Dentistry and Endodontics, Department of Oral Health Science, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yutaka Terao
- Division of Microbiology and Infectious Diseases, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan; Center for Advanced Oral Science, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.
| |
Collapse
|
9
|
Läsche M, Gallwas J, Gründker C. Like Brothers in Arms: How Hormonal Stimuli and Changes in the Metabolism Signaling Cooperate, Leading HPV Infection to Drive the Onset of Cervical Cancer. Int J Mol Sci 2022; 23:5050. [PMID: 35563441 PMCID: PMC9103757 DOI: 10.3390/ijms23095050] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 02/06/2023] Open
Abstract
Despite all precautionary actions and the possibility of using vaccinations to counteract infections caused by human papillomaviruses (HPVs), HPV-related cancers still account for approximately 5% of all carcinomas. Worldwide, many women are still excluded from adequate health care due to their social position and origin. Therefore, immense efforts in research and therapy are still required to counteract the challenges that this disease entails. The special thing about an HPV infection is that it is not only able to trick the immune system in a sophisticated way, but also, through genetic integration into the host genome, to use all the resources available to the host cells to complete the replication cycle of the virus without activating the alarm mechanisms of immune recognition and elimination. The mechanisms utilized by the virus are the metabolic, immune, and hormonal signaling pathways that it manipulates. Since the virus is dependent on replication enzymes of the host cells, it also intervenes in the cell cycle of the differentiating keratinocytes and shifts their terminal differentiation to the uppermost layers of the squamocolumnar transformation zone (TZ) of the cervix. The individual signaling pathways are closely related and equally important not only for the successful replication of the virus but also for the onset of cervical cancer. We will therefore analyze the effects of HPV infection on metabolic signaling, as well as changes in hormonal and immune signaling in the tumor and its microenvironment to understand how each level of signaling interacts to promote tumorigenesis of cervical cancer.
Collapse
Affiliation(s)
| | | | - Carsten Gründker
- Department of Gynecology and Obstetrics, University Medicine Göttingen, 37075 Göttingen, Germany; (M.L.); (J.G.)
| |
Collapse
|
10
|
He L, Wang L, Wang Z, Li T, Chen H, Zhang Y, Hu Z, Dimitrov DS, Du J, Liao X. Immune Modulating Antibody-Drug Conjugate (IM-ADC) for Cancer Immunotherapy. J Med Chem 2021; 64:15716-15726. [PMID: 34730979 DOI: 10.1021/acs.jmedchem.1c00961] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Antibody-drug conjugate (ADC) and immune checkpoint blockade (ICB) offer promising approaches for cancer treatment. Here, we describe an ADC constructed by conjugating anti-PD-L1 THIOMAB with a bifunctional immunomodulator D18 via a redox-cleavable linker. The resulting ADC HE-S2 not only triggers a potent antitumor immune response by blocking the PD-1/PD-L1 interaction and activating the Toll-like receptor 7/8 (TLR7/8) signaling pathway but also upregulates its targeted PD-L1 expression via epigenetic regulation and/or IFN-γ induction, thus conferring more sensitivity to the PD-1/PD-L1 blockade. We identify that ADC HE-S2 treatment could lead to more pronounced tumor suppression than the treatment of D18 in combination with the anti-PD-L1 antibody. Accordingly, this study provides a novel ADC strategy to enhance the antitumor immune response to ICB therapy.
Collapse
Affiliation(s)
- Lei He
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.,Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing 100088, China
| | - Liangliang Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.,Department of Radiation and Cellular Oncology, The Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois 60637, United States
| | - Zhisong Wang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.,Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing 100088, China
| | - Tiantian Li
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Hui Chen
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.,Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing 100088, China
| | - Yaning Zhang
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Dimiter S Dimitrov
- Center for Antibody Therapeutics, University of Pittsburgh, Pittsburgh, Pennsylvania 15216, United States
| | - Juanjuan Du
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Xuebin Liao
- School of Pharmaceutical Sciences, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China.,Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Capital Medical University, Beijing 100088, China
| |
Collapse
|
11
|
Gargan S, Stevenson NJ. Unravelling the Immunomodulatory Effects of Viral Ion Channels, towards the Treatment of Disease. Viruses 2021; 13:2165. [PMID: 34834972 PMCID: PMC8618147 DOI: 10.3390/v13112165] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/07/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
The current COVID-19 pandemic has highlighted the need for the research community to develop a better understanding of viruses, in particular their modes of infection and replicative lifecycles, to aid in the development of novel vaccines and much needed anti-viral therapeutics. Several viruses express proteins capable of forming pores in host cellular membranes, termed "Viroporins". They are a family of small hydrophobic proteins, with at least one amphipathic domain, which characteristically form oligomeric structures with central hydrophilic domains. Consequently, they can facilitate the transport of ions through the hydrophilic core. Viroporins localise to host membranes such as the endoplasmic reticulum and regulate ion homeostasis creating a favourable environment for viral infection. Viroporins also contribute to viral immune evasion via several mechanisms. Given that viroporins are often essential for virion assembly and egress, and as their structural features tend to be evolutionarily conserved, they are attractive targets for anti-viral therapeutics. This review discusses the current knowledge of several viroporins, namely Influenza A virus (IAV) M2, Human Immunodeficiency Virus (HIV)-1 Viral protein U (Vpu), Hepatitis C Virus (HCV) p7, Human Papillomavirus (HPV)-16 E5, Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV) Open Reading Frame (ORF)3a and Polyomavirus agnoprotein. We highlight the intricate but broad immunomodulatory effects of these viroporins and discuss the current antiviral therapies that target them; continually highlighting the need for future investigations to focus on novel therapeutics in the treatment of existing and future emergent viruses.
Collapse
Affiliation(s)
- Siobhan Gargan
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland;
| | - Nigel J. Stevenson
- Viral Immunology Group, School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, D02 R590 Dublin, Ireland;
- Viral Immunology Group, Royal College of Surgeons in Ireland-Medical University of Bahrain, Manama 15503, Bahrain
| |
Collapse
|
12
|
Basukala O, Banks L. The Not-So-Good, the Bad and the Ugly: HPV E5, E6 and E7 Oncoproteins in the Orchestration of Carcinogenesis. Viruses 2021; 13:1892. [PMID: 34696321 PMCID: PMC8541208 DOI: 10.3390/v13101892] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
Infection with HPV starts with the access of the viral particles to basal cells in the epidermis, potentially via microtraumas to the skin. The basal cells are able to keep away these pathogens in normal circumstances through a robust immune response from the host, as HPV infections are, in general, cleared within 2 to 3 weeks. However, the rare instances of persistent infection and/or in cases where the host immune system is compromised are major risk factors for the development of lesions potentially leading to malignancy. Evolutionarily, obligatory pathogens such as HPVs would not be expected to risk exposing the host to lethal cancer, as this would entail challenging their own life cycle, but infection with these viruses is highly correlated with cancer and malignancy-as in cancer of the cervix, which is almost always associated with these viruses. Despite this key associative cause and the availability of very effective vaccines against these viruses, therapeutic interventions against HPV-induced cancers are still a challenge, indicating the need for focused translational research. In this review, we will consider the key roles that the viral proteins play in driving the host cells to carcinogenesis, mainly focusing on events orchestrated by early proteins E5, E6 and E7-the not-so-good, the bad and the ugly-and discuss and summarize the major events that lead to these viruses mechanistically corrupting cellular homeostasis, giving rise to cancer and malignancy.
Collapse
Affiliation(s)
| | - Lawrence Banks
- Tumour Virology Laboratory, International Centre for Genetic Engineering and Biotechnology, Padriciano 99, I-34149 Trieste, Italy;
| |
Collapse
|
13
|
Scarth JA, Patterson MR, Morgan EL, Macdonald A. The human papillomavirus oncoproteins: a review of the host pathways targeted on the road to transformation. J Gen Virol 2021; 102:001540. [PMID: 33427604 PMCID: PMC8148304 DOI: 10.1099/jgv.0.001540] [Citation(s) in RCA: 90] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 11/25/2020] [Indexed: 12/24/2022] Open
Abstract
Persistent infection with high-risk human papillomaviruses (HR-HPVs) is the causal factor in over 99 % of cervical cancer cases, and a significant proportion of oropharyngeal and anogenital cancers. The key drivers of HPV-mediated transformation are the oncoproteins E5, E6 and E7. Together, they act to prolong cell-cycle progression, delay differentiation and inhibit apoptosis in the host keratinocyte cell in order to generate an environment permissive for viral replication. The oncoproteins also have key roles in mediating evasion of the host immune response, enabling infection to persist. Moreover, prolonged infection within the cellular environment established by the HR-HPV oncoproteins can lead to the acquisition of host genetic mutations, eventually culminating in transformation to malignancy. In this review, we outline the many ways in which the HR-HPV oncoproteins manipulate the host cellular environment, focusing on how these activities can contribute to carcinogenesis.
Collapse
Affiliation(s)
- James A. Scarth
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Molly R. Patterson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Ethan L. Morgan
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Present address: Tumour Biology Section, Head and Neck Surgery Branch, National Institute on Deafness and Other Communication Disorders, National Institute of Health, Bethesda, MD 20892, USA
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
- Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| |
Collapse
|
14
|
Waheed AA, Swiderski M, Khan A, Gitzen A, Majadly A, Freed EO. The viral protein U (Vpu)-interacting host protein ATP6V0C down-regulates cell-surface expression of tetherin and thereby contributes to HIV-1 release. J Biol Chem 2020; 295:7327-7340. [PMID: 32291285 PMCID: PMC7247306 DOI: 10.1074/jbc.ra120.013280] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Host proteins with antiviral activity have evolved as first-line defenses to suppress viral replication. The HIV-1 accessory protein viral protein U (Vpu) enhances release of the virus from host cells by down-regulating the cell-surface expression of the host restriction factor tetherin. However, the exact mechanism of Vpu-mediated suppression of antiviral host responses is unclear. To further understand the role of host proteins in Vpu's function, here we carried out yeast two-hybrid screening and identified the V0 subunit C of vacuolar ATPase (ATP6V0C) as a Vpu-binding protein. To examine the role of ATP6V0C in Vpu-mediated tetherin degradation and HIV-1 release, we knocked down ATP6V0C expression in HeLa cells and observed that ATP6V0C depletion impairs Vpu-mediated tetherin degradation, resulting in defective HIV-1 release. We also observed that ATP6V0C overexpression stabilizes tetherin expression. This stabilization effect was specific to ATP6V0C, as overexpression of another subunit of the vacuolar ATPase, ATP6V0C″, had no effect on tetherin expression. ATP6V0C overexpression did not stabilize CD4, another target of Vpu-mediated degradation. Immunofluorescence localization experiments revealed that the ATP6V0C-stabilized tetherin is sequestered in a CD63- and lysosome-associated membrane protein 1 (LAMP1)-positive intracellular compartment. These results indicate that the Vpu-interacting protein ATP6V0C plays a role in down-regulating cell-surface expression of tetherin and thereby contributes to HIV-1 assembly and release.
Collapse
Affiliation(s)
- Abdul A Waheed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702.
| | - Maya Swiderski
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702
| | - Ali Khan
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702
| | - Ariana Gitzen
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702
| | - Ahlam Majadly
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702
| | - Eric O Freed
- Virus-Cell Interaction Section, HIV Dynamics and Replication Program, Center for Cancer Research, NCI, National Institutes of Health, Frederick, Maryland 21702
| |
Collapse
|
15
|
James CD, Morgan IM, Bristol ML. The Relationship between Estrogen-Related Signaling and Human Papillomavirus Positive Cancers. Pathogens 2020; 9:E403. [PMID: 32455952 PMCID: PMC7281727 DOI: 10.3390/pathogens9050403] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/15/2022] Open
Abstract
High risk-human papillomaviruses (HPVs) are known carcinogens. Numerous reports have linked the steroid hormone estrogen, and the expression of estrogen receptors (ERs), to HPV-related cancers, although the exact nature of the interactions remains to be fully elucidated. Here we will focus on estrogen signaling and describe both pro and potentially anti-cancer effects of this hormone in HPV-positive cancers. This review will summarize: (1) cell culture-related evidence, (2) animal model evidence, and (3) clinical evidence demonstrating an interaction between estrogen and HPV-positive cancers. This comprehensive review provides insights into the potential relationship between estrogen and HPV. We suggest that estrogen may provide a potential therapeutic for HPV-related cancers, however additional studies are necessary.
Collapse
Affiliation(s)
- Claire D. James
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
| | - Iain M. Morgan
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
- VCU Massey Cancer Center, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA
| | - Molly L. Bristol
- School of Dentistry, Philips Institute for Oral Health Research, Virginia Commonwealth University (VCU), Richmond, VA 23298, USA;
| |
Collapse
|
16
|
Basto DL, Chaves CBP, Felix SP, Amaro-Filho SM, Vieira VC, Martins LFL, de Carvalho NA, Almeida LM, Moreira MÂM. The papillomavirus E5 gene does not affect EGFR transcription and overall survival in cervical cancer. J Med Virol 2019; 92:1283-1289. [PMID: 31696949 DOI: 10.1002/jmv.25624] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Accepted: 11/04/2019] [Indexed: 01/07/2023]
Abstract
INTRODUCTION The human papillomavirus (HPV) E5 gene encodes a small and highly hydrophobic oncoprotein that affects immune evasion, cell proliferation, loss of apoptotic capacity and angiogenesis in tumors. E5 shows an affinity for biological membranes and was associated with an increase of epidermal growth factor/epidermal growth factor receptor (EGF/EGFR) signaling through the accumulation of EGFR in cellular membranes. Due to the frequent integration of the HPV genome into the host cell genome, E5 is frequently not transcribed in cervical tumors. AIM In this study we looked forward to verifying whether the potential expression of E5 protein in human papillomavirus 16 positive (HPV16+ ) and human papillomavirus 18 positive (HPV18+ ) cervical tumors was associated with levels of EGFR and vascular endothelial growth factor A (VEGFA) transcription and with patients overall survival. RESULTS Association between the presence of E5 transcripts and viral genome disruption was observed for HPV16+ and HPV18+ tumors. Association was not observed between tumors potentially capable of translating E5 and EGFR or VEGFA transcriptional levels. Similarly, the capability of translating E5 and overall survival in patients with HPV16+ squamous cell carcinoma tumors stage ≥ IB2 were not associated. CONCLUSION The likely presence of E5 transcripts was neither associated to a higher activity of the EGFR-VEGFA pathway nor to the overall survival of patients with HPV16+ squamous cell carcinoma in stages ≥ IB2.
Collapse
Affiliation(s)
- Diogo Lisbôa Basto
- Department of Genetics, Post-Graduate Program in Genetics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.,Genetics Program, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Cláudia Bessa Pereira Chaves
- Gynecologic Oncology Department and Clinical Research Division, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | - Valdimara Corrêa Vieira
- Oncovirology Program, Instituto Nacional de Câncer, Rio de Janeiro, Brazil.,Department of Immunology, Harvard Medical School, Boston, Massachusetts
| | | | | | - Liz Maria Almeida
- Population Research Program, Instituto Nacional de Câncer (INCA), Rio de Janeiro, Brazil
| | | |
Collapse
|
17
|
Suppression of Stromal Interferon Signaling by Human Papillomavirus 16. J Virol 2019; 93:JVI.00458-19. [PMID: 31292244 DOI: 10.1128/jvi.00458-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/29/2019] [Indexed: 01/09/2023] Open
Abstract
Human papillomaviruses (HPVs) infect squamous epithelia and cause several important cancers. Immune evasion is critical for viral persistence. Fibroblasts in the stromal microenvironment provide growth signals and cytokines that are required for proper epithelial differentiation, maintenance, and immune responses and are critical in the development of many cancers. In this study, we examined the role of epithelial-stromal interactions in the HPV16 life cycle using organotypic (raft) cultures as a model. Rafts were created using uninfected human foreskin keratinocytes (HFKs) and HFKs containing either wild-type HPV16 or HPV16 with a stop mutation to prevent the expression of the viral oncogene E5. Microarray analysis revealed significant changes in gene expression patterns in the stroma in response to HPV16, some of which were E5 dependent. Interferon (IFN)-stimulated genes (ISGs) and extracellular matrix remodeling genes were suppressed, the most prominent pathways affected. STAT1, IFNAR1, IRF3, and IRF7 were knocked down in stromal fibroblasts using lentiviral short hairpin RNA (shRNA) transduction. HPV late gene expression and viral copy number in the epithelium were increased when the stromal IFN pathway was disrupted, indicating that the stroma helps control the late phase of the HPV life cycle in the epithelium. Increased late gene expression correlated with increased late keratinocyte differentiation but not decreased IFN signaling in the epithelium. These studies show HPV16 has a paracrine effect on stromal innate immunity, reveal a new role for E5 as a stromal innate immune suppressor, and suggest that stromal IFN signaling may influence keratinocyte differentiation.IMPORTANCE The persistence of high-risk human papillomavirus (HPV) infections is the key risk factor for developing HPV-associated cancers. The ability of HPV to evade host immunity is a critical component of its ability to persist. The environment surrounding a tumor is increasingly understood to be critical in cancer development, including immune evasion. Our studies show that HPV can suppress the expression of immune-related genes in neighboring fibroblasts in a three-dimensional (3D) model of human epithelium. This finding is significant, because it indicates that HPV can control innate immunity not only in the infected cell but also in the microenvironment. In addition, the ability of HPV to regulate stromal gene expression depends in part on the viral oncogene E5, revealing a new function for this protein as an immune evasion factor.
Collapse
|
18
|
Young JM, Zine El Abidine A, Gómez-Martinez RA, Ozbun MA. The Known and Potential Intersections of Rab-GTPases in Human Papillomavirus Infections. Front Cell Dev Biol 2019; 7:139. [PMID: 31475144 PMCID: PMC6702953 DOI: 10.3389/fcell.2019.00139] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 07/09/2019] [Indexed: 12/16/2022] Open
Abstract
Papillomaviruses (PVs) were the first viruses recognized to cause tumors and cancers in mammalian hosts by Shope, nearly a century ago (Shope and Hurst, 1933). Over 40 years ago, zur Hausen (1976) first proposed that human papillomaviruses (HPVs) played a role in cervical cancer; in 2008, he shared the Nobel Prize in Medicine for his abundant contributions demonstrating the etiology of HPVs in genital cancers. Despite effective vaccines and screening, HPV infection and morbidity remain a significant worldwide burden, with HPV infections and HPV-related cancers expected increase through 2040. Although HPVs have long-recognized roles in tumorigenesis and cancers, our understanding of the molecular mechanisms by which these viruses interact with cells and usurp cellular processes to initiate infections and produce progeny virions is limited. This is due to longstanding challenges in both obtaining well-characterized infectious virus stocks and modeling tissue-based infection and the replicative cycles in vitro. In the last 20 years, the development of methods to produce virus-like particles (VLPs) and pseudovirions (PsV) along with more physiologically relevant cell- and tissue-based models has facilitated progress in this area. However, many questions regarding HPV infection remain difficult to address experimentally and are, thus, unanswered. Although an obligatory cellular uptake receptor has yet to be identified for any PV species, Rab-GTPases contribute to HPV uptake and transport of viral genomes toward the nucleus. Here, we provide a general overview of the current HPV infection paradigm, the epithelial differentiation-dependent HPV replicative cycle, and review the specifics of how HPVs usurp Rab-related functions during infectious entry. We also suggest other potential interactions based on how HPVs alter cellular activities to complete their replicative-cycle in differentiating epithelium. Understanding how HPVs interface with Rab functions during their complex replicative cycle may provide insight for the development of therapeutic interventions, as current viral counter-measures are solely prophylactic and therapies for HPV-positive individuals remain archaic and limited.
Collapse
Affiliation(s)
- Jesse M. Young
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Amira Zine El Abidine
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Ricardo A. Gómez-Martinez
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
- Department of Obstetrics & Gynecology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| | - Michelle A. Ozbun
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
- Department of Obstetrics & Gynecology, University of New Mexico School of Medicine, UNM Comprehensive Cancer Center, Albuquerque, NM, United States
| |
Collapse
|
19
|
Papillomaviruses and Endocytic Trafficking. Int J Mol Sci 2018; 19:ijms19092619. [PMID: 30181457 PMCID: PMC6163501 DOI: 10.3390/ijms19092619] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 08/24/2018] [Accepted: 08/29/2018] [Indexed: 12/14/2022] Open
Abstract
Endocytic trafficking plays a major role in transport of incoming human papillomavirus (HPVs) from plasma membrane to the trans Golgi network (TGN) and ultimately into the nucleus. During this infectious entry, several cellular sorting factors are recruited by the viral capsid protein L2, which plays a critical role in ensuring successful transport of the L2/viral DNA complex to the nucleus. Later in the infection cycle, two viral oncoproteins, E5 and E6, have also been shown to modulate different aspects of endocytic transport pathways. In this review, we highlight how HPV makes use of and perturbs normal endocytic transport pathways, firstly to achieve infectious virus entry, secondly to produce productive infection and the completion of the viral life cycle and, finally, on rare occasions, to bring about the development of malignancy.
Collapse
|
20
|
Wetherill LF, Wasson CW, Swinscoe G, Kealy D, Foster R, Griffin S, Macdonald A. Alkyl-imino sugars inhibit the pro-oncogenic ion channel function of human papillomavirus (HPV) E5. Antiviral Res 2018; 158:113-121. [PMID: 30096339 PMCID: PMC6156294 DOI: 10.1016/j.antiviral.2018.08.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 07/27/2018] [Accepted: 08/06/2018] [Indexed: 02/07/2023]
Abstract
Despite the availability of prophylactic vaccines the burden of human papillomavirus (HPV) associated malignancy remains high and there is a need to develop additional therapeutic strategies to complement vaccination. We have previously shown that the poorly characterised E5 oncoprotein forms a virus-coded ion channel or viroporin that was sensitive to the amantadine derivative rimantadine. We now demonstrate that alkylated imino sugars, which have antiviral activity against a number of viruses, inhibit E5 channel activity in vitro. Using molecular modelling we predict that imino sugars intercalate between E5 protomers to prevent channel oligomerisation. We explored the ability of these viroporin inhibitors to block E5-mediated activation of mitogenic signalling in keratinocytes. Treatment with either rimantadine or imino sugars prevented ERK-MAPK phosphorylation and reduced cyclin B1 expression in cells expressing E5 from a number of high-risk HPV types. Moreover, viroporin inhibitors also reduced ERK-MAPK activation and cyclin B1 expression in differentiating primary human keratinocytes containing high-risk HPV18. These observations provide evidence of a key role for E5 viroporin function during the HPV life cycle. Viroporin inhibitors could be utilised for stratified treatment of HPV associated tumours prior to virus integration, or as true antiviral therapies to eliminate virus prior to malignant transformation. Imino sugars inhibit the viroporin activity of the E5 oncoprotein. Imino sugars likely interact at E5 protomer interfaces within a channel to prevent oligomerisation. Imino sugars and adamantanes block mitogenic signalling mediated by E5 from a range of high-risk HPV types. Viroporin inhibitors reduce mitogenic signalling in differentiating primary keratinocytes containing high-risk HPV18.
Collapse
Affiliation(s)
- Laura F Wetherill
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UK; School of Medicine, Faculty of Medicine & Health, University of Leeds, Wellcome Trust Brenner Building, St James' University Hospital, Beckett St., Leeds, LS9 7TF, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Christopher W Wasson
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Gemma Swinscoe
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - David Kealy
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Richard Foster
- School of Chemistry, Faculty of Mathematics and Physical Sciences, University of Leeds, Leeds, LS2 9JT, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Stephen Griffin
- School of Medicine, Faculty of Medicine & Health, University of Leeds, Wellcome Trust Brenner Building, St James' University Hospital, Beckett St., Leeds, LS9 7TF, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
21
|
Development of Novel Single-Chain Antibodies against the Hydrophobic HPV-16 E5 Protein. BIOMED RESEARCH INTERNATIONAL 2018; 2018:5809028. [PMID: 30027096 PMCID: PMC6031085 DOI: 10.1155/2018/5809028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 05/24/2018] [Accepted: 05/29/2018] [Indexed: 11/18/2022]
Abstract
The human papilloma virus type 16 infects genital mucosa with high prevalence in the oncogenesis of cervical and oropharyngeal cancers. The E5 protein of this virus is a small hydrophobic protein, whose expression generally decreases as the infection progresses to malignancy. These characteristics point to a role of E5 in the establishment of HPV infection and the initiation into cell transformation. The study of the HPV-16 E5 functions has been hindered because of the lack of antibodies. Detection is very difficult because of its hydrophobic nature, membrane location, and very low levels of expression. Thus, the objective of this study was to select single-chain antibodies against the full size E5 protein, which was coexpressed with maltose-binding protein. We report that the E5 protein was recognized by the antibody and was validated in W12 cells by fluorescent microscopy, including a colocalization with one of its host substrates. The use of this antibody could increase our knowledge about the functions of the oncogenic HPV-16 E5 protein during the earliest stages of keratinocyte infection in human.
Collapse
|
22
|
Wechsler EI, Tugizov S, Herrera R, Da Costa M, Palefsky JM. E5 can be expressed in anal cancer and leads to epidermal growth factor receptor-induced invasion in a human papillomavirus 16-transformed anal epithelial cell line. J Gen Virol 2018; 99:631-644. [PMID: 29624161 DOI: 10.1099/jgv.0.001061] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
We created the first human papillomavirus (HPV)-16-immortalized anal epithelial cell line, known as AKC2 cells to establish an in vitro model of HPV-16-induced anal carcinogenesis. Consistent with detection of E6, E7 and E5 expression in anal cancer biopsies, AKC2 cells expressed high levels of all three HPV oncogenes. Also, similar to findings in anal cancer biopsies, epidermal growth factor receptor (EGFR) was overexpressed in AKC2 cells. AKC2 cells exhibited a poorly differentiated and invasive phenotype in three-dimensional raft culture and inhibition of EGFR function abrogated AKC2 invasion. Reducing E5 expression using E5-targeted siRNAs in AKC2 cells led to knockdown of E5 expression, but also HPV-16 E2, E6 and E7 expression. AKC2 cells treated with E5-targeted siRNA had reduced levels of total and phosphorylated EGFR, and reduced invasion. Rescue of E6/E7 expression with simultaneous E5 knockdown confirmed that E5 plays a key role in EGFR overexpression and EGFR-induced invasion.
Collapse
Affiliation(s)
- Erin Isaacson Wechsler
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Sharof Tugizov
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Rossana Herrera
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Maria Da Costa
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| | - Joel M Palefsky
- Division of Infectious Diseases, Department of Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
23
|
Wasson CW, Morgan EL, Müller M, Ross RL, Hartley M, Roberts S, Macdonald A. Human papillomavirus type 18 E5 oncogene supports cell cycle progression and impairs epithelial differentiation by modulating growth factor receptor signalling during the virus life cycle. Oncotarget 2017; 8:103581-103600. [PMID: 29262586 PMCID: PMC5732752 DOI: 10.18632/oncotarget.21658] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 08/16/2017] [Indexed: 02/04/2023] Open
Abstract
Deregulation of proliferation and differentiation-dependent signalling pathways is a hallmark of human papillomavirus (HPV) infection. Although the manipulation of these pathways by E6 and E7 has been extensively studied, controversies surround the role of the E5 oncoprotein during a productive virus life cycle. By integrating primary keratinocytes harbouring wild type or E5 knockout HPV18 genomes with pharmacological and gain/loss of function models, this study aimed to provide molecular information about the role of E5 in epithelial proliferation and differentiation. We show that E5 contributes to cell cycle progression and unscheduled host DNA synthesis in differentiating keratinocytes. E5 function correlates with increased EGFR activation in differentiating cells and blockade of this pathway impairs differentiation-dependent cell cycle progression of HPV18 containing cells. Our findings provide a functional requirement of enhanced EGFR signalling for suprabasal cellular DNA synthesis during the virus life cycle. They also reveal an unrecognised contribution of E5 towards the impaired keratinocyte differentiation observed during a productive HPV infection. E5 suppresses a signalling axis consisting of the keratinocyte growth factor receptor (KGFR) pathway. Inhibition of this pathway compensates for the loss of E5 in knockout cells and re-instates the delay in differentiation. The negative regulation of KGFR involves suppression by the EGFR pathway. Thus our data reveal an unappreciated role for E5-mediated EGFR signalling in orchestrating the balance between proliferation and differentiation in suprabasal cells.
Collapse
Affiliation(s)
- Christopher W Wasson
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Ethan L Morgan
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Marietta Müller
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Rebecca L Ross
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Margaret Hartley
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sally Roberts
- Institute of Cancer and Genomic Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Macdonald
- School of Molecular and Cellular Biology, Astbury Centre for Structural and Molecular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
24
|
Conditional cell reprogramming involves non-canonical β-catenin activation and mTOR-mediated inactivation of Akt. PLoS One 2017; 12:e0180897. [PMID: 28700668 PMCID: PMC5507294 DOI: 10.1371/journal.pone.0180897] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 06/22/2017] [Indexed: 11/19/2022] Open
Abstract
The combination of irradiated fibroblast feeder cells and Rho kinase inhibitor, Y-267362, converts primary epithelial cells growing in vitro into an undifferentiated adult stem cell-like state that is characterized by long-term proliferation. This cell culture method also maintains the proliferation of adult epithelial stem cells from various tissues. Both primary and adult stem cells retain their tissue-specific differentiation potential upon removal of the culture conditions. Due to the ability to modulate the proliferation and differentiation of the cells, this method is referred to as conditional reprogramming and it is increasingly being used in studies of tumor heterogeneity, personalized medicine and regenerative medicine. However, little is known about the biology of these conditionally reprogrammed (CR) cells. Previously we showed that β-catenin activation, a hallmark of stem cells in vivo, occurs in CR human ectocervical cells (HECs). Here we show that β-catenin-dependent transcription is necessary for the induction of epithelial stem cell markers, and that β-catenin is activated via a non-canonical pathway that is independent of Wnt and Akt/GSK-3. Active Akt actually decreases due to increased mTOR signaling, with a consequent increase in dephosphorylated, active GSK-3. Despite the increase in active GSK-3, β-catenin associates with protein phosphatase 2A (PP2A) and is activated. Inhibition of PP2A catalytic activity reduces both the level of active β-catenin and the acute induction of stem cell markers, suggesting an important role for PP2A in the activation of β-catenin. Moreover, we demonstrate similar results using human prostate and breast cells, indicating that these changes are not restricted to ectocervical epithelial cells and may represent a more fundamental property of conditional reprogramming.
Collapse
|
25
|
Brand TM, Hartmann S, Bhola NE, Peyser ND, Li H, Zeng Y, Isaacson Wechsler E, Ranall MV, Bandyopadhyay S, Duvvuri U, LaVallee TM, Jordan RCK, Johnson DE, Grandis JR. Human Papillomavirus Regulates HER3 Expression in Head and Neck Cancer: Implications for Targeted HER3 Therapy in HPV + Patients. Clin Cancer Res 2016; 23:3072-3083. [PMID: 27986750 DOI: 10.1158/1078-0432.ccr-16-2203] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Revised: 11/17/2016] [Accepted: 12/09/2016] [Indexed: 12/31/2022]
Abstract
Purpose: Human papillomavirus (HPV) 16 plays an etiologic role in a growing subset of head and neck squamous cell carcinomas (HNSCC), where viral expression of the E6 and E7 oncoproteins is necessary for tumor growth and maintenance. Although patients with HPV+ tumors have a more favorable prognosis, there are currently no HPV-selective therapies. Recent studies identified differential receptor tyrosine kinase (RTK) profiles in HPV+ versus HPV- tumors. One such RTK, HER3, is overexpressed and interacts with phosphoinositide-3-kinase (PI3K) in HPV+ tumors. Therefore, we investigated the role of HPV oncoproteins in regulating HER3-mediated signaling and determined whether HER3 could be a molecular target in HPV+ HNSCC.Experimental Design: HER3 was investigated as a molecular target in HPV+ HNSCC using established cell lines, patient-derived xenografts (PDX), and human tumor specimens. A mechanistic link between HPV and HER3 was examined by augmenting E6 and E7 expression levels in HNSCC cell lines. The dependency of HPV+ and HPV- HNSCC models on HER3 was evaluated with anti-HER3 siRNAs and the clinical stage anti-HER3 monoclonal antibody KTN3379.Results: HER3 was overexpressed in HPV+ HNSCC, where it was associated with worse overall survival in patients with pharyngeal cancer. Further investigation indicated that E6 and E7 regulated HER3 protein expression and downstream PI3K pathway signaling. Targeting HER3 with siRNAs or KTN3379 significantly inhibited the growth of HPV+ cell lines and PDXs.Conclusions: This study uncovers a direct relationship between HPV infection and HER3 in HNSCC and provides a rationale for the clinical evaluation of targeted HER3 therapy for the treatment of HPV+ patients. Clin Cancer Res; 23(12); 3072-83. ©2016 AACR.
Collapse
MESH Headings
- Animals
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/virology
- Cell Line, Tumor
- Elafin/genetics
- Gene Expression Regulation, Neoplastic/genetics
- Gene Expression Regulation, Viral/genetics
- Head and Neck Neoplasms/drug therapy
- Head and Neck Neoplasms/genetics
- Head and Neck Neoplasms/pathology
- Head and Neck Neoplasms/virology
- Human papillomavirus 16/pathogenicity
- Humans
- Mice
- Molecular Targeted Therapy
- Oncogene Proteins, Viral/genetics
- Papillomavirus E7 Proteins/genetics
- Papillomavirus Infections/genetics
- Papillomavirus Infections/virology
- RNA, Small Interfering/administration & dosage
- RNA, Small Interfering/genetics
- Receptor, ErbB-3/antagonists & inhibitors
- Receptor, ErbB-3/genetics
- Repressor Proteins/genetics
- Squamous Cell Carcinoma of Head and Neck
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Toni M Brand
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Stefan Hartmann
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
- Department of Oral and Maxillofacial Plastic Surgery, University Hospital Würzburg, Würzburg, Germany
| | - Neil E Bhola
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Noah D Peyser
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Hua Li
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Yan Zeng
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Erin Isaacson Wechsler
- Division of Infectious Diseases, Department of Medicine, University of California San Francisco, San Francisco, California
| | - Max V Ranall
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Sourav Bandyopadhyay
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California
| | - Umamaheswar Duvvuri
- Department of Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | | | - Richard C K Jordan
- Departments of Orofacial Sciences and Pathology, University of California San Francisco, San Francisco, California
| | - Daniel E Johnson
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California
| | - Jennifer R Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, California.
| |
Collapse
|
26
|
Wu W, Panté N. Vimentin plays a role in the release of the influenza A viral genome from endosomes. Virology 2016; 497:41-52. [PMID: 27423069 DOI: 10.1016/j.virol.2016.06.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/26/2016] [Accepted: 06/27/2016] [Indexed: 12/28/2022]
Abstract
Influenza A virus exploits the subcellular transport machinery during the early stages of infection. Actin filaments and microtubules facilitate the trafficking of virus-containing endosomes towards the perinuclear region; however, the role of vimentin remains to be determined. In this study, we followed influenza A virus infection in vimentin-null cells and found that vimentin depletion severely reduced influenza viral RNA and protein expression, and production of infectious progeny virions. Furthermore, we show that in vimentin-null cells endosomal distribution and acidification were affected, and incoming influenza virions accumulated in late endosomes of these cells. We propose that this accumulation resulted from the impaired acidification of late endosomes in vimentin-null cells, which blocked the release of the viral genome from these organelles. These findings are the first to demonstrate that vimentin is critical for influenza viral infection as it facilitates endosomal trafficking and acidification, and mediates viral genome penetration into the cytoplasm to propagate the infection.
Collapse
Affiliation(s)
- Wei Wu
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4
| | - Nelly Panté
- Department of Zoology, University of British Columbia, 6270 University Boulevard, Vancouver, British Columbia, Canada V6T 1Z4.
| |
Collapse
|
27
|
Gutierrez-Xicotencatl L, Salazar-Piña DA, Pedroza-Saavedra A, Chihu-Amparan L, Rodriguez-Ocampo AN, Maldonado-Gama M, Esquivel-Guadarrama FR. Humoral Immune Response Against Human Papillomavirus as Source of Biomarkers for the Prediction and Detection of Cervical Cancer. Viral Immunol 2016; 29:83-94. [PMID: 26780189 DOI: 10.1089/vim.2015.0087] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cervical cancer (CC) is one of the main causes of death among women of reproductive age. Although there are different tests, the disease tends to be diagnosed at late stages. In recent years, the use of complementary tests or sequential diagnostic tests has been implemented. Nevertheless, the results are variable and not conclusive; therefore, more studies for improving the usefulness of these tests in diagnostics are necessary. The human papillomavirus (HPV) infection has been associated with both benign and malignant proliferation of skin and mucosal tissues. Furthermore, some HPV types have been classified as high risk due to their potential to cause cancer, and HPV16 is most frequently associated with this disease. Although between 70% and 80% of precancerous lesions are eliminated by the host's immune system, there is no available test to distinguish between regressive lesions from those that could progress to CC. An HPV infection generates a humoral immune response against L1 and L2 capsid proteins, which can be protective and a response against early proteins. The latter is not a protective response, but these antibodies can be used as markers to determine the stage of the infection and/or the stage of the cervical lesion. Up to now, the humoral immune response resulting from the HPV infection has been used to study the biology of the virus and the efficacy of the HPV vaccines. Although there are no conclusive results regarding the use of these antibodies for diagnosis, we hereby review the actual panorama of the antibody response against the HPV proteins during the development of the disease as well as their possible use as biomarkers for the progression of cervical lesions and of CC.
Collapse
Affiliation(s)
- Lourdes Gutierrez-Xicotencatl
- 1 Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos, México
| | - Dolores Azucena Salazar-Piña
- 1 Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos, México
| | - Adolfo Pedroza-Saavedra
- 1 Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos, México
| | - Lilia Chihu-Amparan
- 1 Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos, México
| | | | - Minerva Maldonado-Gama
- 1 Centro de Investigación Sobre Enfermedades Infecciosas, Instituto Nacional de Salud Pública , Cuernavaca, Morelos, México
| | | |
Collapse
|
28
|
Doorbar J, Egawa N, Griffin H, Kranjec C, Murakami I. Human papillomavirus molecular biology and disease association. Rev Med Virol 2015; 25 Suppl 1:2-23. [PMID: 25752814 PMCID: PMC5024016 DOI: 10.1002/rmv.1822] [Citation(s) in RCA: 544] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 06/12/2014] [Accepted: 06/25/2014] [Indexed: 12/27/2022]
Abstract
Human papillomaviruses (HPVs) have evolved over millions of years to propagate themselves in a range of different animal species including humans. Viruses that have co‐evolved slowly in this way typically cause chronic inapparent infections, with virion production in the absence of apparent disease. This is the case for many Beta and Gamma HPV types. The Alpha papillomavirus types have however evolved immunoevasion strategies that allow them to cause persistent visible papillomas. These viruses activate the cell cycle as the infected epithelial cell differentiates in order to create a replication competent environment that allows viral genome amplification and packaging into infectious particles. This is mediated by the viral E6, E7, and E5 proteins. High‐risk E6 and E7 proteins differ from their low‐risk counterparts however in being able to drive cell cycle entry in the upper epithelial layers and also to stimulate cell proliferation in the basal and parabasal layers. Deregulated expression of these cell cycle regulators underlies neoplasia and the eventual progression to cancer in individuals who cannot resolve high‐risk HPV infection. Most work to date has focused on the study of high‐risk HPV types such as HPV 16 and 18, which has led to an understanding of the molecular pathways subverted by these viruses. Such approaches will lead to the development of better strategies for disease treatment, including targeted antivirals and immunotherapeutics. Priorities are now focused toward understanding HPV neoplasias at sites other than the cervix (e.g. tonsils, other transformation zones) and toward understanding the mechanisms by which low‐risk HPV types can sometimes give rise to papillomatosis and under certain situations even cancers. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| | | | | | | | | |
Collapse
|
29
|
Müller M, Wasson CW, Bhatia R, Boxall S, Millan D, Goh GYS, Haas J, Stonehouse NJ, Macdonald A. YIP1 family member 4 (YIPF4) is a novel cellular binding partner of the papillomavirus E5 proteins. Sci Rep 2015; 5:12523. [PMID: 26235900 PMCID: PMC4522686 DOI: 10.1038/srep12523] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/26/2015] [Indexed: 01/14/2023] Open
Abstract
E5 proteins are amongst the least understood of the Human Papillomavirus (HPV) encoded gene products. They are small, membrane-integrated proteins known to modulate a number of critical host pathways associated with pathogenesis including growth factor receptor signaling and immune evasion. Their role in the virus life cycle is less clear, indicating a role in the productive stages of the life cycle. However, a mechanism for this is currently lacking. Here we describe the identification of a novel binding partner of E5, YIPF4 using yeast two-hybrid analysis. YIPF4 is also a poorly characterized membrane spanning protein. Mutagenesis studies implicated the transmembrane regions of each protein as important for their interaction. Binding to YIPF4 was found for all E5 proteins tested suggesting that this interaction may mediate a conserved E5 function. In normal human keratinocytes YIPF4 expression was down-regulated upon differentiation and this reduction was partially rescued in cells harbouring HPV. Despite the conserved nature of the interaction with E5, siRNA mediated depletion of YIPF4 failed to impede two well-characterized functions of E5, namely EGFR trafficking or HLA class I presentation. Continued studies of YIPF4 are warranted to determine its role in the PV life cycle.
Collapse
Affiliation(s)
| | | | - Ramya Bhatia
- Division of Pathway Medicine, The University of Edinburgh, UK
| | | | - David Millan
- Department of Pathology, Southern General Hospital, Glasgow, Scotland, UK
| | | | - Jürgen Haas
- Division of Pathway Medicine, The University of Edinburgh, UK
| | | | | |
Collapse
|
30
|
Zhang J, Saba NF, Chen GZ, Shin DM. Targeting HER (ERBB) signaling in head and neck cancer: An essential update. Mol Aspects Med 2015; 45:74-86. [PMID: 26163475 DOI: 10.1016/j.mam.2015.07.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 12/13/2022]
Abstract
HNC (head and neck cancer) remains the 6th most common carcinoma worldwide. The suboptimal survival and toxicities observed with conventional approaches warrant exploration of novel therapeutic strategies such as targeted therapies. Although targeting EGFR (epidermal growth factor receptor) with cetuximab demonstrated clinical promise, HER (human epidermal growth factor receptor) or ERBB (erythroblastic leukemia viral oncogene homolog) targeted therapy in HNC has overall been suboptimal to date in clinical settings. Overcoming the resistance as well as identifying new strategies therefore remains a significant challenge. In this review, we will discuss the emerging roles of HER members besides EGFR. A comprehensive "three-dimensional" view of HER signaling pathway from the importance of EGFR nuclear translocation to our maturing concept of receptors' "spatial regulation", as well as the interdependence and interaction among different HER members will also be addressed to complete an essential update of HER signaling in HNC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, NE, Atlanta, GA 30322, USA; Department of Internal Medicine, Division of Hematology, Oncology and Blood & Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Nabil F Saba
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, NE, Atlanta, GA 30322, USA
| | - Georgia Zhuo Chen
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, NE, Atlanta, GA 30322, USA
| | - Dong M Shin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, 1365-C Clifton Road, NE, Atlanta, GA 30322, USA.
| |
Collapse
|
31
|
Scott C, Griffin S. Viroporins: structure, function and potential as antiviral targets. J Gen Virol 2015; 96:2000-2027. [PMID: 26023149 DOI: 10.1099/vir.0.000201] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
The channel-forming activity of a family of small, hydrophobic integral membrane proteins termed 'viroporins' is essential to the life cycles of an increasingly diverse range of RNA and DNA viruses, generating significant interest in targeting these proteins for antiviral development. Viroporins vary greatly in terms of their atomic structure and can perform multiple functions during the virus life cycle, including those distinct from their role as oligomeric membrane channels. Recent progress has seen an explosion in both the identification and understanding of many such proteins encoded by highly significant pathogens, yet the prototypic M2 proton channel of influenza A virus remains the only example of a viroporin with provenance as an antiviral drug target. This review attempts to summarize our current understanding of the channel-forming functions for key members of this growing family, including recent progress in structural studies and drug discovery research, as well as novel insights into the life cycles of many viruses revealed by a requirement for viroporin activity. Ultimately, given the successes of drugs targeting ion channels in other areas of medicine, unlocking the therapeutic potential of viroporins represents a valuable goal for many of the most significant viral challenges to human and animal health.
Collapse
Affiliation(s)
- Claire Scott
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| | - Stephen Griffin
- Leeds Institute of Cancer & Pathology and Leeds CRUK Clinical Centre, Faculty of Medicine and Health, St James's University Hospital, University of Leeds, Beckett Street, Leeds LS9 7TF, UK
| |
Collapse
|
32
|
Kondapalli KC, Llongueras JP, Capilla-González V, Prasad H, Hack A, Smith C, Guerrero-Cázares H, Quiñones-Hinojosa A, Rao R. A leak pathway for luminal protons in endosomes drives oncogenic signalling in glioblastoma. Nat Commun 2015; 6:6289. [PMID: 25662504 PMCID: PMC4354686 DOI: 10.1038/ncomms7289] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 01/12/2015] [Indexed: 12/15/2022] Open
Abstract
Epidermal growth factor receptor (EGFR) signalling is a potent driver of glioblastoma, a malignant and lethal form of brain cancer. Disappointingly, inhibitors targeting receptor tyrosine kinase activity are not clinically effective and EGFR persists on the plasma membrane to maintain tumour growth and invasiveness. Here we show that endolysosomal pH is critical for receptor sorting and turnover. By functioning as a leak pathway for protons, the Na(+)/H(+) exchanger NHE9 limits luminal acidification to circumvent EGFR turnover and prolong downstream signalling pathways that drive tumour growth and migration. In glioblastoma, NHE9 expression is associated with stem/progenitor characteristics, radiochemoresistance, poor prognosis and invasive growth in vitro and in vivo. Silencing or inhibition of NHE9 in brain tumour-initiating cells attenuates tumoursphere formation and improves efficacy of EGFR inhibitor. Thus, NHE9 mediates inside-out control of oncogenic signalling and is a highly druggable target for pan-specific receptor clearance in cancer therapy.
Collapse
Affiliation(s)
- Kalyan C. Kondapalli
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Jose P. Llongueras
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Vivian Capilla-González
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hari Prasad
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Anniesha Hack
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Christopher Smith
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Hugo Guerrero-Cázares
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| | - Rajini Rao
- Department of Physiology, The Johns Hopkins University School of Medicine, 725 North Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
33
|
Müller M, Prescott EL, Wasson CW, Macdonald A. Human papillomavirus E5 oncoprotein: function and potential target for antiviral therapeutics. Future Virol 2015. [DOI: 10.2217/fvl.14.99] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
ABSTRACT Mucosal human papillomaviruses express a small, hydrophobic, protein called E5, which plays an important role in the HPV life cycle by delaying normal epithelial cell differentiation while maintaining cell cycle progression. In addition, E5 exhibits transforming abilities in a number of cell culture systems and transgenic mouse models. Lacking any described enzymatic activity, E5 is thought to function by binding to host proteins and modulating their activities. In particular, members of the growth factor receptor family are known targets for subversion. This review article summarizes our latest understanding of this enigmatic oncoprotein, including its role in the HPV life cycle, interactions with host proteins and contribution toward tumorigenesis.
Collapse
Affiliation(s)
- Marietta Müller
- School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Emma L Prescott
- School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Christopher W Wasson
- School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| | - Andrew Macdonald
- School of Molecular & Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, West Yorkshire, LS2 9JT, UK
| |
Collapse
|
34
|
Ajiro M, Zheng ZM. Oncogenes and RNA splicing of human tumor viruses. Emerg Microbes Infect 2014; 3:e63. [PMID: 26038756 PMCID: PMC4185361 DOI: 10.1038/emi.2014.62] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2014] [Revised: 06/29/2014] [Accepted: 06/29/2014] [Indexed: 02/07/2023]
Abstract
Approximately 10.8% of human cancers are associated with infection by an oncogenic virus. These viruses include human papillomavirus (HPV), Epstein–Barr virus (EBV), Merkel cell polyomavirus (MCV), human T-cell leukemia virus 1 (HTLV-1), Kaposi's sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV) and hepatitis B virus (HBV). These oncogenic viruses, with the exception of HCV, require the host RNA splicing machinery in order to exercise their oncogenic activities, a strategy that allows the viruses to efficiently export and stabilize viral RNA and to produce spliced RNA isoforms from a bicistronic or polycistronic RNA transcript for efficient protein translation. Infection with a tumor virus affects the expression of host genes, including host RNA splicing factors, which play a key role in regulating viral RNA splicing of oncogene transcripts. A current prospective focus is to explore how alternative RNA splicing and the expression of viral oncogenes take place in a cell- or tissue-specific manner in virus-induced human carcinogenesis.
Collapse
Affiliation(s)
- Masahiko Ajiro
- Tumor Virus RNA Biology Section, Gene Regulation and Chromosome Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, MD 21702, USA
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, Gene Regulation and Chromosome Biology Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health , Frederick, MD 21702, USA
| |
Collapse
|
35
|
Um SH, Mundi N, Yoo J, Palma DA, Fung K, MacNeil D, Wehrli B, Mymryk JS, Barrett JW, Nichols AC. Variable expression of the forgotten oncogene E5 in HPV-positive oropharyngeal cancer. J Clin Virol 2014; 61:94-100. [PMID: 25027574 DOI: 10.1016/j.jcv.2014.06.019] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/19/2014] [Accepted: 06/20/2014] [Indexed: 01/25/2023]
Abstract
BACKGROUND The role of the HPV E6/E7 oncogenes in head and neck squamous cell cancer (HNSCC) has been studied extensively, but the role of the viral E5 protein remains poorly understood. Studies in cervical cancer indicate that E5 increases epidermal growth factor receptor (EGFR) recycling to the cell surface and enhances growth factor signal transduction. OBJECTIVE This study was designed to examine the relationship between HPV E5, EGFR, and survival in HPV-positive HNSCC. STUDY DESIGN A retrospective search of the London Health Sciences Centre pathology database was performed to identify oropharyngeal cancer samples. HPV E5 and EGFR expression was measured by reverse transcriptase real-time PCR. RESULTS The majority of oropharyngeal tumor samples (59/82, 72%) were HPV-16 positive. Among the HPV-positive tumors, highly variable E5 expression was detected from early polycistronic transcripts. Tumors with high E5 expression levels had significantly higher EGFR levels (p=0.03). High E5 levels were correlated with improved recurrence-free survival, but not overall survival (p=0.02 and 0.71, respectively), whereas high EGFR was strongly associated with decreased recurrence-free and overall survival (p<0.001 and 0.006 respectively). Multivariate analysis revealed E5 and EGFR to be the strongest predictors of recurrence-free survival (p<0.01). CONCLUSIONS HPV E5-encoded transcripts are variably expressed in HPV-positive HNSCC and this is correlated with EGFR expression in HPV-positive OPC. However, E5 and EGFR independently predict recurrence-free survival in opposing manners. These findings require further validation to determine if E5 and EGFR are useful biomarkers to stratify treatment intensity for patients with HPV-positive HNSCC.
Collapse
Affiliation(s)
- Sung Ho Um
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada
| | - Neil Mundi
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada
| | - John Yoo
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada
| | - David A Palma
- London Regional Cancer Program, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada
| | - Kevin Fung
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada
| | - Danielle MacNeil
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada
| | - Bret Wehrli
- Department of Pathology, The University of Western Ontario, London, Ontario, Canada
| | - Joe S Mymryk
- London Regional Cancer Program, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada; Department of Microbiology and Immunology, The University of Western Ontario, London, Ontario, Canada
| | - John W Barrett
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada
| | - Anthony C Nichols
- Department of Otolaryngology Head & Neck Surgery, The University of Western Ontario, London, Ontario, Canada; London Regional Cancer Program, London, Ontario, Canada; Lawson Health Research Institute, London, Ontario, Canada; Department of Oncology, The University of Western Ontario, London, Ontario, Canada; Department of Pathology, The University of Western Ontario, London, Ontario, Canada.
| |
Collapse
|
36
|
Kotnik Halavaty K, Regan J, Mehta K, Laimins L. Human papillomavirus E5 oncoproteins bind the A4 endoplasmic reticulum protein to regulate proliferative ability upon differentiation. Virology 2014; 452-453:223-30. [PMID: 24606699 DOI: 10.1016/j.virol.2014.01.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 11/16/2013] [Accepted: 01/17/2014] [Indexed: 10/25/2022]
Abstract
Human papillomaviruses (HPV) infect stratified epithelia and link their life cycles to epithelial differentiation. The HPV E5 protein plays a role in the productive phase of the HPV life cycle but its mechanism of action is still unclear. We identify a new binding partner of E5, A4, using a membrane-associated yeast-two hybrid system. The A4 protein co-localizes with HPV 31 E5 in perinuclear regions and forms complexes with E5 and Bap31. In normal keratinocytes, A4 is found primarily in basal cells while in HPV positive cells high levels of A4 are seen in both undifferentiated and differentiated cells. Reduction of A4 expression by shRNAs, enhanced HPV genome amplification and increased cell proliferation ability following differentiation but this was not seen in cells lacking E5. Our studies suggest that the A4 protein is an important E5 binding partner that plays a role in regulating cell proliferation ability upon differentiation.
Collapse
Affiliation(s)
- Katarina Kotnik Halavaty
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Jennifer Regan
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Kavi Mehta
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA
| | - Laimonis Laimins
- Department of Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, 303 E. Chicago Avenue, Chicago, IL 60611, USA.
| |
Collapse
|
37
|
Dolman NJ, Kilgore JA, Davidson MW. A review of reagents for fluorescence microscopy of cellular compartments and structures, part I: BacMam labeling and reagents for vesicular structures. ACTA ACUST UNITED AC 2014; Chapter 12:12.30.1-12.30.27. [PMID: 23835803 DOI: 10.1002/0471142956.cy1230s65] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Fluorescent labeling of vesicular structures in cultured cells, particularly for live cells, can be challenging for a number of reasons. The first challenge is to identify a reagent that will be specific enough where some structures have a number of potential reagents and others very few options. The emergence of BacMam constructs has allowed more easy-to-use choices. Presented here is a discussion of BacMam constructs as well as a review of commercially-available reagents for labeling vesicular structures in cells, including endosomes, peroxisomes, lysosomes, and autophagosomes, complete with a featured reagent for each structure, recommended protocol, troubleshooting guide, and example image.
Collapse
Affiliation(s)
- Nick J Dolman
- Molecular Probes Labeling and Detection, Life Technologies, Eugene, OR, USA
| | | | | |
Collapse
|
38
|
Chow LT, Broker TR. Human papillomavirus infections: warts or cancer? Cold Spring Harb Perspect Biol 2013; 5:cshperspect.a012997. [PMID: 23685995 DOI: 10.1101/cshperspect.a012997] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Human papillomaviruses (HPVs) are prevalent pathogens of mucosal and cutaneous epithelia. Productive infections of squamous epithelia lead to benign hyperproliferative warts, condylomata, or papillomas. Persistent infections of the anogenital mucosa by high-risk HPV genotypes 16 and 18 and closely related types can infrequently progress to high-grade intraepithelial neoplasias, carcinomas-in-situ, and invasive cancers in women and men. HPV-16 is also associated with a fraction of head and neck cancers. We discuss the interactions of the mucosotropic HPVs with the host regulatory proteins and pathways that lead to benign coexistence and enable HPV DNA amplification or, alternatively, to cancers that no longer support viral production.
Collapse
Affiliation(s)
- Louise T Chow
- Department of Biochemistry and Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294-0005, USA
| | | |
Collapse
|
39
|
DiMaio D, Petti LM. The E5 proteins. Virology 2013; 445:99-114. [PMID: 23731971 DOI: 10.1016/j.virol.2013.05.006] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 05/01/2013] [Accepted: 05/03/2013] [Indexed: 12/23/2022]
Abstract
The E5 proteins are short transmembrane proteins encoded by many animal and human papillomaviruses. These proteins display transforming activity in cultured cells and animals, and they presumably also play a role in the productive virus life cycle. The E5 proteins are thought to act by modulating the activity of cellular proteins. Here, we describe the biological activities of the best-studied E5 proteins and discuss the evidence implicating specific protein targets and pathways in mediating these activities. The primary target of the 44-amino acid BPV1 E5 protein is the PDGF β receptor, whereas the EGF receptor appears to be an important target of the 83-amino acid HPV16 E5 protein. Both E5 proteins also bind to the vacuolar ATPase and affect MHC class I expression and cell-cell communication. Continued studies of the E5 proteins will elucidate important aspects of transmembrane protein-protein interactions, cellular signal transduction, cell biology, virus replication, and tumorigenesis.
Collapse
Affiliation(s)
- Daniel DiMaio
- Department of Genetics, Yale School of Medicine, USA; Department of Therapeutic Radiology, Yale School of Medicine, USA; Department of Molecular Biophysics & Biochemistry, Yale University, USA; Yale Cancer Center, USA.
| | | |
Collapse
|
40
|
HPV-18 E6 mutants reveal p53 modulation of viral DNA amplification in organotypic cultures. Proc Natl Acad Sci U S A 2013; 110:7542-9. [PMID: 23572574 DOI: 10.1073/pnas.1304855110] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human papillomaviruses (HPVs) amplify in differentiated strata of a squamous epithelium. The HPV E7 protein destabilizes the p130/retinoblastoma susceptibility protein family of tumor suppressors and reactivates S-phase reentry, thereby facilitating viral DNA amplification. The high-risk HPV E6 protein destabilizes the p53 tumor suppressor and many other host proteins. However, the critical E6 targets relevant to viral DNA amplification have not been identified, because functionally significant E6 mutants are not stably maintained in transfected cells. Using Cre-loxP recombination, which efficiently generates HPV genomic plasmids in transfected primary human keratinocytes, we have recapitulated a highly productive infection of HPV-18 in organotypic epithelial cultures. By using this system, we now report the characterization of four HPV-18 E6 mutations. An E6 null mutant accumulated high levels of p53 and amplified very poorly. p53 siRNA or ectopic WT E6 partially restored amplification, whereas three missense E6 mutations that did not effectively destabilize p53 complemented the null mutant poorly. Unexpectedly, in cis, two of the missense mutants amplified, albeit to a lower extent than the WT and only in cells with undetectable p53. These observations and others implicate p53 and additional host proteins in regulating viral DNA amplification and also suggest an inhibitory effect of E6 overexpression. We show that high levels of viral DNA amplification are critical for late protein expression and report several previously undescribed viral RNAs, including bicistronic transcripts predicted to encode E5 and L2 or an alternative form of E1^E4 and L1.
Collapse
|
41
|
Yuan CH, Filippova M, Duerksen-Hughes P. Modulation of apoptotic pathways by human papillomaviruses (HPV): mechanisms and implications for therapy. Viruses 2012; 4:3831-50. [PMID: 23250450 PMCID: PMC3528293 DOI: 10.3390/v4123831] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 12/12/2012] [Accepted: 12/14/2012] [Indexed: 12/14/2022] Open
Abstract
The ability of the host to trigger apoptosis in infected cells is perhaps the most powerful tool by which viruses can be cleared from the host organism. To avoid elimination by this mechanism, human papillomaviruses (HPV) have developed several mechanisms that enable the cells they infect to elude both extrinsic and intrinsic apoptosis. In this manuscript, we review the current literature regarding how HPV-infected cells avoid apoptosis and the molecular mechanisms involved in these events. In particular, we will discuss the modifications in intrinsic and extrinsic apoptotic pathways caused by proteins encoded by HPV early genes. Many of the current efforts regarding anti-cancer drug development are focused on directing tumor cells to undergo apoptosis. However, the ability of HPV-infected cells to resist apoptotic signals renders such therapies ineffective. Possible mechanisms for overcoming the resistance of HPV-infected tumor cells to anticancer drugs will be discussed.
Collapse
Affiliation(s)
- Chung-Hsiang Yuan
- Department of Basic Sciences, Loma Linda University School of Medicine, 11085 Campus St., Loma Linda, CA 92354, USA.
| | | | | |
Collapse
|
42
|
Doorbar J, Quint W, Banks L, Bravo IG, Stoler M, Broker TR, Stanley MA. The biology and life-cycle of human papillomaviruses. Vaccine 2012; 30 Suppl 5:F55-70. [PMID: 23199966 DOI: 10.1016/j.vaccine.2012.06.083] [Citation(s) in RCA: 913] [Impact Index Per Article: 70.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 06/04/2012] [Accepted: 06/05/2012] [Indexed: 12/20/2022]
Abstract
Human papillomaviruses (HPVs) comprise a diverse group, and have different epithelial tropisms and life-cycle strategies. Many HPVs are classified as low-risk, as they are only very rarely associated with neoplasia or cancer in the general population. These HPVs typically cause inapparent/inconspicuous infections, or benign papillomas, which can persist for months or years, but which are eventually resolved by the host's immune system. Low-risk HPVs are difficult to manage in immunosuppressed people and in individuals with genetic predispositions, and can give rise to papillomatosis, and in rare instances, to cancer. The high-risk HPV types are, by contrast, a cause of several important human cancers, including almost all cases of cervical cancer, a large proportion of other anogenital cancers and a growing number of head and neck tumours. The high-risk HPV types constitute a subset of the genus Alphapapillomavirus that are prevalent in the general population, and in most individuals cause only inconspicuous oral and genital lesions. Cancer progression is associated with persistent high-risk HPV infection and with deregulated viral gene expression, which leads to excessive cell proliferation, deficient DNA repair, and the accumulation of genetic damage in the infected cell. Although their life-cycle organisation is broadly similar to that of the low-risk HPV types, the two groups differ significantly in their capacity to drive cell cycle entry and cell proliferation in the basal/parabasal cell layers. This is thought to be linked, at least in part, to different abilities of the high- and low-risk E6 proteins to modulate the activity of p53 and PDZ-domain proteins, and the differential ability of the E7 proteins to target the several different members of the retinoblastoma protein family. This article forms part of a special supplement entitled "Comprehensive Control of HPV Infections and Related Diseases" Vaccine Volume 30, Supplement 5, 2012.
Collapse
Affiliation(s)
- John Doorbar
- Division of Virology, National Institute for Medical Research, London, United Kingdom.
| | | | | | | | | | | | | |
Collapse
|
43
|
Conditionally reprogrammed cells represent a stem-like state of adult epithelial cells. Proc Natl Acad Sci U S A 2012; 109:20035-40. [PMID: 23169653 DOI: 10.1073/pnas.1213241109] [Citation(s) in RCA: 224] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The combination of irradiated fibroblast feeder cells and Rho kinase inhibitor, Y-27632, conditionally induces an indefinite proliferative state in primary mammalian epithelial cells. These conditionally reprogrammed cells (CRCs) are karyotype-stable and nontumorigenic. Because self-renewal is a recognized property of stem cells, we investigated whether Y-27632 and feeder cells induced a stem-like phenotype. We found that CRCs share characteristics of adult stem cells and exhibit up-regulated expression of α6 and β1 integrins, ΔNp63α, CD44, and telomerase reverse transcriptase, as well as decreased Notch signaling and an increased level of nuclear β-catenin. The induction of CRCs is rapid (occurs within 2 d) and results from reprogramming of the entire cell population rather than the selection of a minor subpopulation. CRCs do not overexpress the transcription factor sets characteristic of embryonic or induced pluripotent stem cells (e.g., Sox2, Oct4, Nanog, or Klf4). The induction of CRCs is also reversible, and removal of Y-27632 and feeders allows the cells to differentiate normally. Thus, when CRCs from ectocervical epithelium or tracheal epithelium are placed in an air-liquid interface culture system, the cervical cells form a well differentiated stratified squamous epithelium, whereas the tracheal cells form a ciliated airway epithelium. We discuss the diagnostic and therapeutic opportunities afforded by a method that can generate adult stem-like cells in vitro without genetic manipulation.
Collapse
|
44
|
Ajiro M, Jia R, Zhang L, Liu X, Zheng ZM. Intron definition and a branch site adenosine at nt 385 control RNA splicing of HPV16 E6*I and E7 expression. PLoS One 2012; 7:e46412. [PMID: 23056301 PMCID: PMC3464268 DOI: 10.1371/journal.pone.0046412] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Accepted: 08/29/2012] [Indexed: 11/19/2022] Open
Abstract
HPV16 E6 and E7, two viral oncogenes, are expressed from a single bicistronic pre-mRNA. In this report, we provide the evidence that the bicistronic pre-mRNA intron 1 contains three 5' splice sites (5' ss) and three 3' splice sites (3' ss) normally used in HPV16(+) cervical cancer and its derived cell lines. The choice of two novel alternative 5' ss (nt 221 5' ss and nt 191 5' ss) produces two novel isoforms of E6E7 mRNAs (E6*V and E6*VI). The nt 226 5' ss and nt 409 3' ss is preferentially selected over the other splice sites crossing over the intron to excise a minimal length of the intron in RNA splicing. We identified AACAAAC as the preferred branch point sequence (BPS) and an adenosine at nt 385 (underlined) in the BPS as a branch site to dictate the selection of the nt 409 3' ss for E6*I splicing and E7 expression. Introduction of point mutations into the mapped BPS led to reduced U2 binding to the BPS and thereby inhibition of the second step of E6E7 splicing at the nt 409 3' ss. Importantly, the E6E7 bicistronic RNA with a mutant BPS and inefficient splicing makes little or no E7 and the resulted E6 with mutations of (91)QYNK(94) to (91)PSFW(94) displays attenuate activity on p53 degradation. Together, our data provide structural basis of the E6E7 intron 1 for better understanding of how viral E6 and E7 expression is regulated by alternative RNA splicing. This study elucidates for the first time a mapped branch point in HPV16 genome involved in viral oncogene expression.
Collapse
Affiliation(s)
- Masahiko Ajiro
- Tumor Virus RNA Biology Section, HIV and AIDS Malignancy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | - Rong Jia
- Tumor Virus RNA Biology Section, HIV and AIDS Malignancy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | - Lifang Zhang
- Tumor Virus RNA Biology Section, HIV and AIDS Malignancy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | - Xuefeng Liu
- Tumor Virus RNA Biology Section, HIV and AIDS Malignancy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
| | - Zhi-Ming Zheng
- Tumor Virus RNA Biology Section, HIV and AIDS Malignancy Branch, Center for Cancer Research, NCI, NIH, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
45
|
Sahab Z, Sudarshan SR, Liu X, Zhang Y, Kirilyuk A, Kamonjoh CM, Simic V, Dai Y, Byers SW, Doorbar J, Suprynowicz FA, Schlegel R. Quantitative measurement of human papillomavirus type 16 e5 oncoprotein levels in epithelial cell lines by mass spectrometry. J Virol 2012; 86:9465-73. [PMID: 22740411 PMCID: PMC3416135 DOI: 10.1128/jvi.01032-12] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Accepted: 06/19/2012] [Indexed: 11/20/2022] Open
Abstract
The high-risk human papillomavirus type 16 (HPV-16) E5 protein (16E5) induces tumors in a transgenic mouse model and may contribute to early stages of cervical carcinogenesis. Although high-risk E5 expression is generally thought to be lost during the progression to cervical carcinoma following integration of HPV DNA into the host genome, episomal viral DNA has been documented in a subset of HPV-16-positive malignant lesions. Numerous studies have shown that transcripts that could potentially encode 16E5 are present in cervical biopsy specimens and cervical cancer cell lines, but the presence of E5 protein has been demonstrated in only two reports that have not been corroborated. In the present study, we show that trypsin cleavage of 16E5 generates a unique four-amino-acid C-terminal peptide (FLIT) that serves as a marker for E5 expression in transfected cells and epithelial cell lines containing integrated and episomal HPV-16 DNA. Following trypsin cleavage, reversed-phase chromatography and mass spectrometry (MS) were used to detect FLIT. Immunoprecipitation assays using a newly generated anti-16E5 antibody confirmed that 16E5 was solely responsible for the FLIT signal, and deuterated FLIT peptide provided an internal standard that enabled us to quantify the number of 16E5 molecules per cell. We show that 16E5 is expressed in the Caski but not in the SiHa cervical cancer cell line, suggesting that 16E5 may contribute to the malignant phenotype of some cervical cancers, even in cells exclusively containing an integrated HPV genome.
Collapse
Affiliation(s)
- Ziad Sahab
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, DC, USA
| | - Sawali R. Sudarshan
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - YiYu Zhang
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - Alexander Kirilyuk
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, DC, USA
| | | | - Vera Simic
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - Yuhai Dai
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - Stephen W. Byers
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University Medical School, Washington, DC, USA
| | - John Doorbar
- Division of Virology, National Institute for Medical Research, London, United Kingdom
| | - Frank A. Suprynowicz
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| | - Richard Schlegel
- Department of Pathology, Georgetown University Medical School, Washington, DC, USA
| |
Collapse
|
46
|
Kivi N, Rönty M, Tarkkanen J, Auvinen P, Auvinen E. Cell culture model predicts human disease: Altered expression of junction proteins and matrix metalloproteinases in cervical dysplasia. BMC Clin Pathol 2012; 12:9. [PMID: 22863036 PMCID: PMC3495715 DOI: 10.1186/1472-6890-12-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2012] [Accepted: 07/30/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cervical cancer is necessarily caused by human papillomaviruses, which encode three oncogenes manifesting their functions by interfering with a number of cellular proteins and pathways: the E5, E6, and E7 proteins. We have earlier found in our microarray studies that the E5 oncogene crucially affects the expression of cellular genes involved in adhesion and motility of epithelial cells. METHODS In order to biologically validate our previous experimental findings we performed immunohistochemical staining of a representative set of tissue samples from different grades of high-risk human papillomavirus associated cervical disease as well as normal squamous and columnar cervical epithelium. Three-dimensional collagen raft cultures established from E5-expressing and control epithelial cells were also examined. The expression of p16, matrix metalloproteinase (MMP) -7, MMP-16, cytokeratin (CK) 8/18, laminin, E-cadherin and beta-catenin was studied. RESULTS In agreement with our previous microarray studies, we found intense staining for E-cadherin and beta-catenin in adherens junctions even in high-grade cervical lesions. Staining for MMP-16 was increased in severe disease as well. No significant change in staining for MMP-7 and cytokeratin 8/18 along with the grade of cervical squamous epithelial disease was observed. CONCLUSIONS Here we have confirmed, using tissue material from human papillomavirus associated lesions, some of the cellular gene expression modifications that we earlier reported in an experimental system studying specifically the E5 oncogene of papillomaviruses. These findings were partially surprising in the context of cervical carcinogenesis and emphasize that the complexity of carcinogenesis is not yet fully understood. Microarray approaches provide a wide overwiev of gene expression in experimental settings, which may yield biologically valid biomarkers for disease diagnostics, prognosis, and follow-up.
Collapse
Affiliation(s)
- Niina Kivi
- Haartman Institute, Department of Virology, University of Helsinki, POB 21 (Haartmaninkatu 3), FIN-00014, Helsinki, Finland
| | | | | | | | | |
Collapse
|
47
|
Wetherill LF, Holmes KK, Verow M, Müller M, Howell G, Harris M, Fishwick C, Stonehouse N, Foster R, Blair GE, Griffin S, Macdonald A. High-risk human papillomavirus E5 oncoprotein displays channel-forming activity sensitive to small-molecule inhibitors. J Virol 2012; 86:5341-51. [PMID: 22357280 PMCID: PMC3347351 DOI: 10.1128/jvi.06243-11] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 02/15/2012] [Indexed: 01/12/2023] Open
Abstract
High-risk human papillomavirus type 16 (HPV16) is the primary causative agent of cervical cancer and therefore is responsible for significant morbidity and mortality worldwide. Cellular transformation is mediated directly by the expression of viral oncogenes, the least characterized of which, E5, subverts cellular proliferation and immune recognition processes. Despite a growing catalogue of E5-specific host interactions, little is understood regarding the molecular basis of its function. Here we describe a novel function for HPV16 E5 as an oligomeric channel-forming protein, placing it within the virus-encoded "viroporin" family. The development of a novel recombinant E5 expression system showed that E5 formed oligomeric assemblies of a defined luminal diameter and stoichiometry in membranous environments and that such channels mediated fluorescent dye release from liposomes. Hexameric E5 channel stoichiometry was suggested by native PAGE studies. In lieu of high-resolution structural information, established de novo molecular modeling and design methods permitted the development of the first specific small-molecule E5 inhibitor, capable of both abrogating channel activity in vitro and reducing E5-mediated effects on cell signaling pathways. The identification of channel activity should enhance the future understanding of the physiological function of E5 and could represent an important target for antiviral intervention.
Collapse
Affiliation(s)
- Laura F. Wetherill
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Kristopher K. Holmes
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Mark Verow
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Marietta Müller
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Gareth Howell
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Mark Harris
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Colin Fishwick
- School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - Nicola Stonehouse
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Richard Foster
- School of Chemistry, University of Leeds, Leeds, United Kingdom
| | - G. Eric Blair
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Stephen Griffin
- Leeds Institute of Molecular Medicine, Faculty of Medicine and Health, St. James's University Hospital, University of Leeds, Leeds, United Kingdom
| | - Andrew Macdonald
- Institute of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
48
|
Klingelhutz AJ, Roman A. Cellular transformation by human papillomaviruses: lessons learned by comparing high- and low-risk viruses. Virology 2012; 424:77-98. [PMID: 22284986 DOI: 10.1016/j.virol.2011.12.018] [Citation(s) in RCA: 113] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Revised: 12/17/2011] [Accepted: 12/27/2011] [Indexed: 12/19/2022]
Abstract
The oncogenic potential of papillomaviruses (PVs) has been appreciated since the 1930s yet the mechanisms of virally-mediated cellular transformation are still being revealed. Reasons for this include: a) the oncoproteins are multifunctional, b) there is an ever-growing list of cellular interacting proteins, c) more than one cellular protein may bind to a given region of the oncoprotein, and d) there is only limited information on the proteins encoded by the corresponding non-oncogenic PVs. The perspective of this review will be to contrast the activities of the viral E6 and E7 proteins encoded by the oncogenic human PVs (termed high-risk HPVs) to those encoded by their non-oncogenic counterparts (termed low-risk HPVs) in an attempt to sort out viral life cycle-related functions from oncogenic functions. The review will emphasize lessons learned from the cell culture studies of the HPVs causing mucosal/genital tract cancers.
Collapse
|
49
|
Woodworth CD, Diefendorf LP, Jette DF, Mohammed A, Moses MA, Searleman SA, Stevens DA, Wilton KM, Mondal S. Inhibition of the epidermal growth factor receptor by erlotinib prevents immortalization of human cervical cells by Human Papillomavirus type 16. Virology 2011; 421:19-27. [PMID: 21982220 DOI: 10.1016/j.virol.2011.09.014] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Revised: 08/11/2011] [Accepted: 09/14/2011] [Indexed: 11/19/2022]
Abstract
The Human Papillomavirus type-16 (HPV-16) E6 and E7 oncogenes are selectively retained and expressed in cervical carcinomas, and expression of E6 and E7 is sufficient to immortalize human cervical epithelial cells. Expression of the epidermal growth factor receptor (EGFR) is often increased in cervical dysplasia and carcinoma, and HPV oncoproteins stimulate cell growth via the EGFR pathway. We found that erlotinib, a specific inhibitor of EGFR tyrosine kinase activity, prevented immortalization of cultured human cervical epithelial cells by the complete HPV-16 genome or the E6/E7 oncogenes. Erlotinib stimulated apoptosis in cells that expressed HPV-16 E6/E7 proteins and induced senescence in a subpopulation of cells that did not undergo apoptosis. Since immortalization by HPV E6/E7 is an important early event in cervical carcinogenesis, the EGFR is a potential target for chemoprevention or therapy in women who have a high risk for cervical cancer.
Collapse
Affiliation(s)
- Craig D Woodworth
- Department of Biology, Clarkson University, Potsdam, NY 13699-5805, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
The human papillomavirus type 16 E5 oncoprotein translocates calpactin I to the perinuclear region. J Virol 2011; 85:10968-75. [PMID: 21849434 DOI: 10.1128/jvi.00706-11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The human papillomavirus type 16 (HPV-16) E5 oncoprotein is embedded in membranes of the endoplasmic reticulum and nuclear envelope with its C terminus exposed to the cytoplasm. Among other activities, E5 cooperates with the HPV E6 oncoprotein to induce koilocytosis in human cervical cells and keratinocytes in vitro. The effect of E5 on infected cells may rely on its interactions with various cellular proteins. In this study we identify calpactin I, a heterotetrameric, Ca(2+)- and phospholipid-binding protein complex that regulates membrane fusion, as a new cellular target for E5. Both the annexin A2 and p11 subunits of calpactin I coimmunoprecipitate with E5 in COS cells and in human epithelial cell lines, and an intact E5 C terminus is required for binding. Moreover, E5-expressing cells exhibit a perinuclear redistribution of annexin A2 and p11 and show increased fusion of perinuclear membrane vesicles. The C terminus of E5 is required for both the perinuclear redistribution of calpactin I and increased formation of perinuclear vacuoles. These results support the hypothesis that the E5-induced relocalization of calpactin I to the perinuclear region promotes perinuclear membrane fusion, which may underlie the development of koilocytotic vacuoles.
Collapse
|