1
|
Mikuličić S, Massenberg A, Döring T, Brandenburg K, Lang T, Florin L. HSPG-binding peptide Pep19-2.5 is a potent inhibitor of HPV16 infection. Antimicrob Agents Chemother 2025; 69:e0157524. [PMID: 39807885 PMCID: PMC11823620 DOI: 10.1128/aac.01575-24] [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: 10/23/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025] Open
Abstract
Peptide-based therapeutics are gaining attention for their potential to target various viral and host cell factors. One notable example is Pep19-2.5 (Aspidasept), a synthetic anti-lipopolysaccharide peptide that binds to heparan sulfate proteoglycans (HSPGs) and has demonstrated inhibitory effects against certain bacteria and enveloped viruses. This study explores, for the first time, the effectiveness of Pep19-2.5 against a non-enveloped virus, using pseudoviruses of the oncogenic human papillomavirus type 16 (HPV16) as a model. HPV16 infects epithelial cells of the skin and mucosa by using multiple cell surface receptors with initial attachment to HSPGs. Pharmacological inhibition with Pep19-2.5 in HeLa and HaCaT cells resulted in a concentration-dependent reduction of HPV16 PsV infection, with near-complete blockade observed at higher concentrations. The half-maximal inhibitory concentration (IC50) was determined to be 116 nM in HeLa cells and 183 nM in HaCaT cells, highlighting its potent antiviral activity. Our results demonstrate that Pep19-2.5 not only inhibits HPV16 PsV binding to the cell surface but also significantly reduces infection when administered post-binding. Imaging analyses revealed Pep19-2.5-dependent release of large cell-associated crowds of viral particles, suggesting interference with the transfer to secondary receptor molecules. This was corroborated by the effectiveness of Pep19-2.5 in an HSPG-negative cell line, indicating that the peptide disrupts virus binding to both primary and secondary interaction partners. Based on these findings, we propose that the antimicrobial effect of Pep19-2.5 is not limited to HSPG-dependent infections. Additionally, Pep19-2.5 may be a valuable tool for dissecting specific steps in the viral entry process.
Collapse
Affiliation(s)
- Snježana Mikuličić
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Annika Massenberg
- University of Bonn, Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, Bonn, Germany
| | - Tatjana Döring
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Klaus Brandenburg
- Brandenburg Antiinfektiva GmbH, Research Center Borstel-Leibniz-Lung Center, Borstel, Germany
| | - Thorsten Lang
- University of Bonn, Faculty of Mathematics and Natural Sciences, Life & Medical Sciences (LIMES) Institute, Bonn, Germany
| | - Luise Florin
- Institute for Virology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
- Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
2
|
Feng Y, van Bodegraven D, Kádek A, L B Munguira I, Soria-Martinez L, Nentwich S, Saha S, Chardon F, Kavan D, Uetrecht C, Schelhaas M, Roos WH. Glycan-induced structural activation softens the human papillomavirus capsid for entry through reduction of intercapsomere flexibility. Nat Commun 2024; 15:10076. [PMID: 39572555 PMCID: PMC11582657 DOI: 10.1038/s41467-024-54373-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 11/05/2024] [Indexed: 11/24/2024] Open
Abstract
High-risk human papillomaviruses (HPVs) cause various cancers. While type-specific prophylactic vaccines are available, additional anti-viral strategies are highly desirable. Initial HPV cell entry involves receptor-switching induced by structural capsid modifications. These modifications are initiated by interactions with cellular heparan sulphates (HS), however, their molecular nature and functional consequences remain elusive. Combining virological assays with hydrogen/deuterium exchange mass spectrometry, and atomic force microscopy, we investigate the effect of capsid-HS binding and structural activation. We show how HS-induced structural activation requires a minimal HS-chain length and simultaneous engagement of several binding sites by a single HS molecule. This engagement introduces a pincer-like force that stabilizes the capsid in a conformation with extended capsomer linkers. It results in capsid enlargement and softening, thereby likely facilitating L1 proteolytic cleavage and subsequent L2-externalization, as needed for cell entry. Our data supports the further devising of prophylactic strategies against HPV infections.
Collapse
Affiliation(s)
- Yuzhen Feng
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
| | | | - Alan Kádek
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY & Leibniz Institute of Virology (LIV), Notkestraße 85, Hamburg, Germany
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Ignacio L B Munguira
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands
| | | | - Sarah Nentwich
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY & Leibniz Institute of Virology (LIV), Notkestraße 85, Hamburg, Germany
| | - Sreedeepa Saha
- Institute of Cellular Virology, ZMBE, University of Münster, Münster, Germany
| | - Florian Chardon
- Institute of Cellular Virology, ZMBE, University of Münster, Münster, Germany
| | - Daniel Kavan
- Institute of Microbiology of the Czech Academy of Sciences, Videnska 1083, Prague, Czech Republic
| | - Charlotte Uetrecht
- CSSB Centre for Structural Systems Biology, Deutsches Elektronen-Synchrotron DESY & Leibniz Institute of Virology (LIV), Notkestraße 85, Hamburg, Germany.
- Institute of Chemistry and Metabolomics, University of Lübeck, Ratzeburger Allee 160, Lübeck, Germany.
| | - Mario Schelhaas
- Institute of Cellular Virology, ZMBE, University of Münster, Münster, Germany.
| | - Wouter H Roos
- Moleculaire Biofysica, Zernike Instituut, Rijksuniversiteit Groningen, Groningen, Netherlands.
| |
Collapse
|
3
|
Ishii Y, Yamaji T, Sekizuka T, Homma Y, Mori S, Takeuchi T, Kukimoto I. Folliculin Prevents Lysosomal Degradation of Human Papillomavirus To Support Infectious Cell Entry. J Virol 2023; 97:e0005623. [PMID: 37167561 PMCID: PMC10231244 DOI: 10.1128/jvi.00056-23] [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: 01/12/2023] [Accepted: 04/20/2023] [Indexed: 05/13/2023] Open
Abstract
Human papillomavirus (HPV) infects epithelial basal cells in the mucosa and either proliferates with the differentiation of the basal cells or persists in them. Multiple host factors are required to support the HPV life cycle; however, the molecular mechanisms involved in cell entry are not yet fully understood. In this study, we performed a genome-wide clustered regularly interspaced short palindromic repeat (CRISPR)-CRISPR-associated protein 9 (Cas9) knockout (KO) screen in HeLa cells and identified folliculin (FLCN), a GTPase-activating protein for Rag GTPases, as an important host factor for HPV infection. The introduction of single guide RNAs for the FLCN gene into HeLa, HaCaT, and ectocervical Ect1 cells reduced infection by HPV18 pseudovirions (18PsVs) and 16PsVs. FLCN KO HeLa cells also exhibited strong resistance to infection with 18PsVs and 16PsVs; nevertheless, they remained highly susceptible to infections with vesicular stomatitis virus glycoprotein-pseudotyped lentivirus and adeno-associated virus. Immunofluorescence microscopy revealed that the numbers of virions binding to the cell surface were slightly increased in FLCN KO cells. However, virion internalization analysis showed that the internalized virions were rapidly degraded in FLCN KO cells. This degradation was blocked by treatment with the lysosome inhibitor bafilomycin A1. Furthermore, the virion degradation phenotype was also observed in Ras-related GTP-binding protein C (RagC) KO cells. These results suggest that FLCN prevents the lysosomal degradation of incoming HPV virions by enhancing lysosomal RagC activity. IMPORTANCE Cell entry by human papillomavirus (HPV) involves a cellular retrograde transport pathway from the endosome to the trans-Golgi network/Golgi apparatus. However, the mechanism by which this viral trafficking is safeguarded is poorly understood. This is the first study showing that the GTPase-activating protein folliculin (FLCN) protects incoming HPV virions from lysosomal degradation and supports infectious cell entry by activating the Rag GTPases, presumably through the suppression of excessive lysosomal biosynthesis. These findings provide new insights into the effects of small GTPase activity regulation on HPV cell entry and enhance our understanding of the HPV degradation pathway.
Collapse
Affiliation(s)
- Yoshiyuki Ishii
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Toshiyuki Yamaji
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Tsuyoshi Sekizuka
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuta Homma
- Department of Biochemistry and Cell Biology, National Institute of Infectious Diseases, Tokyo, Japan
| | - Seiichiro Mori
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takamasa Takeuchi
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| | - Iwao Kukimoto
- Pathogen Genomics Center, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
4
|
Mayerberger EA, Yazdanparast Tafti S, Jedlicka SS, Jellison KL. Effect of Glycosaminoglycans on Cryptosporidium Oocyst Attachment and Excystation. Appl Environ Microbiol 2023; 89:e0173722. [PMID: 36790186 PMCID: PMC10056967 DOI: 10.1128/aem.01737-22] [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: 10/18/2022] [Accepted: 01/11/2023] [Indexed: 02/16/2023] Open
Abstract
Cryptosporidium causes severe gastrointestinal disease resulting from the ingestion of oocysts, followed by oocyst excystation in the small intestine and the release of infective sporozoites. An understudied strategy for Cryptosporidium inactivation is purposeful oocyst excystation, as sporozoites do not survive long in the environment. This study showed that C. parvum oocyst excystation was induced by direct contact with various glycosaminoglycans (GAGs), including heparin (Hep), chondroitin sulfate A (CSA), and hyaluronan (HA), assembled on polydopamine (PD)-functionalized surfaces. PD surfaces elicited 97.9 ± 3.6% oocyst attachment, with some of the attached oocysts partially (7.3 ± 1.3%) or fully (4.0 ± 0.6%) excysted after 4 days. The PD-GAG surfaces (GAG concentration = 2 mg/mL) elicited similarly high attachment (>97%) and higher oocyst excystation efficiencies after 4 days. The PD-Hep surfaces elicited the highest number of attached excysted oocysts (11.8 ± 0.63% partially excysted; 11.9 ± 0.49% fully excysted), and the PD-HA surfaces elicited the lowest (8.8 ± 2.1% partially excysted; 7.8 ± 1.2% fully excysted). Surface characterization revealed that the addition of GAGs to the PD surface changed both the surface roughness as well as the surface wettability. Treatment of oocysts with an enzyme that degraded the surface glycocalyx markedly reduced excystation (to <2%) of the oocysts attached to the PD and PD-GAG surfaces. These findings suggest that GAGs provide an important local signal for the excystation of C. parvum oocysts and that certain surface-expressed oocyst receptors are necessary for efficient excystation. These oocyst-receptor relationships may be useful in the design of functionalized surfaces for the purposeful inactivation of oocysts in the environment or in water treatment systems. IMPORTANCE Polydopamine surfaces functionalized with glycosaminoglycans were shown to facilitate the attachment and excystation of Cryptosporidium parvum oocysts. Our findings suggest that a surface-expressed receptor on the oocyst wall plays a key role in excystation, with glycosaminoglycans serving as ligands that trigger the initiation of the process. Future technologies and treatment strategies designed to promote premature excystation of oocysts will minimize the ingestion of sporozoites that initiate infection. Therefore, the results from this study have important implications for the protection of public health from waterborne cryptosporidiosis and may serve as a foundation for engineered surfaces designed to remove oocysts from surface waters or inactivate oocysts in water treatment systems.
Collapse
Affiliation(s)
- Elisa A. Mayerberger
- Department of Civil and Environmental Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | | | - Sabrina S. Jedlicka
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania, USA
- Department of Materials Science and Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| | - Kristen L. Jellison
- Department of Civil and Environmental Engineering, Lehigh University, Bethlehem, Pennsylvania, USA
| |
Collapse
|
5
|
Ishii Y, Mori S, Kukimoto I. [Identification of new host factors supporting the human papillomavirus life cycle]. Uirusu 2023; 73:189-198. [PMID: 39343554 DOI: 10.2222/jsv.73.189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
|
6
|
Chondroitin Sulfate Proteoglycans Are De Facto Cellular Receptors for Human Papillomavirus 16 under High Serum Conditions. J Virol 2022; 96:e0185721. [DOI: 10.1128/jvi.01857-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Heparan sulfate proteoglycans (HSPGs) have previously been identified as primary attachment factors for the initial binding of human papillomaviruses (HPVs) prior to infection. Here, we demonstrate that
in vitro
, HPV binding to HSPGs is strongly dependent on the surrounding experimental conditions, including the concentration of fetal bovine serum (FBS).
Collapse
|
7
|
Mikuličić S, Strunk J, Florin L. HPV16 Entry into Epithelial Cells: Running a Gauntlet. Viruses 2021; 13:v13122460. [PMID: 34960729 PMCID: PMC8706107 DOI: 10.3390/v13122460] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/16/2022] Open
Abstract
During initial infection, human papillomaviruses (HPV) take an unusual trafficking pathway through their host cell. It begins with a long period on the cell surface, during which the capsid is primed and a virus entry platform is formed. A specific type of clathrin-independent endocytosis and subsequent retrograde trafficking to the trans-Golgi network follow this. Cellular reorganization processes, which take place during mitosis, enable further virus transport and the establishment of infection while evading intrinsic cellular immune defenses. First, the fragmentation of the Golgi allows the release of membrane-encased virions, which are partially protected from cytoplasmic restriction factors. Second, the nuclear envelope breakdown opens the gate for these virus–vesicles to the cell nucleus. Third, the dis- and re-assembly of the PML nuclear bodies leads to the formation of modified virus-associated PML subnuclear structures, enabling viral transcription and replication. While remnants of the major capsid protein L1 and the viral DNA remain in a transport vesicle, the viral capsid protein L2 plays a crucial role during virus entry, as it adopts a membrane-spanning conformation for interaction with various cellular proteins to establish a successful infection. In this review, we follow the oncogenic HPV type 16 during its long journey into the nucleus, and contrast pro- and antiviral processes.
Collapse
|
8
|
Ozbun MA, Campos SK. The long and winding road: human papillomavirus entry and subcellular trafficking. Curr Opin Virol 2021; 50:76-86. [PMID: 34416595 DOI: 10.1016/j.coviro.2021.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 07/26/2021] [Accepted: 07/27/2021] [Indexed: 12/28/2022]
Abstract
Human papillomaviruses (HPVs) infect and replicate in differentiating mucosal and cutaneous epithelium. Most HPV infections are asymptomatic or cause transient benign neoplasia. However, persistent infections by oncogenic HPV types can progress to cancer. During infectious entry into host keratinocytes, HPV particles interact with many host proteins, beginning with major capsid protein L1 binding to cellular heparan sulfate and a series of enzymatic capsid modifications that promote infectious cellular entry. After utilizing the endosomal pathway to uncoat the viral genome (vDNA), the minor capsid protein L2/vDNA complex is retrograde trafficked to the Golgi, and thereafter, to the nucleus where viral transcription initiates. Post-Golgi trafficking is dependent on mitosis, with L2-dependent tethering of vDNA to mitotic chromosomes before accumulation at nuclear substructures in G1. This review summarizes the current knowledge of the HPV entry pathway, the role of cellular proteins in this process, and notes many gaps in our understanding.
Collapse
Affiliation(s)
- Michelle A Ozbun
- Departments of Molecular Genetics & Microbiology, Obstetrics & Gynecology, The University of New Mexico School of Medicine, Albuquerque, NM 87131, USA; The University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87131, USA.
| | - Samuel K Campos
- Departments of Immunobiology, Molecular & Cellular Biology, and the Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, AZ 85721, USA; The BIO5 Institute, University of Arizona, Tucson, AZ 85721, USA.
| |
Collapse
|
9
|
Lai KY, Rizzato M, Aydin I, Villalonga-Planells R, Drexler HCA, Schelhaas M. A Ran-binding protein facilitates nuclear import of human papillomavirus type 16. PLoS Pathog 2021; 17:e1009580. [PMID: 33974675 PMCID: PMC8139508 DOI: 10.1371/journal.ppat.1009580] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/21/2021] [Accepted: 04/23/2021] [Indexed: 01/01/2023] Open
Abstract
Human papillomaviruses (HPVs) utilize an atypical mode of nuclear import during cell entry. Residing in the Golgi apparatus until mitosis onset, a subviral complex composed of the minor capsid protein L2 and viral DNA (L2/vDNA) is imported into the nucleus after nuclear envelope breakdown by associating with mitotic chromatin. In this complex, L2 plays a crucial role in the interactions with cellular factors that enable delivery and ultimately tethering of the viral genome to mitotic chromatin. To date, the cellular proteins facilitating these steps remain unknown. Here, we addressed which cellular proteins may be required for this process. Using label-free mass spectrometry, biochemical assays, microscopy, and functional virological assays, we discovered that L2 engages a hitherto unknown protein complex of Ran-binding protein 10 (RanBP10), karyopherin alpha2 (KPNA2), and dynein light chain DYNLT3 to facilitate transport towards mitotic chromatin. Thus, our study not only identifies novel cellular interactors and mechanism that facilitate a poorly understood step in HPV entry, but also a novel cellular transport complex. Human papillomaviruses (HPVs) cause proliferative lesions such as benign warts or malignant invasive cancers. Like other DNA viruses, HPV has to deliver its genome to the nucleus for viral genome transcription and replication. After initial attachment, HPVs are endocytosed to be eventually directed to the trans-Golgi-network (TGN) by intracellular trafficking, where they reside until cell division. Mitosis onset enables access of the virus to cellular chromatin after nuclear envelope breakdown. Tethering of the virus to mitotic chromatin ensures nuclear delivery upon reformation of the nuclear envelope after mitosis. Our previous work showed that the minor capsid protein L2 facilitates nuclear delivery. However, the detailed mechanism, namely, how HPV trafficks from cytosol to the nuclear space, is barely understood. Here, we identified for the first time cellular proteins that interacted with L2 for nuclear import. Mechanistically, the proteins formed a hitherto unknown cellular transport complex that interacted with L2 to direct the virus to mitotic chromosomes by microtubular transport. Our findings provided not only evidence for a transport mechanism of a poorly understood step of HPV entry, but also discovered a novel cellular transport complex.
Collapse
Affiliation(s)
- Kun-Yi Lai
- Institute of Cellular Virology, Westphalian Wilhelms-University of Münster, Münster, Germany
- Interfaculty Centre ‘Cells in Motion’ (CiM), Westphalian Wilhelms-University of Münster, Münster, Germany
| | - Matteo Rizzato
- Institute of Cellular Virology, Westphalian Wilhelms-University of Münster, Münster, Germany
| | - Inci Aydin
- Institute of Cellular Virology, Westphalian Wilhelms-University of Münster, Münster, Germany
| | | | - Hannes C. A. Drexler
- Biomolecular Mass Spectrometry Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Mario Schelhaas
- Institute of Cellular Virology, Westphalian Wilhelms-University of Münster, Münster, Germany
- Interfaculty Centre ‘Cells in Motion’ (CiM), Westphalian Wilhelms-University of Münster, Münster, Germany
- * E-mail:
| |
Collapse
|
10
|
Carse S, Bergant M, Schäfer G. Advances in Targeting HPV Infection as Potential Alternative Prophylactic Means. Int J Mol Sci 2021; 22:2201. [PMID: 33672181 PMCID: PMC7926419 DOI: 10.3390/ijms22042201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/15/2021] [Accepted: 02/19/2021] [Indexed: 01/22/2023] Open
Abstract
Infection by oncogenic human papillomavirus (HPV) is the primary cause of cervical cancer and other anogenital cancers. The majority of cervical cancer cases occur in low- and middle- income countries (LMIC). Concurrent infection with Human Immunodeficiency Virus (HIV) further increases the risk of HPV infection and exacerbates disease onset and progression. Highly effective prophylactic vaccines do exist to combat HPV infection with the most common oncogenic types, but the accessibility to these in LMIC is severely limited due to cost, difficulties in accessing the target population, cultural issues, and maintenance of a cold chain. Alternative preventive measures against HPV infection that are more accessible and affordable are therefore also needed to control cervical cancer risk. There are several efforts in identifying such alternative prophylactics which target key molecules involved in early HPV infection events. This review summarizes the current knowledge of the initial steps in HPV infection, from host cell-surface engagement to cellular trafficking of the viral genome before arrival in the nucleus. The key molecules that can be potentially targeted are highlighted, and a discussion on their applicability as alternative preventive means against HPV infection, with a focus on LMIC, is presented.
Collapse
Affiliation(s)
- Sinead Carse
- International Centre for Genetic Engineering and Biotechnology (ICGEB) Cape Town, Observatory 7925, South Africa;
- Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| | - Martina Bergant
- Laboratory for Environmental and Life Sciences, University of Nova Gorica, Vipavska 13, 5000 Nova Gorica, Slovenia;
| | - Georgia Schäfer
- International Centre for Genetic Engineering and Biotechnology (ICGEB) Cape Town, Observatory 7925, South Africa;
- Institute of Infectious Disease and Molecular Medicine (IDM), Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
- Division of Medical Biochemistry and Structural Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Observatory 7925, South Africa
| |
Collapse
|
11
|
Uhlorn BL, Jackson R, Li S, Bratton SM, Van Doorslaer K, Campos SK. Vesicular trafficking permits evasion of cGAS/STING surveillance during initial human papillomavirus infection. PLoS Pathog 2020; 16:e1009028. [PMID: 33253291 PMCID: PMC7728285 DOI: 10.1371/journal.ppat.1009028] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 12/10/2020] [Accepted: 10/02/2020] [Indexed: 12/26/2022] Open
Abstract
Oncogenic human papillomaviruses (HPVs) replicate in differentiating epithelium, causing 5% of cancers worldwide. Like most other DNA viruses, HPV infection initiates after trafficking viral genome (vDNA) to host cell nuclei. Cells possess innate surveillance pathways to detect microbial components or physiological stresses often associated with microbial infections. One of these pathways, cGAS/STING, induces IRF3-dependent antiviral interferon (IFN) responses upon detection of cytosolic DNA. Virion-associated vDNA can activate cGAS/STING during initial viral entry and uncoating/trafficking, and thus cGAS/STING is an obstacle to many DNA viruses. HPV has a unique vesicular trafficking pathway compared to many other DNA viruses. As the capsid uncoats within acidic endosomal compartments, minor capsid protein L2 protrudes across vesicular membranes to facilitate transport of vDNA to the Golgi. L2/vDNA resides within the Golgi lumen until G2/M, whereupon vesicular L2/vDNA traffics along spindle microtubules, tethering to chromosomes to access daughter cell nuclei. L2/vDNA-containing vesicles likely remain intact until G1, following nuclear envelope reformation. We hypothesize that this unique vesicular trafficking protects HPV from cGAS/STING surveillance. Here, we investigate cGAS/STING responses to HPV infection. DNA transfection resulted in acute cGAS/STING activation and downstream IFN responses. In contrast, HPV infection elicited minimal cGAS/STING and IFN responses. To determine the role of vesicular trafficking in cGAS/STING evasion, we forced premature viral penetration of vesicular membranes with membrane-perturbing cationic lipids. Such treatment renders a non-infectious trafficking-defective mutant HPV infectious, yet susceptible to cGAS/STING detection. Overall, HPV evades cGAS/STING by its unique subcellular trafficking, a property that may contribute to establishment of infection.
Collapse
Affiliation(s)
- Brittany L. Uhlorn
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona, United States of America
| | - Robert Jackson
- School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, United States of America
| | - Shuaizhi Li
- Department of Immunobiology, The University of Arizona, Tucson, Arizona, United States of America
| | - Shauna M. Bratton
- Department of Physiology, The University of Arizona, Tucson, Arizona, United States of America
| | - Koenraad Van Doorslaer
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona, United States of America
- School of Animal & Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, The University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, The University of Arizona, Tucson, Arizona, United States of America
- Genetics Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona, United States of America
| | - Samuel K. Campos
- Cancer Biology Graduate Interdisciplinary Program, The University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, The University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, The University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular & Cellular Biology, The University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
12
|
Harwood MC, Dupzyk AJ, Inoue T, DiMaio D, Tsai B. p120 catenin recruits HPV to γ-secretase to promote virus infection. PLoS Pathog 2020; 16:e1008946. [PMID: 33085724 PMCID: PMC7577436 DOI: 10.1371/journal.ppat.1008946] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 08/28/2020] [Indexed: 12/29/2022] Open
Abstract
During internalization and trafficking, human papillomavirus (HPV) moves from the cell surface to the endosome where the transmembrane protease γ-secretase promotes insertion of the viral L2 capsid protein into the endosome membrane. Protrusion of L2 through the endosome membrane into the cytosol allows the recruitment of cytosolic host factors that target the virus to the Golgi en route for productive infection. How endosome-localized HPV is delivered to γ-secretase, a decisive infection step, is unclear. Here we demonstrate that cytosolic p120 catenin, likely via an unidentified transmembrane protein, interacts with HPV at early time-points during viral internalization and trafficking. In the endosome, p120 is not required for low pH-dependent disassembly of the HPV L1 capsid protein from the incoming virion. Rather, p120 is required for HPV to interact with γ-secretase-an interaction that ensures the virus is transported along a productive route. Our findings clarify an enigmatic HPV infection step and provide critical insights into HPV infection that may lead to new therapeutic strategies against HPV-induced diseases.
Collapse
Affiliation(s)
- Mara Calypso Harwood
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Allison Jade Dupzyk
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, United States of America
| | - Takamasa Inoue
- Pathogen Research Section, Central Research Laboratory, Research and Development Division, Japan Blood Products Organization, Kobe, Japan
| | - Daniel DiMaio
- Department of Genetics, Yale School of Medicine, New Haven, CT, United States of America
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- * E-mail:
| |
Collapse
|
13
|
Epidermal Growth Factor Receptor and Abl2 Kinase Regulate Distinct Steps of Human Papillomavirus 16 Endocytosis. J Virol 2020; 94:JVI.02143-19. [PMID: 32188731 DOI: 10.1128/jvi.02143-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 03/11/2020] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus 16 (HPV16), the leading cause of cervical cancer, exploits a novel endocytic pathway during host cell entry. This mechanism shares many requirements with macropinocytosis but differs in the mode of vesicle formation. Previous work indicated a role of the epidermal growth factor receptor (EGFR) in HPV16 endocytosis. However, the functional outcome of EGFR signaling and its downstream targets during HPV16 uptake are not well characterized. Here, we analyzed the functional importance of signal transduction via EGFR and its downstream effectors for endocytosis of HPV16. Our findings indicate two phases of EGFR signaling as follows: a-likely dispensable-transient activation with or shortly after cell binding and signaling required throughout the process of asynchronous internalization of HPV16. Interestingly, EGFR inhibition interfered with virus internalization and strongly reduced the number of endocytic pits, suggesting a role for EGFR signaling in the induction of HPV16 endocytosis. Moreover, we identified the Src-related kinase Abl2 as a novel regulator of virus uptake. Inhibition of Abl2 resulted in an accumulation of misshaped endocytic pits, indicating Abl2's importance for endocytic vesicle maturation. Since Abl2 rather than Src, a regulator of membrane ruffling during macropinocytosis, mediated downstream signaling of EGFR, we propose that the selective effector targeting downstream of EGFR determines whether HPV16 endocytosis or macropinocytosis is induced.IMPORTANCE Human papillomaviruses are small, nonenveloped DNA viruses that infect skin and mucosa. The so-called high-risk HPVs (e.g., HPV16, HPV18, HPV31) have transforming potential and are associated with various anogenital and oropharyngeal tumors. These viruses enter host cells by a novel endocytic pathway with unknown cellular function. To date, it is unclear how endocytic vesicle formation occurs mechanistically. Here, we addressed the role of epidermal growth factor receptor signaling, which has previously been implicated in HPV16 endocytosis and identified the kinase Abl2 as a novel regulator of virus uptake. Since other viruses, such as influenza A virus and lymphocytic choriomeningitis virus, possibly make use of related mechanisms, our findings shed light on fundamental strategies of virus entry and may in turn help to develop new host cell-targeted antiviral strategies.
Collapse
|
14
|
De Gregorio V, Urciuolo F, Netti PA, Imparato G. In Vitro Organotypic Systems to Model Tumor Microenvironment in Human Papillomavirus (HPV)-Related Cancers. Cancers (Basel) 2020; 12:E1150. [PMID: 32375253 PMCID: PMC7281263 DOI: 10.3390/cancers12051150] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022] Open
Abstract
Despite the well-known role of chronic human papillomavirus (HPV) infections in causing tumors (i.e., all cervical cancers and other human malignancies from the mucosal squamous epithelia, including anogenital and oropharyngeal cavity), its persistence is not sufficient for cancer development. Other co-factors contribute to the carcinogenesis process. Recently, the critical role of the underlying stroma during the HPV life cycle and HPV-induced disease have been investigated. The tumor stroma is a key component of the tumor microenvironment (TME), which is a specialized entity. The TME is dynamic, interactive, and constantly changing-able to trigger, support, and drive tumor initiation, progression, and metastasis. In previous years, in vitro organotypic raft cultures and in vivo genetically engineered mouse models have provided researchers with important information on the interactions between HPVs and the epithelium. Further development for an in-depth understanding of the interaction between HPV-infected tissue and the surrounding microenvironment is strongly required. In this review, we critically describe the HPV-related cancers modeled in vitro from the simplified 'raft culture' to complex three-dimensional (3D) organotypic models, focusing on HPV-associated cervical cancer disease platforms. In addition, we review the latest knowledge in the field of in vitro culture systems of HPV-associated malignancies of other mucosal squamous epithelia (anogenital and oropharynx), as well as rare cutaneous non-melanoma associated cancer.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Francesco Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
| | - Paolo Antonio Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, 80125 Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
15
|
Soria-Martinez L, Bauer S, Giesler M, Schelhaas S, Materlik J, Janus K, Pierzyna P, Becker M, Snyder NL, Hartmann L, Schelhaas M. Prophylactic Antiviral Activity of Sulfated Glycomimetic Oligomers and Polymers. J Am Chem Soc 2020; 142:5252-5265. [PMID: 32105452 DOI: 10.1021/jacs.9b13484] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
In this work, we investigate the potential of highly sulfated synthetic glycomimetics to act as inhibitors of viral binding/infection. Our results indicate that both long-chain glycopolymers and short-chain glycooligomers are capable of preventing viral infection. Notably, glycopolymers efficiently inhibit Human Papillomavirus (HPV16) infection in vitro and maintain their antiviral activity in vivo, while the glycooligomers exert their inhibitory function post attachment of viruses to cells. Moreover, when we tested the potential for broader activity against several other human pathogenic viruses, we observed broad-spectrum antiviral activity of these compounds beyond our initial assumptions. While the compounds tested displayed a range of antiviral efficacies, viruses with rather diverse glycan specificities such as Herpes Simplex Virus (HSV), Influenza A Virus (IAV), and Merkel Cell Polyomavirus (MCPyV) could be targeted. This opens new opportunities to develop broadly active glycomimetic inhibitors of viral entry and infection.
Collapse
Affiliation(s)
- Laura Soria-Martinez
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany
| | - Sebastian Bauer
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Markus Giesler
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Sonja Schelhaas
- European Institute for Molecular Imaging, University of Münster, Münster 48149, Germany.,Cells in Motion Interfaculty Centre CiMIC, University of Münster, Münster 48149, Germany
| | - Jennifer Materlik
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Kevin Janus
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Patrick Pierzyna
- Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Miriam Becker
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany
| | - Nicole L Snyder
- Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Department of Chemistry, Davidson College, Davidson, North Carolina 28035, United States
| | - Laura Hartmann
- Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Institute of Organic Chemistry and Macromolecular Chemistry, Heinrich-Heine-University Düsseldorf, Düsseldorf 40225, Germany
| | - Mario Schelhaas
- Institute of Cellular Virology, ZMBE, University of Münster, Münster 48149, Germany.,Research Group "ViroCarb: glycans controlling non-enveloped virus infections" (FOR2327), Coordinating University of Tübingen, Tübingen 72074, Germany.,Cells in Motion Interfaculty Centre CiMIC, University of Münster, Münster 48149, Germany
| |
Collapse
|
16
|
Influence of cell-penetrating peptides on the activity and stability of virus-based nanoparticles. Int J Pharm 2020; 576:119008. [DOI: 10.1016/j.ijpharm.2019.119008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/23/2019] [Accepted: 12/29/2019] [Indexed: 02/06/2023]
|
17
|
Infectious Entry of Merkel Cell Polyomavirus. J Virol 2019; 93:JVI.02004-18. [PMID: 30626687 DOI: 10.1128/jvi.02004-18] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/23/2018] [Indexed: 12/17/2022] Open
Abstract
Merkel cell polyomavirus (MCPyV) is a small, nonenveloped tumor virus associated with an aggressive form of skin cancer, Merkel cell carcinoma (MCC). MCPyV infections are highly prevalent in the human population, with MCPyV virions being continuously shed from human skin. However, the precise host cell tropism(s) of MCPyV remains unclear: MCPyV is able to replicate within a subset of dermal fibroblasts, but MCPyV DNA has also been detected in a variety of other tissues. However, MCPyV appears different from other polyomaviruses, as it requires sulfated polysaccharides, such as heparan sulfates and/or chondroitin sulfates, for initial attachment. Like other polyomaviruses, MCPyV engages sialic acid as a (co)receptor. To explore the infectious entry process of MCPyV, we analyzed the cell biological determinants of MCPyV entry into A549 cells, a highly transducible lung carcinoma cell line, in comparison to well-studied simian virus 40 and a number of other viruses. Our results indicate that MCPyV enters cells via caveolar/lipid raft-mediated endocytosis but not macropinocytosis, clathrin-mediated endocytosis, or glycosphingolipid-enriched carriers. The viruses were internalized in small endocytic pits that led the virus to endosomes and from there to the endoplasmic reticulum (ER). Similar to other polyomaviruses, trafficking required microtubular transport, acidification of endosomes, and a functional redox environment. To our surprise, the virus was found to acquire a membrane envelope within endosomes, a phenomenon not reported for other viruses. Only minor amounts of viruses reached the ER, while the majority was retained in endosomal compartments, suggesting that endosome-to-ER trafficking is a bottleneck during infectious entry.IMPORTANCE MCPyV is the first polyomavirus directly implicated in the development of an aggressive human cancer, Merkel cell carcinoma (MCC). Although MCPyV is constantly shed from healthy skin, the MCC incidence increases among aging and immunocompromised individuals. To date, the events connecting initial MCPyV infection and subsequent transformation still remain elusive. MCPyV differs from other known polyomaviruses concerning its cell tropism, entry receptor requirements, and infection kinetics. In this study, we examined the cellular requirements for endocytic entry as well as the subcellular localization of incoming virus particles. A thorough understanding of the determinants of the infectious entry pathway and the specific biological niche will benefit prevention of virus-derived cancers such as MCC.
Collapse
|
18
|
Glycans Controlling Virus Infections: Meeting Report on the 1st International Symposium on Glycovirology Schöntal, Germany, 02⁻04 May 2018. Viruses 2018; 10:v10110636. [PMID: 30445709 PMCID: PMC6266297 DOI: 10.3390/v10110636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 11/12/2018] [Indexed: 12/30/2022] Open
Abstract
Glycans are, with nucleic acids, proteins and lipids, one of the four founding structures of cellular life. Due to their non-template synthesis, they are inherently heterogeneous and difficult to study with regards to their structure and function. Since 2016, the research group ViroCarb, funded by the German Research Foundation, has investigated the role of glycans in non-enveloped virus infections with a highly interdisciplinary approach. The core idea was to bring together scientists and students from various disciplines such as structural biology, cell biology, virology and chemistry to advance research by an interdisciplinary means. In 2018, ViroCarb hosted the 1st International Symposium on Glycovirology in Schöntal, Germany, with a similar aim. Scientists from various disciplines gathered to discuss their area of study, present recent findings, establish or strengthen collaborations, and mentor the next generation of glycovirologists through formal presentations and informal discussions. The secluded meeting at the monastery of Schöntal gave ample time for in-depth discussions. On behalf of ViroCarb, this report summarizes the reports and highlights advances in the field.
Collapse
|
19
|
Inoue T, Zhang P, Zhang W, Goodner-Bingham K, Dupzyk A, DiMaio D, Tsai B. γ-Secretase promotes membrane insertion of the human papillomavirus L2 capsid protein during virus infection. J Cell Biol 2018; 217:3545-3559. [PMID: 30006461 PMCID: PMC6168257 DOI: 10.1083/jcb.201804171] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/05/2018] [Accepted: 06/25/2018] [Indexed: 01/02/2023] Open
Abstract
Despite their importance as human pathogens, entry of human papillomaviruses (HPVs) into cells is poorly understood. The transmembrane protease γ-secretase executes a crucial function during the early stages of HPV infection, but the role of γ-secretase in infection and the identity of its critical substrate are unknown. Here we demonstrate that γ-secretase harbors a previously uncharacterized chaperone function, promoting low pH-dependent insertion of the HPV L2 capsid protein into endosomal membranes. Upon membrane insertion, L2 recruits the cytosolic retromer, which enables the L2 viral genome complex to enter the retrograde transport pathway and traffic to the Golgi en route for infection. Although a small fraction of membrane-inserted L2 is also cleaved by γ-secretase, this proteolytic event appears dispensable for HPV infection. Our findings demonstrate that γ-secretase is endowed with an activity that can promote membrane insertion of L2, thereby targeting the virus to the productive infectious pathway.
Collapse
Affiliation(s)
- Takamasa Inoue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| | - Pengwei Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT
| | - Wei Zhang
- Department of Genetics, Yale School of Medicine, New Haven, CT
| | | | - Allison Dupzyk
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI
| | - Daniel DiMaio
- Department of Genetics, Yale School of Medicine, New Haven, CT
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT
- Department of Molecular Biophysics and Biochemistry, Yale School of Medicine, New Haven, CT
- Yale Cancer Center, New Haven, CT
| | - Billy Tsai
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI
| |
Collapse
|
20
|
Non-human papillomaviruses for gene delivery in vitro and in vivo. PLoS One 2018; 13:e0198996. [PMID: 29912929 PMCID: PMC6005490 DOI: 10.1371/journal.pone.0198996] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/18/2018] [Indexed: 12/17/2022] Open
Abstract
Papillomavirus capsids are known to have the ability to package DNA plasmids and deliver them both in vitro and in vivo. Of all known papillomavirus types, human papillomaviruses (HPVs) are by far the most intensely studied. Although HPVs work well as gene transfer vectors, their use is limited as most individuals are exposed to this virus either through a HPV vaccination or natural infection. To circumvent these constraints, we produced pseudovirions (PsVs) of ten non-human papillomavirus types and tested their transduction efficiencies in vitro. PsVs based on Macaca fascicularis papillomavirus-11 and Puma concolor papillomavirus-1 were further tested in vivo. Intramuscular transduction by PsVs led to months-long expression of a reporter plasmid, indicating that PsVs have potential as gene delivery vectors.
Collapse
|
21
|
Extracellular Conformational Changes in the Capsid of Human Papillomaviruses Contribute to Asynchronous Uptake into Host Cells. J Virol 2018; 92:JVI.02106-17. [PMID: 29593032 DOI: 10.1128/jvi.02106-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 03/17/2018] [Indexed: 11/20/2022] Open
Abstract
Human papillomavirus 16 (HPV16) is the leading cause of cervical cancer. For initial infection, HPV16 utilizes a novel endocytic pathway for host cell entry. Unique among viruses, uptake occurs asynchronously over a protracted period of time, with half-times between 9 and 12 h. To trigger endocytic uptake, the virus particles need to undergo a series of structural modifications after initial binding to heparan sulfate proteoglycans (HSPGs). These changes involve proteolytic cleavage of the major capsid protein L1 by kallikrein-8 (KLK8), exposure of the N terminus of the minor capsid protein L2 by cyclophilins, and cleavage of this N terminus by furin. Overall, the structural changes are thought to facilitate the engagement of an elusive secondary receptor for internalization. Here, we addressed whether structural changes are the rate-limiting steps during infectious internalization of HPV16 by using structurally primed HPV16 particles. Our findings indicate that the structural modifications mediated by cyclophilins and furin, which lead to exposure and cleavage, respectively, of the L2 N terminus contribute to the slow and asynchronous internalization kinetics, whereas conformational changes elicited by HSPG binding and KLK8 cleavage did not. However, these structural modifications accounted for only 30 to 50% of the delay in internalization. Therefore, we propose that limited internalization receptor availability for engagement of HPV16 causes slow and asynchronous internalization in addition to rate-limiting structural changes in the viral capsid.IMPORTANCE HPVs are the main cause of anogenital cancers. Their unique biology is linked to the differentiation program of skin or mucosa. Here, we analyzed another unique aspect of HPV infections using the prototype HPV16. After initial cell binding, HPVs display an unusually protracted residence time on the plasma membrane prior to asynchronous uptake. As viruses typically do not expose themselves to host immune sensing, we analyzed the underlying reasons for this unusual behavior. This study provides evidence that both extracellular structural modifications and possibly a limited availability of the internalization receptor contribute to the slow internalization process of the virus. These findings indicate that perhaps a unique niche for initial infection that could allow for rapid infection exists. In addition, our results may help to develop novel, preventive antiviral measures.
Collapse
|
22
|
Superinfection Exclusion between Two High-Risk Human Papillomavirus Types during a Coinfection. J Virol 2018; 92:JVI.01993-17. [PMID: 29437958 DOI: 10.1128/jvi.01993-17] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 01/25/2018] [Indexed: 12/12/2022] Open
Abstract
Superinfection exclusion is a common phenomenon whereby a single cell is unable to be infected by two types of the same pathogen. Superinfection exclusion has been described for various viruses, including vaccinia virus, measles virus, hepatitis C virus, influenza A virus, and human immunodeficiency virus. Additionally, the mechanism of exclusion has been observed at various steps of the viral life cycle, including attachment, entry, viral genomic replication, transcription, and exocytosis. Human papillomavirus (HPV) is the causative agent of cervical cancer. Recent epidemiological studies indicate that up to 50% women who are HPV positive (HPV+) are infected with more than one HPV type. However, no mechanism of superinfection exclusion has ever been identified for HPV. Here, we show that superinfection exclusion exists during a HPV coinfection and that it occurs on the cell surface during the attachment/entry phase of the viral life cycle. Additionally, we are able to show that the minor capsid protein L2 plays a role in this exclusion. This study shows, for the first time, that superinfection exclusion occurs during HPV coinfections and describes a potential molecular mechanism through which it occurs.IMPORTANCE Superinfection exclusion is a phenomenon whereby one cell is unable to be infected by multiple related pathogens. This phenomenon has been described for many viruses and has been shown to occur at various points in the viral life cycle. HPV is the causative agent of cervical cancer and is involved in other anogenital and oropharyngeal cancers. Recent epidemiological research has shown that up to 50% of HPV-positive individuals harbor more than one type of HPV. We investigated the interaction between two high-risk HPV types, HPV16 and HPV18, during a coinfection. We present data showing that HPV16 is able to block or exclude HPV18 on the cell surface during a coinfection. This exclusion is due in part to differences in the HPV minor capsid protein L2. This report provides, for the first time, evidence of superinfection exclusion for HPV and leads to a better understanding of the complex interactions between multiple HPV types during coinfections.
Collapse
|
23
|
A central region in the minor capsid protein of papillomaviruses facilitates viral genome tethering and membrane penetration for mitotic nuclear entry. PLoS Pathog 2017; 13:e1006308. [PMID: 28464022 PMCID: PMC5412989 DOI: 10.1371/journal.ppat.1006308] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Accepted: 03/22/2017] [Indexed: 12/18/2022] Open
Abstract
Incoming papillomaviruses (PVs) depend on mitotic nuclear envelope breakdown to gain initial access to the nucleus for viral transcription and replication. In our previous work, we hypothesized that the minor capsid protein L2 of PVs tethers the incoming vDNA to mitotic chromosomes to direct them into the nascent nuclei. To re-evaluate how dynamic L2 recruitment to cellular chromosomes occurs specifically during prometaphase, we developed a quantitative, microscopy-based assay for measuring the degree of chromosome recruitment of L2-EGFP. Analyzing various HPV16 L2 truncation-mutants revealed a central chromosome-binding region (CBR) of 147 amino acids that confers binding to mitotic chromosomes. Specific mutations of conserved motifs (IVAL286AAAA, RR302/5AA, and RTR313EEE) within the CBR interfered with chromosomal binding. Moreover, assembly-competent HPV16 containing the chromosome-binding deficient L2(RTR313EEE) or L2(IVAL286AAAA) were inhibited for infection despite their ability to be transported to intracellular compartments. Since vDNA and L2 were not associated with mitotic chromosomes either, the infectivity was likely impaired by a defect in tethering of the vDNA to mitotic chromosomes. However, L2 mutations that abrogated chromatin association also compromised translocation of L2 across membranes of intracellular organelles. Thus, chromatin recruitment of L2 may in itself be a requirement for successful penetration of the limiting membrane thereby linking both processes mechanistically. Furthermore, we demonstrate that the association of L2 with mitotic chromosomes is conserved among the alpha, beta, gamma, and iota genera of Papillomaviridae. However, different binding patterns point to a certain variance amongst the different genera. Overall, our data suggest a common strategy among various PVs, in which a central region of L2 mediates tethering of vDNA to mitotic chromosomes during cell division thereby coordinating membrane translocation and delivery to daughter nuclei. Papillomaviruses can cause carcinogenic malignancies such as cervical cancer. Like most DNA viruses, papillomaviruses must deliver their genome to the cell nucleus during initial infection, where it is expressed and replicated. However, papillomaviruses make use of unconventional mechanisms for genome delivery. They reside on the cell surface for protracted, hour-long times, before they are taken up by a novel endocytic mechanism. Moreover, they are delivered to the trans-Golgi-network by non-canonical endosomal trafficking prior to nuclear delivery. For entry into the nucleus, papillomaviruses access the nuclear space after nuclear envelope breakdown during mitosis unlike most other intranuclear viruses. The detailed mechanism how the viral genome is directed to nascent nuclei during mitosis remains elusive. Our previous work suggested that the minor capsid protein L2 may tether the incoming viral genome to mitotic chromosomes to direct it to the nascent nuclei. This work identifies a conserved central region in L2 protein to be necessary and sufficient for tethering. Moreover, it demonstrates that this mechanism is conserved across different papillomavirus genera. Importantly, this report also provides evidence that the processes of nuclear import by tethering and membrane penetration are mechanistically linked.
Collapse
|
24
|
Calton CM, Bronnimann MP, Manson AR, Li S, Chapman JA, Suarez-Berumen M, Williamson TR, Molugu SK, Bernal RA, Campos SK. Translocation of the papillomavirus L2/vDNA complex across the limiting membrane requires the onset of mitosis. PLoS Pathog 2017; 13:e1006200. [PMID: 28463988 PMCID: PMC5412990 DOI: 10.1371/journal.ppat.1006200] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 01/25/2017] [Indexed: 11/20/2022] Open
Abstract
The human papillomavirus type 16 (HPV16) L2 protein acts as a chaperone to ensure that the viral genome (vDNA) traffics from endosomes to the trans-Golgi network (TGN) and eventually the nucleus, where HPV replication occurs. En route to the nucleus, the L2/vDNA complex must translocate across limiting intracellular membranes. The details of this critical process remain poorly characterized. We have developed a system based on subcellular compartmentalization of the enzyme BirA and its cognate substrate to detect membrane translocation of L2-BirA from incoming virions. We find that L2 translocation requires transport to the TGN and is strictly dependent on entry into mitosis, coinciding with mitotic entry in synchronized cells. Cell cycle arrest causes retention of L2/vDNA at the TGN; only release and progression past G2/M enables translocation across the limiting membrane and subsequent infection. Microscopy of EdU-labeled vDNA reveals a rapid and dramatic shift in vDNA localization during early mitosis. At late G2/early prophase vDNA egresses from the TGN to a pericentriolar location, accumulating there through prometaphase where it begins to associate with condensed chromosomes. By metaphase and throughout anaphase the vDNA is seen bound to the mitotic chromosomes, ensuring distribution into both daughter nuclei. Mutations in a newly defined chromatin binding region of L2 potently blocked translocation, suggesting that translocation is dependent on chromatin binding during prometaphase. This represents the first time a virus has been shown to functionally couple the penetration of limiting membranes to cellular mitosis, explaining in part the tropism of HPV for mitotic basal keratinocytes.
Collapse
Affiliation(s)
- Christine M. Calton
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Matthew P. Bronnimann
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
| | - Ariana R. Manson
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Shuaizhi Li
- Department of Cellular & Molecular Medicine, University of Arizona, Tucson, Arizona, United States of America
| | - Janice A. Chapman
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| | - Marcela Suarez-Berumen
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Tatum R. Williamson
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
| | - Sudheer K. Molugu
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Ricardo A. Bernal
- Department of Chemistry, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Samuel K. Campos
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
- Department of Immunobiology, University of Arizona, Tucson, Arizona, United States of America
- Department of Molecular & Cellular Biology, University of Arizona, Tucson, Arizona, United States of America
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
25
|
Aksoy P, Gottschalk EY, Meneses PI. HPV entry into cells. MUTATION RESEARCH. REVIEWS IN MUTATION RESEARCH 2017; 772:13-22. [PMID: 28528686 PMCID: PMC5443120 DOI: 10.1016/j.mrrev.2016.09.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 08/22/2016] [Accepted: 09/16/2016] [Indexed: 12/20/2022]
Abstract
Human papillomavirus (HPV) is a sexually transmitted virus responsible for the development of cervical cancer, anal cancer, head and throat cancers, as well as genital area warts. A major focus of current HPV research is on preventing the virus from entering a cell and transferring its genetic material to the nucleus, thus potentially preventing the development of cancer. Although the available HPV vaccines are extremely successful, approximately 15 additional cancer-causing HPVs have been identified that the vaccines do not protect against. Therefore, roughly 150,000 cancer cases will not be prevented annually with the current vaccines. Research efforts focused on the basic cell biology of HPV infection have a goal of identifying common infectious events that may lead to inexpensive vaccines or anti-virals to prevent infection by most, if not all, HPVs. In this review we attempt to summarize what is known regarding the process of HPV binding, entry, and intracellular trafficking.
Collapse
Affiliation(s)
- Pinar Aksoy
- Department of Biological Sciences, Fordham University, Bronx, NY, USA
| | | | | |
Collapse
|
26
|
Weber C, Berberich E, von Rhein C, Henß L, Hildt E, Schnierle BS. Identification of Functional Determinants in the Chikungunya Virus E2 Protein. PLoS Negl Trop Dis 2017; 11:e0005318. [PMID: 28114368 PMCID: PMC5289616 DOI: 10.1371/journal.pntd.0005318] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 02/02/2017] [Accepted: 01/10/2017] [Indexed: 11/20/2022] Open
Abstract
Background Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that causes high fever, rash, and recurrent arthritis in humans. It has efficiently adapted to Aedes albopictus, which also inhabits temperate regions, including Europe and the United States of America. In the past, CHIKV has mainly affected developing countries, but has recently caused large outbreaks in the Caribbean and Latin America. No treatment or licensed CHIKV vaccine exists. Methodology/Principal Findings Here, we have identified determinants in the CHIKV cell-attachment protein E2 that facilitate cell binding. The extracellular part of the E2 gene is subdivided into the three domains, A, B, and C. These domains were expressed in E. coli and as Fc-fusion proteins generated from HEK293T cells and used for cell-binding assays. Domains A and B bound to all cells tested, independently of their permissiveness to CHIKV infection. Domain C did not bind to cells at all. Furthermore, CHIKV cell entry was promoted by cell-surface glycosaminoglycans (GAGs) and domain B interacted exclusively with GAG-expressing cells. Domain A also bound, although only moderately, to GAG-deficient cells. Soluble GAGs were able to inhibit CHIKV infection up to 90%; however, they enhanced the transduction rate of CHIKV Env pseudotyped vectors in GAG-negative cells. Conclusion/Significance These data imply that CHIKV uses at least two mechanisms to enter cells, one GAG-dependent, via initial attachment through domain B, and the other GAG-independent, via attachment of domain A. These data give indications that CHIKV uses multiple mechanisms to enter cells and shows the potential of GAGs as lead structures for developing antiviral drugs. The chikungunya virus (CHIKV) glycoprotein E2 mediates cell attachment and consists of three domains A, B and C. Since the cell entry process of CHIKV is not understood in detail, we analyzed the binding properties of the three E2 domains with proteins expressed in E. coli or as Fc-fusion proteins and the role of glycosaminoglycans (GAGs) on E2 cell binding and CHIKV entry. The two surface-exposed E2 domains, A and B, both bound to cells and domain B bound only to cells expressing GAGs. Domain A bound additionally to GAG-deficient cells and domain C did not bind to cells. CHIKV-pseudotyped lentiviral vector and CHIKV entry were enhanced in cells expressing GAGs. Our results suggest that CHIKV uses at least two entry mechanisms, one GAG-dependent, via attachment through E2 domain B, and the other GAG-independent, via binding of domain A. These data give indications that CHIKV uses multiple mechanisms to enter cells and shows the potential of GAGs as lead structures for developing antiviral drugs. In addition, it shows that domain A and B might constitute good targets for vaccine development.
Collapse
Affiliation(s)
- Christopher Weber
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
| | - Eva Berberich
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
| | - Christine von Rhein
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
| | - Lisa Henß
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
| | - Eberhard Hildt
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
| | - Barbara S. Schnierle
- Paul-Ehrlich-Institut, Department of Virology, Paul-Ehrlich Strasse, Langen, Germany
- * E-mail:
| |
Collapse
|
27
|
The CD63-Syntenin-1 Complex Controls Post-Endocytic Trafficking of Oncogenic Human Papillomaviruses. Sci Rep 2016; 6:32337. [PMID: 27578500 PMCID: PMC5006017 DOI: 10.1038/srep32337] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 08/02/2016] [Indexed: 12/31/2022] Open
Abstract
Human papillomaviruses enter host cells via a clathrin-independent endocytic pathway involving tetraspanin proteins. However, post-endocytic trafficking required for virus capsid disassembly remains unclear. Here we demonstrate that the early trafficking pathway of internalised HPV particles involves tetraspanin CD63, syntenin-1 and ESCRT-associated adaptor protein ALIX. Following internalisation, viral particles are found in CD63-positive endosomes recruiting syntenin-1, a CD63-interacting adaptor protein. Electron microscopy and immunofluorescence experiments indicate that the CD63-syntenin-1 complex controls delivery of internalised viral particles to multivesicular endosomes. Accordingly, infectivity of high-risk HPV types 16, 18 and 31 as well as disassembly and post-uncoating processing of viral particles was markedly suppressed in CD63 or syntenin-1 depleted cells. Our analyses also present the syntenin-1 interacting protein ALIX as critical for HPV infection and CD63-syntenin-1-ALIX complex formation as a prerequisite for intracellular transport enabling viral capsid disassembly. Thus, our results identify the CD63-syntenin-1-ALIX complex as a key regulatory component in post-endocytic HPV trafficking.
Collapse
|
28
|
Furin Cleavage of L2 during Papillomavirus Infection: Minimal Dependence on Cyclophilins. J Virol 2016; 90:6224-6234. [PMID: 27122588 DOI: 10.1128/jvi.00038-16] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/21/2016] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Despite an abundance of evidence supporting an important role for the cleavage of minor capsid protein L2 by cellular furin, direct cleavage of capsid-associated L2 during human papillomavirus 16 (HPV16) infection remains poorly characterized. The conserved cleavage site, close to the L2 N terminus, confounds observation and quantification of the small cleavage product by SDS-PAGE. To overcome this difficulty, we increased the size shift by fusing a compact protein domain, the Propionibacterium shermanii transcarboxylase domain (PSTCD), to the N terminus of L2. The infectious PSTCD-L2 virus displayed an appreciable L2 size shift during infection of HaCaT keratinocytes. Cleavage under standard cell culture conditions rarely exceeded 35% of total L2. Cleavage levels were enhanced by the addition of exogenous furin, and the absolute levels of infection correlated to the level of L2 cleavage. Cleavage occurred on both the HaCaT cell surface and extracellular matrix (ECM). Contrary to current models, experiments on the involvement of cyclophilins revealed little, if any, role for these cellular enzymes in the modulation of furin cleavage. HPV16 L2 contains two consensus cleavage sites, Arg5 (2RHKR5) and Arg12 (9RTKR12). Mutant PSTCD-L2 viruses demonstrated that although furin can cleave either site, cleavage must occur at Arg12, as cleavage at Arg5 alone is insufficient for successful infection. Mutation of the conserved cysteine residues revealed that the Cys22-Cys28 disulfide bridge is not required for cleavage. The PSTCD-L2 virus or similar N-terminal fusions will be valuable tools to study additional cellular and viral determinants of furin cleavage. IMPORTANCE Furin cleavage of minor capsid protein L2 during papillomavirus infection has been difficult to directly visualize and quantify, confounding efforts to study this important step of HPV infection. Fusion of a small protein domain to the N terminus greatly facilitates direct visualization of the cleavage product, revealing important characteristics of this critical process. Contrary to the current model, we found that cleavage is largely independent of cyclophilins, suggesting that cyclophilins act either in parallel to or downstream of furin to trigger exposure of a conserved N-terminal L2 epitope (RG-1) during infection. Based on this finding, we strongly caution against using L2 RG-1 epitope exposure as a convenient but indirect proxy of furin cleavage.
Collapse
|
29
|
Abstract
Glycosaminoglycans (GAGs) are complex linear polysaccharides expressed in intracellular compartments, at the cell surface, and in the extracellular environment where they interact with various molecules to regulate many cellular processes implicated in health and disease. Subversion of GAGs is a pathogenic strategy shared by a wide variety of microbial pathogens, including viruses, bacteria, parasites, and fungi. Pathogens use GAGs at virtually every major portals of entry to promote their attachment and invasion of host cells, movement from one cell to another, and to protect themselves from immune attack. Pathogens co-opt fundamental activities of GAGs to accomplish these tasks. This ingenious strategy to subvert essential activities of GAGs likely prevented host organisms from deleting or inactivating these mechanisms during their evolution. The goal of this review is to provide a mechanistic overview of our current understanding of how microbes subvert GAGs at major steps of pathogenesis, using select GAG-pathogen interactions as representative examples.
Collapse
Affiliation(s)
- Rafael S Aquino
- Division of Respiratory Diseases and 2Division of Newborn Medicine, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Pyong Woo Park
- Division of Respiratory Diseases Children's Hospital, Harvard Medical School, Boston, MA 02115, USA and Division of Newborn Medicine, Children's Hospital, Harvard Medical School, Boston, MA 02115, USA,
| |
Collapse
|
30
|
Cruz L, Biryukov J, Conway MJ, Meyers C. Cleavage of the HPV16 Minor Capsid Protein L2 during Virion Morphogenesis Ablates the Requirement for Cellular Furin during De Novo Infection. Viruses 2015; 7:5813-30. [PMID: 26569287 PMCID: PMC4664983 DOI: 10.3390/v7112910] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 10/23/2015] [Accepted: 10/29/2015] [Indexed: 12/17/2022] Open
Abstract
Infections by high-risk human papillomaviruses (HPV) are the causative agents for the development of cervical cancer. As with other non-enveloped viruses, HPVs are taken up by the cell through endocytosis following primary attachment to the host cell. Through studies using recombinant pseudovirus particles (PsV), many host cellular proteins have been implicated in the process. The proprotein convertase furin has been demonstrated to cleave the minor capsid protein, L2, post-attachment to host cells and is required for infectious entry by HPV16 PsV. In contrast, using biochemical inhibition by a furin inhibitor and furin-negative cells, we show that tissue-derived HPV16 native virus (NV) initiates infection independent of cellular furin. We show that HPV16 L2 is cleaved during virion morphogenesis in differentiated tissue. In addition, HPV45 is also not dependent on cellular furin, but two other alpha papillomaviruses, HPV18 and HPV31, are dependent on the activity of cellular furin for infection.
Collapse
Affiliation(s)
- Linda Cruz
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Jennifer Biryukov
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Michael J Conway
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| | - Craig Meyers
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033, USA.
| |
Collapse
|
31
|
Abstract
Human papillomavirus (HPV) is the most prevalent sexually transmitted disease in the United States and can cause cancer with persistent infection. The most common cancer caused by HPV is cervical carcinoma with an average of 12,000 cases reported every year in the United States. Worldwide, over 500,000 cases of cervical cancer are reported yearly with over 250,000 deaths attributed to the disease. Although much is known about the serious health risks associated with HPV infection, there is still much to be discovered about how HPV binds and enters target cells. Understanding is required on how HPV infections will lead to strategies and therapies for reducing the number of infections and HPV-related diseases, including cancers. The HPV viral particle is composed of 2 viral proteins, L1 and L2. Data suggest that binding of the viral capsid to cells is dependent on the L1 protein. We hypothesize that this initial binding to a heparan sulfate is composed of 2 independent events: the first results in a structural change that exposes a hidden portion of the L1 protein leading to a second binding event on the heparan sulfate. Our experiments tested if this "hidden" portion of L1 is necessary for infection and explored the nature of this binding. We generated a peptide with the sequence of the "hidden" portion of L1. Infection of HaCaT cells in the presence of this peptide is highly reduced. Our results suggest that the binding of the L1 C-terminal domain is dependent on amino acid sequence and is necessary for infection.
Collapse
|
32
|
Surviladze Z, Sterkand RT, Ozbun MA. Interaction of human papillomavirus type 16 particles with heparan sulfate and syndecan-1 molecules in the keratinocyte extracellular matrix plays an active role in infection. J Gen Virol 2015; 96:2232-2241. [PMID: 26289843 PMCID: PMC4681067 DOI: 10.1099/vir.0.000147] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 04/13/2015] [Indexed: 12/18/2022] Open
Abstract
Oncogenic human papillomaviruses (HPVs) attach predominantly to extracellular matrix (ECM) components during infection of cultured keratinocytes and in the rodent vaginal challenge model in vivo. However, the mechanism of virion transfer from the ECM to receptors that mediate entry into host cells has not been determined. In this work we strove to assess the role of heparan sulfate (HS) chains in HPV16 binding to the ECM and determine how HPV16 release from the ECM is regulated. We also assessed the extent to which capsids released from the ECM are infectious. We show that a large fraction of HPV16 particles binds to the ECM via HS chains, and that syndecan-1 (snd-1) molecules present in the ECM are involved in virus binding. Inhibiting the normal processing of snd-1 and HS molecules via matrix metalloproteinases and heparanase dramatically reduces virus release from the ECM, cellular uptake and infection. Conversely, exogenous heparinase activates each of these processes. We confirm that HPV16 released from the ECM is infectious in keratinocytes. Use of a specific inhibitor shows furin is not involved in HPV16 release from ECM attachment factors and corroborates other studies showing only the intracellular activity of furin is responsible for modulating HPV infectivity. These data suggest that our recently proposed model, describing the action of HS proteoglycan processing enzymes in releasing HPV16 from the cell surface in complex with the attachment factor snd-1, is also relevant to the release of HPV16 particles from the ECM to promote efficient infection of keratinocytes.
Collapse
Affiliation(s)
- Zurab Surviladze
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | - Rosa T. Sterkand
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| | - Michelle A. Ozbun
- Department of Molecular Genetics & Microbiology, University of New Mexico School of Medicine, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
33
|
Song D, Li H, Li H, Dai J. Effect of human papillomavirus infection on the immune system and its role in the course of cervical cancer. Oncol Lett 2015; 10:600-606. [PMID: 26622540 DOI: 10.3892/ol.2015.3295] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 04/13/2015] [Indexed: 02/07/2023] Open
Abstract
Human papillomavirus (HPV) is widely known as a cause of cervical intraepithelial neoplasia (CIN) and cervical cancer. The mechanisms involved have been studied by numerous studies. The integration of the virus genome into the host cells results in the abnormal regulation of cell cycle control. HPV can also induce immune evasion of the infected cells, which enable the virus to be undetectable for long periods of time. The induction of immunotolerance of the host's immune system by the persistent infection of HPV is one of the most important mechanisms for cervical lesions. The present review elaborates on the roles of several types of immune cells, such as macrophages and natural killer cells, which are classified as innate immune cells, and dendritic cells (DCs), cluster of differentiation (CD)4+/CD8+ T cells and regulatory T cells, which are classified as adaptive immune cells. HPV infection could effect the differentiation of these immune cells in a unique way, resulting in the host's immune tolerance to the infection. The immune system modifications induced by HPV infection include tumor-associated macrophage differentiation, a compromised cellular immune response, an abnormal imbalance between type 1 T-helper cells (Th1) and Th2 cells, regulatory T cell infiltration, and downregulated DC activation and maturation. To date, numerous types of preventative vaccines have been created to slow down carcinogenesis. Immune response activation-based therapeutic vaccine is becoming more and more attractive for the treatment of HPV-associated diseases.
Collapse
Affiliation(s)
- Dan Song
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Hong Li
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Haibo Li
- Center for Reproduction and Genetics, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Jianrong Dai
- Department of Gynecology, Suzhou Municipal Hospital, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu 215002, P.R. China
| |
Collapse
|
34
|
Kallikrein-8 Proteolytically Processes Human Papillomaviruses in the Extracellular Space To Facilitate Entry into Host Cells. J Virol 2015; 89:7038-52. [PMID: 25926655 DOI: 10.1128/jvi.00234-15] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/20/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED The entry of human papillomaviruses into host cells is a complex process. It involves conformational changes at the cell surface, receptor switching, internalization by a novel endocytic mechanism, uncoating in endosomes, trafficking of a subviral complex to the Golgi complex, and nuclear entry during mitosis. Here, we addressed how the stabilizing contacts in the capsid of human papillomavirus 16 (HPV16) may be reversed to allow uncoating of the viral genome. Using biochemical and cell-biological analyses, we determined that the major capsid protein L1 underwent proteolytic cleavage during entry. In addition to a dispensable cathepsin-mediated proteolysis that occurred likely after removal of capsomers from the subviral complex in endosomes, at least two further proteolytic cleavages of L1 were observed, one of which was independent of the low-pH environment of endosomes. This cleavage occurred extracellularly. Further analysis showed that the responsible protease was the secreted trypsin-like serine protease kallikrein-8 (KLK8) involved in epidermal homeostasis and wound healing. Required for infection, the cleavage was facilitated by prior interaction of viral particles with heparan sulfate proteoglycans. KLK8-mediated cleavage was crucial for further conformational changes exposing an important epitope of the minor capsid protein L2. Occurring independently of cyclophilins and of furin that mediate L2 exposure, KLK8-mediated cleavage of L1 likely facilitated access to L2, located in the capsid lumen, and potentially uncoating. Since HPV6 and HPV18 also required KLK8 for entry, we propose that the KLK8-dependent entry step is conserved. IMPORTANCE Our analysis of the proteolytic processing of incoming HPV16, an etiological agent of cervical cancer, demonstrated that the capsid is cleaved extracellularly by a serine protease active during wound healing and that this cleavage was crucial for infection. The cleavage of L1 is one of at least four structural alterations that prime the virus extracellularly for receptor switching, internalization, and possibly uncoating. This step was also important for HPV6 and HPV18, which may suggest that it is conserved among the papillomaviruses. This study advances the understanding of how HPV16 initially infects cells, strengthens the notion that wounding facilitates infection of epidermal tissue, and may help the development of antiviral measures.
Collapse
|
35
|
The HPV16 and MusPV1 papillomaviruses initially interact with distinct host components on the basement membrane. Virology 2015; 481:79-94. [PMID: 25771496 DOI: 10.1016/j.virol.2015.02.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 01/12/2015] [Accepted: 02/13/2015] [Indexed: 11/24/2022]
Abstract
To understand and compare the mechanisms of murine and human PV infection, we examined pseudovirion binding and infection of the newly described MusPV1 using the murine cervicovaginal challenge model. These analyses revealed primary tissue interactions distinct from those previously described for HPV16. Unlike HPV16, MusPV1 bound basement membrane (BM) in an HSPG-independent manner. Nevertheless, subsequent HSPG interactions were critical. L2 antibodies or low doses of VLP antibodies, sufficient to prevent infection, did not lead to disassociation of the MusPV1 pseudovirions from the BM, in contrast to previous findings with HPV16. Similarly, furin inhibition did not lead to loss of MusPV1 from the BM. Therefore, phylogenetically distant PV types differ in their initial interactions with host attachment factors, but initiate their lifecycle on the acellular BM. Despite these differences, these distantly related PV types displayed similar intracellular trafficking patterns and susceptibilities to biochemical inhibition of infection.
Collapse
|
36
|
Human papillomavirus species-specific interaction with the basement membrane-resident non-heparan sulfate receptor. Viruses 2014; 6:4856-79. [PMID: 25490765 PMCID: PMC4276933 DOI: 10.3390/v6124856] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 11/22/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
Using a cell culture model where virus is bound to the extracellular matrix (ECM) prior to cell surface binding, we determined that human papillomavirus type 16 (HPV16) utilizes ECM resident laminin (LN) 332 as an attachment receptor for infectious entry. In presence of LN332, soluble heparin can function as ligand activator rather than competitive inhibitor of HPV16 infection. We also show that the ability to use LN332 binding as a productive attachment step for infectious entry is not conserved amongst HPV types. In the alpha genus, species 9 members (HPV16) attach to ECM via LN332, while members of species 7 (HPV18) are completely inhibited by heparin pre-incubation due to an inability to use LN332. Since HPV species 7 and 9 are preferentially associated with adenocarcinoma and squamous cell carcinoma of the cervix, respectively, our data provide first evidence that pre-entry events may contribute to the anatomical-site preference of HPV species.
Collapse
|
37
|
Wang JW, Jagu S, Wang C, Kitchener HC, Daayana S, Stern PL, Pang S, Day PM, Huh WK, Roden RBS. Measurement of neutralizing serum antibodies of patients vaccinated with human papillomavirus L1 or L2-based immunogens using furin-cleaved HPV Pseudovirions. PLoS One 2014; 9:e101576. [PMID: 24999962 PMCID: PMC4084990 DOI: 10.1371/journal.pone.0101576] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 06/09/2014] [Indexed: 11/26/2022] Open
Abstract
Antibodies specific for neutralizing epitopes in either Human papillomavirus (HPV) capsid protein L1 or L2 can mediate protection from viral challenge and thus their accurate and sensitive measurement at high throughput is likely informative for monitoring response to prophylactic vaccination. Here we compare measurement of L1 and L2-specific neutralizing antibodies in human sera using the standard Pseudovirion-Based Neutralization Assay (L1-PBNA) with the newer Furin-Cleaved Pseudovirion-Based Neutralization Assay (FC-PBNA), a modification of the L1-PBNA intended to improve sensitivity towards L2-specific neutralizing antibodies without compromising assay of L1-specific responses. For detection of L1-specific neutralizing antibodies in human sera, the FC- PBNA and L1-PBNA assays showed similar sensitivity and a high level of correlation using WHO standard sera (n = 2), and sera from patients vaccinated with Gardasil (n = 30) or an experimental human papillomavirus type 16 (HPV16) L1 VLP vaccine (n = 70). The detection of L1-specific cross-neutralizing antibodies in these sera using pseudovirions of types phylogenetically-related to those targeted by the L1 virus-like particle (VLP) vaccines was also consistent between the two assays. However, for sera from patients (n = 17) vaccinated with an L2-based immunogen (TA-CIN), the FC-PBNA was more sensitive than the L1-PBNA in detecting L2-specific neutralizing antibodies. Further, the neutralizing antibody titers measured with the FC-PBNA correlated with those determined with the L2-PBNA, another modification of the L1-PBNA that spacio-temporally separates primary and secondary receptor engagement, as well as the protective titers measured using passive transfer studies in the murine genital-challenge model. In sum, the FC-PBNA provided sensitive measurement for both L1 VLP and L2-specific neutralizing antibody in human sera. Vaccination with TA-CIN elicits weak cross-protective antibody in a subset of patients, suggesting the need for an adjuvant.
Collapse
MESH Headings
- Antibodies, Neutralizing/blood
- Antibodies, Neutralizing/immunology
- Antibodies, Viral/blood
- Antibodies, Viral/immunology
- Antibody Specificity
- Capsid Proteins/immunology
- Capsid Proteins/metabolism
- Furin/metabolism
- Human Papillomavirus Recombinant Vaccine Quadrivalent, Types 6, 11, 16, 18
- Humans
- Neutralization Tests/methods
- Neutralization Tests/standards
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Papillomaviridae/immunology
- Papillomavirus Vaccines/immunology
- Papillomavirus Vaccines/metabolism
- Proteolysis
- Reference Standards
- Vaccination
- Virion/immunology
Collapse
Affiliation(s)
- Joshua W. Wang
- Departments of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Subhashini Jagu
- Departments of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Chenguang Wang
- Department of Biostatistics, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Henry C. Kitchener
- Woman's Cancer Centre, St Mary's Hospital, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Sai Daayana
- Woman's Cancer Centre, St Mary's Hospital, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Peter L. Stern
- Paterson Building, Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom
| | - Susana Pang
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Patricia M. Day
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, Maryland, United States of America
| | - Warner K. Huh
- Division of Gynecologic Oncology, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Richard B. S. Roden
- Departments of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
38
|
Aydin I, Weber S, Snijder B, Samperio Ventayol P, Kühbacher A, Becker M, Day PM, Schiller JT, Kann M, Pelkmans L, Helenius A, Schelhaas M. Large scale RNAi reveals the requirement of nuclear envelope breakdown for nuclear import of human papillomaviruses. PLoS Pathog 2014; 10:e1004162. [PMID: 24874089 PMCID: PMC4038628 DOI: 10.1371/journal.ppat.1004162] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Accepted: 04/21/2014] [Indexed: 11/24/2022] Open
Abstract
A two-step, high-throughput RNAi silencing screen was used to identify host cell factors required during human papillomavirus type 16 (HPV16) infection. Analysis of validated hits implicated a cluster of mitotic genes and revealed a previously undetermined mechanism for import of the viral DNA (vDNA) into the nucleus. In interphase cells, viruses were endocytosed, routed to the perinuclear area, and uncoated, but the vDNA failed to be imported into the nucleus. Upon nuclear envelope perforation in interphase cells HPV16 infection occured. During mitosis, the vDNA and L2 associated with host cell chromatin on the metaphase plate. Hence, we propose that HPV16 requires nuclear envelope breakdown during mitosis for access of the vDNA to the nucleoplasm. The results accentuate the value of genes found by RNAi screens for investigation of viral infections. The list of cell functions required during HPV16 infection will, moreover, provide a resource for future virus-host cell interaction studies. Certain human papillomaviruses (HPV) are the etiological cause of cervical cancers and other epithelial tumors. Recent advances in the development of anti-HPV vaccines and their increasing deployment provide hope for a significant decrease of these cancers in the future. However, many details of the transmission of HPV between infected and uninfected individuals are still not well understood. In particular, the multistep process of entry into host cells is only partially characterized. Here, we used a systematic RNAi silencing approach to identify host cell proteins required during HPV16 infection, the most prevalent cancer-causing HPV. Our results indicated that HPV entry requires cells to divide. Cell division is important, since it removes the barrier between the nucleoplasm and the cytosol, so that the virus can access the nucleus for transcription and replication. Our study provides new mechanistic insights into the entry process of an important pathogen, and explains why it can only infect the stem cells or transiently amplifying cells of human skin or mucosa. This work also highlights a novel nuclear import strategy for DNA viruses, which typically use the nuclear import machinery to access the nucleoplasm for infection, Thus, HPV entry displays another unique aspect uncommon to most other viruses.
Collapse
Affiliation(s)
- Inci Aydin
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, Münster, Germany
| | - Susanne Weber
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, Münster, Germany
| | - Berend Snijder
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Pilar Samperio Ventayol
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, Münster, Germany
| | | | - Miriam Becker
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, Münster, Germany
| | - Patricia M Day
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John T Schiller
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Kann
- Laboratoire de Microbiologie Fondamentale et Pathogénicité, Université Bordeaux Segalen, Bordeaux, France
| | - Lucas Pelkmans
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Ari Helenius
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| | - Mario Schelhaas
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, Münster, Germany
| |
Collapse
|
39
|
Kwak K, Jiang R, Wang JW, Jagu S, Kirnbauer R, Roden RBS. Impact of inhibitors and L2 antibodies upon the infectivity of diverse alpha and beta human papillomavirus types. PLoS One 2014; 9:e97232. [PMID: 24816794 PMCID: PMC4016295 DOI: 10.1371/journal.pone.0097232] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2013] [Accepted: 04/16/2014] [Indexed: 11/19/2022] Open
Abstract
The licensed human papillomavirus (HPV) vaccines elicit type-restricted immunity but do not target cutaneous HPV types of the beta genus that are associated with non-melanoma skin cancer in immune-compromised patients, and it is unclear if these diverse types share a common mechanism of infection. Residues 11-88 of minor capsid protein L2 contain cross-protective epitopes, and vaccination with concatamers of this region derived from as many as eight alpha HPV (L2 α11-88x8) is being developed as an alternative prophylactic vaccine with potentially broader efficacy. There is also interest in developing broadly protective topical microbicides, such as carrageenan or heparin that block HPV receptor interactions, or small molecule inhibitors of infection. Here we have examined several inhibitors of HPV infection and antisera to L2 α11-88x8 for their breadth of activity against infection by 34 HPV types from within both the alpha and beta families using pseudovirions (PsV) carrying a luciferase reporter as surrogates for native virus. We observed that both heparin and carrageenan prevented infection by mucosatropic HPV types, but surprisingly PsV of several epidermotropic alpha4 and beta HPV types exhibited increased infectivity especially at low inhibitor concentrations. Furin and γ-secretase inhibitors and L2 α11-88x8 antiserum blocked infection by all HPV PsV types tested. These findings suggest that the distinct tropism of mucosal and cutaneous HPV may reflect distinct cell surface receptor interactions, but a common uptake mechanism dependent upon furin and γ-secretase proteolytic activities. Carrageenan, which is being tested as a vaginal microbicide, broadly inhibited infection by the high-risk mucosatropic HPV PsV, but not most skin tropic alpha and beta HPV. Vaccination with an L2 multimer derived exclusively from alpha papillomavirus sequences induced antibodies that broadly neutralized PsV of all 34 HPVs from within both the alpha and beta families, suggesting each displays conserved L2 neutralizing epitopes.
Collapse
Affiliation(s)
- Kihyuck Kwak
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Rosie Jiang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Joshua W. Wang
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Subhashini Jagu
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Reinhard Kirnbauer
- Laboratory of Viral Oncology, Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University Vienna (MUW), Vienna, Austria
| | - Richard B. S. Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, Maryland, United States of America
- Department of Oncology, The Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
40
|
The tetraspanin CD151 in papillomavirus infection. Viruses 2014; 6:893-908. [PMID: 24553111 PMCID: PMC3939487 DOI: 10.3390/v6020893] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/11/2014] [Accepted: 02/12/2014] [Indexed: 12/18/2022] Open
Abstract
Human papillomaviruses (HPV) are non-enveloped DNA tumor viruses that infect skin and mucosa. The most oncogenic subtype, HPV16, causes various types of cancer, including cervical, anal, and head and neck cancers. During the multistep process of infection, numerous host proteins are required for the delivery of virus genetic information into the nucleus of target cells. Over the last two decades, many host-cell proteins such as heparan sulfate proteoglycans, integrins, growth factor receptors, actin and the tetraspanin CD151 have been described to be involved in the process of infectious entry of HPV16. Tetraspanins have the ability to organize membrane microdomains and to directly influence the function of associated molecules, including binding of receptors to their ligands, receptor oligomerization and signal transduction. Here, we summarize the current knowledge on CD151, and CD151-associated partners during HPV infection and discuss the underlying mechanisms.
Collapse
|
41
|
Day PM, Schelhaas M. Concepts of papillomavirus entry into host cells. Curr Opin Virol 2014; 4:24-31. [PMID: 24525291 PMCID: PMC3951680 DOI: 10.1016/j.coviro.2013.11.002] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 11/17/2013] [Accepted: 11/19/2013] [Indexed: 12/17/2022]
Abstract
Papillomaviruses enter basal cells of stratified epithelia. Assembly of new virions occurs in infected cells during terminal differentiation. This unique biology is reflected in the mechanism of entry. Extracellularly, the interaction of nonenveloped capsids with several host cell proteins, after binding, results in discrete conformational changes. Asynchronous internalization occurs over several hours by an endocytic mechanism related to, but distinct from macropinocytosis. Intracellular trafficking leads virions through the endosomal system, and from late endosomes to the trans-Golgi-network, before nuclear delivery. Here, we discuss the existing data with the aim to synthesize an integrated model of the stepwise process of entry, thereby highlighting key open questions. Additionally, we relate data from experiments with cultured cells to in vivo results.
Collapse
Affiliation(s)
- Patricia M Day
- Laboratory of Cellular Oncology, National Cancer Institute, National Institutes of Health, Bethesda, USA
| | - Mario Schelhaas
- Emmy-Noether Group: Virus Endocytosis, Institutes of Molecular Virology and Medical Biochemistry, ZMBE, University of Münster, Münster, Germany; Cluster of Excellence EXC1003, Cells in Motion, CiM, Münster, Germany.
| |
Collapse
|
42
|
Theisen LL, Erdelmeier CAJ, Spoden GA, Boukhallouk F, Sausy A, Florin L, Muller CP. Tannins from Hamamelis virginiana bark extract: characterization and improvement of the antiviral efficacy against influenza A virus and human papillomavirus. PLoS One 2014; 9:e88062. [PMID: 24498245 PMCID: PMC3909258 DOI: 10.1371/journal.pone.0088062] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 12/23/2013] [Indexed: 11/18/2022] Open
Abstract
Antiviral activity has been demonstrated for different tannin-rich plant extracts. Since tannins of different classes and molecular weights are often found together in plant extracts and may differ in their antiviral activity, we have compared the effect against influenza A virus (IAV) of Hamamelis virginiana L. bark extract, fractions enriched in tannins of different molecular weights and individual tannins of defined structures, including pseudotannins. We demonstrate antiviral activity of the bark extract against different IAV strains, including the recently emerged H7N9, and show for the first time that a tannin-rich extract inhibits human papillomavirus (HPV) type 16 infection. As the best performing antiviral candidate, we identified a highly potent fraction against both IAV and HPV, enriched in high molecular weight condensed tannins by ultrafiltration, a simple, reproducible and easily upscalable method. This ultrafiltration concentrate and the bark extract inhibited early and, to a minor extent, later steps in the IAV life cycle and tannin-dependently inhibited HPV attachment. We observed interesting mechanistic differences between tannin structures: High molecular weight tannin containing extracts and tannic acid (1702 g/mol) inhibited both IAV receptor binding and neuraminidase activity. In contrast, low molecular weight compounds (<500 g/mol) such as gallic acid, epigallocatechin gallate or hamamelitannin inhibited neuraminidase but not hemagglutination. Average molecular weight of the compounds seemed to positively correlate with receptor binding (but not neuraminidase) inhibition. In general, neuraminidase inhibition seemed to contribute little to the antiviral activity. Importantly, antiviral use of the ultrafiltration fraction enriched in high molecular weight condensed tannins and, to a lesser extent, the unfractionated bark extract was preferable over individual isolated compounds. These results are of interest for developing and improving plant-based antivirals.
Collapse
Affiliation(s)
- Linda L. Theisen
- Institute of Immunology, Centre de Recherche Public de la Santé/Laboratoire National de Santé, Luxembourg, Luxembourg
| | | | - Gilles A. Spoden
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Fatima Boukhallouk
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Aurélie Sausy
- Institute of Immunology, Centre de Recherche Public de la Santé/Laboratoire National de Santé, Luxembourg, Luxembourg
| | - Luise Florin
- Department of Medical Microbiology and Hygiene, University Medical Centre of the Johannes Gutenberg University, Mainz, Germany
| | - Claude P. Muller
- Institute of Immunology, Centre de Recherche Public de la Santé/Laboratoire National de Santé, Luxembourg, Luxembourg
- * E-mail:
| |
Collapse
|
43
|
Wang JW, Jagu S, Kwak K, Wang C, Peng S, Kirnbauer R, Roden RBS. Preparation and properties of a papillomavirus infectious intermediate and its utility for neutralization studies. Virology 2013; 449:304-16. [PMID: 24418565 DOI: 10.1016/j.virol.2013.10.038] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 10/18/2013] [Accepted: 10/29/2013] [Indexed: 02/07/2023]
Abstract
We show that minor capsid protein L2 is full length in clinical virion isolates and prepare furin-cleaved pseudovirus (fcPsV) as a model of the infectious intermediate for multiple human papillomavirus (HPV) types. These fcPsV do not require furin for in vitro infection, and are fully infectious in vivo. Both the γ-secretase inhibitor XXI and carrageenan block fcPsV infection in vitro and in vivo implying that they act after furin-cleavage of L2. Despite their enhanced exposure of L2 epitopes, vaccination with fcPsV particles fails to induce L2 antibody, although L1-specific responses are similar to PsV with intact L2. FcPsV can be applied in a simple, high-throughput neutralization assay that detects L2-specific neutralizing antibodies with >10-fold enhanced sensitivity compared with the PsV-based assay. The PsV and fcPsV-based assays exhibit similar sensitivity for type-specific antibodies elicited by L1 virus-like particles (VLP), but the latter improves detection of L1-specific cross-type neutralizing antibodies.
Collapse
Affiliation(s)
- Joshua W Wang
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Subhashini Jagu
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Kihyuck Kwak
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Chenguang Wang
- Department of Biostatistics, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Shiwen Peng
- Department of Oncology, The Johns Hopkins University, Baltimore, MD 21231, USA
| | - Reinhard Kirnbauer
- Laboratory of Viral Oncology, Division of Immunology, Allergy and Infectious Diseases, Department of Dermatology, Medical University Vienna (MUW), Vienna, Austria
| | - Richard B S Roden
- Department of Pathology, The Johns Hopkins University, Baltimore, MD 21231, USA; Department of Oncology, The Johns Hopkins University, Baltimore, MD 21231, USA; Department of Gynecology and Obstetrics, The Johns Hopkins University, Baltimore, MD 21231, USA.
| |
Collapse
|
44
|
Multiple heparan sulfate binding site engagements are required for the infectious entry of human papillomavirus type 16. J Virol 2013; 87:11426-37. [PMID: 23966387 DOI: 10.1128/jvi.01721-13] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Human papillomavirus (HPV) entry is accompanied by multiple receptor-induced conformational changes (CCs) affecting both the major and minor capsid proteins, L1 and L2. Interaction of heparan sulfate (HS) with L1 is essential for successful HPV16 entry. Recently, cocrystallization of HPV16 with heparin revealed four distinct binding sites. Here we characterize mutant HPV16 to delineate the role of engagement with HS binding sites during infectious internalization. Site 1 (Lys278, Lys361), which mediates primary binding, is sufficient to trigger an L2 CC, exposing the amino terminus. Site 2 (Lys54, Lys356) and site 3 (Asn57, Lys59, Lys442, Lys443) are engaged following primary attachment and are required for infectious entry. Site 2 mutant particles are efficiently internalized but fail to undergo an L1 CC on the cell surface and subsequent uncoating in the endocytic compartment. After initial attachment to the cell, site 3 mutants undergo L1 and L2 CCs and then accumulate on the extracellular matrix (ECM). We conclude that the induction of CCs following site 1 and site 2 interactions results in reduced affinity for the primary HS binding site(s) on the cell surface, which allows engagement with site 3. Taken together, our findings suggest that HS binding site engagement induces CCs that prepare the virus for downstream events, such as the exposure of secondary binding sites, CCs, transfer to the uptake receptor, and uncoating.
Collapse
|
45
|
Cruz L, Meyers C. Differential dependence on host cell glycosaminoglycans for infection of epithelial cells by high-risk HPV types. PLoS One 2013; 8:e68379. [PMID: 23861898 PMCID: PMC3701689 DOI: 10.1371/journal.pone.0068379] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/27/2013] [Indexed: 12/19/2022] Open
Abstract
Human papillomavirus (HPV) infection is the leading cause of cervical cancer world-wide. Here, we show that native HPV particles produced in a differentiated epithelium have developed different strategies to infect the host. Using biochemical inhibition assays and glycosaminoglycan (GAG)-negative cells, we show that of the four most common cancer-causing HPV types, HPV18, HPV31, and HPV45 are largely dependent on GAGs to initiate infection. In contrast, HPV16 can bind and enter through a GAG-independent mechanism. Infections of primary human keratinocytes, natural host cells for HPV infections, support our conclusions. Further, this renders the different virus types differentially susceptible to carrageenan, a microbicide targeting virus entry. Our data demonstrates that ordered maturation of papillomavirus particles in a differentiating epithelium may alter the virus entry mechanism. This study should facilitate a better understanding of the attachment and infection by the main oncogenic HPV types, and development of inhibitors of HPV infection.
Collapse
Affiliation(s)
- Linda Cruz
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, Pennsylvania, United States of America
| | | |
Collapse
|