Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Virol ; 97(2): e0187922, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36749071

RESUMEN

The current model of human papillomavirus (HPV) replication is comprised of three modes of replication. Following infectious delivery, the viral genome is amplified during the establishment phase to reach up to some hundred copies per cell. The HPV genome copy number remains constant during the maintenance stage. The differentiation of infected cells induces HPV genome amplification. Using highly sensitive in situ hybridization (DNAscope) and freshly HPV16-infected as well as established HPV16-positive cell lines, we observed that the viral genome is amplified in each S phase of undifferentiated keratinocytes cultured as monolayers. The nuclear viral genome copy number is reset to pre-S-phase levels during mitosis. The majority of the viral genome fails to tether to host chromosomes and is lost to the cytosol. Cytosolic viral genomes gradually decrease during cell cycle progression. The loss of cytosolic genomes is blocked in the presence of NH4Cl or other drugs that interfere with lysosomal acidification, suggesting the involvement of autophagy in viral genome degradation. These observations were also made with HPV31 cell lines obtained from patient samples. Cytosolic viral genomes were not detected in UMSCC47 cells carrying integrated HPV16 DNA. Analyses of organotypic raft cultures derived from keratinocytes harboring episomal HPV16 revealed the presence of cytosolic viral genomes as well. We conclude that HPV maintains viral genome copy numbers by balancing viral genome amplification during S phase with the loss of viral genomes to the cytosol during mitosis. It seems plausible that restrictions to viral genome tethering to mitotic chromosomes reset genome copy numbers in each cell cycle. IMPORTANCE HPV genome maintenance is currently thought to be achieved by regulating the expression and activity of the viral replication factors E1 and E2. In addition, the E8^E2 repressor has been shown to be important for restricting genome copy numbers by competing with E1 and E2 for binding to the viral origin of replication and by recruiting repressor complexes. Here, we demonstrate that the HPV genome is amplified in each S phase. The nuclear genome copy number is reset during mitosis by a failure of the majority of the genomes to tether to mitotic chromosomes. Rather, HPV genomes accumulate in the cytoplasm of freshly divided cells. Cytosolic viral DNA is degraded in G1 in a lysosome-dependent manner, contributing to the genome copy reset. Our data imply that the mode of replication during establishment and maintenance is the same and further suggest that restrictions to genome tethering significantly contribute to viral genome maintenance.


Asunto(s)
Variaciones en el Número de Copia de ADN , Virus del Papiloma Humano , Mitosis , Proteínas Oncogénicas Virales , Replicación Viral , Humanos , Citosol/metabolismo , ADN Viral/genética , Papillomavirus Humano 16/genética , Virus del Papiloma Humano/genética , Queratinocitos , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus , Fase S , Genoma Viral
2.
J Virol ; 97(6): e0021423, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37223953

RESUMEN

Even though replication and transcription of human papillomavirus type 16 (HPV16) has been intensively studied, little is known about immediate-early events of the viral life cycle due to the lack of an efficient infection model allowing genetic dissection of viral factors. We employed the recently developed infection model (Bienkowska-Haba M, Luszczek W, Myers JE, Keiffer TR, et al. 2018. PLoS Pathog 14:e1006846) to investigate genome amplification and transcription immediately after infectious delivery of viral genome to nuclei of primary keratinocytes. Using 5-ethynyl-2'-deoxyuridine (EdU) pulse-labeling and highly sensitive fluorescence in situ hybridization, we observed that the HPV16 genome is replicated and amplified in an E1- and E2-dependent manner. Knockout of E1 resulted in failure of the viral genome to replicate and amplify. In contrast, knockout of the E8^E2 repressor led to increased viral genome copy number, confirming previous reports. Genome copy control by E8^E2 was confirmed for differentiation-induced genome amplification. Lack of functional E1 had no effect on transcription from the early promoter, suggesting that viral genome replication is not required for p97 promoter activity. However, infection with an HPV16 mutant virus defective for E2 transcriptional function revealed a requirement of E2 for efficient transcription from the early promoter. In the absence of the E8^E2 protein, early transcript levels are unaltered and even decreased when normalized to genome copy number. Surprisingly, a lack of functional E8^E2 repressor did not affect E8^E2 transcript levels when normalized to genome copy number. These data suggest that the main function of E8^E2 in the viral life cycle is to control genome copy number. IMPORTANCE It is being assumed that human papillomavirus (HPV) utilizes three different modes of replication during its life cycle: initial amplification during the establishment phase, genome maintenance, and differentiation-induced amplification. However, HPV16 initial amplification was never formally proven due to a lack of an infection model. Using our recently established infection model (Bienkowska-Haba M, Luszczek W, Myers JE, Keiffer TR, et al. 2018. PLoS Pathog 14:e1006846), we demonstrate herein that viral genome is indeed amplified in an E1- and E2-dependent manner. Furthermore, we find that the main function of the viral repressor E8^E2 is to control viral genome copy number. We did not find evidence that it regulates its own promoter in a negative feedback loop. Our data also suggest that the E2 transactivator function is required for stimulation of early promoter activity, which has been debated in the literature. Overall, this report confirms the usefulness of the infection model for studying early events of the HPV life cycle using mutational approaches.


Asunto(s)
Genoma Viral , Papillomavirus Humano 16 , Infecciones por Papillomavirus , Humanos , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/virología , Replicación Viral/genética , Genoma Viral/genética , Células 3T3 NIH , Animales , Ratones , Línea Celular , Células HEK293 , Transcripción Viral/genética
3.
J Virol ; 97(2): e0103222, 2023 02 28.
Artículo en Inglés | MEDLINE | ID: mdl-36719239

RESUMEN

Coinfection of human papillomavirus (HPV) and Epstein-Barr virus (EBV) has been detected in oropharyngeal squamous cell carcinoma. Although HPV and EBV replicate in differentiated epithelial cells, we previously reported that HPV epithelial immortalization reduces EBV replication within organotypic raft culture and that the HPV16 oncoprotein E7 was sufficient to inhibit EBV replication. A well-established function of HPV E7 is the degradation of the retinoblastoma (Rb) family of pocket proteins (pRb, p107, and p130). Here, we show that pRb knockdown in differentiated epithelia and EBV-positive Burkitt lymphoma (BL) reduces EBV lytic replication following de novo infection and reactivation, respectively. In differentiated epithelia, EBV immediate early (IE) transactivators were expressed, but loss of pRb blocked expression of the early gene product, EA-D. Although no alterations were observed in markers of epithelial differentiation, DNA damage, and p16, increased markers of S-phase progression and altered p107 and p130 levels were observed in suprabasal keratinocytes after pRb knockdown. In contrast, pRb interference in Akata BX1 Burkitt lymphoma cells showed a distinct phenotype from differentiated epithelia with no significant effect on EBV IE or EA-D expression. Instead, pRb knockdown reduced the levels of the plasmablast differentiation marker PRDM1/Blimp1 and increased the abundance of c-Myc protein in reactivated Akata BL with pRb knockdown. c-Myc RNA levels also increased following the loss of pRb in epithelial rafts. These results suggest that pRb is required to suppress c-Myc for efficient EBV replication in BL cells and identifies a mechanism for how HPV immortalization, through degradation of the retinoblastoma pocket proteins, interferes with EBV replication in coinfected epithelia. IMPORTANCE Terminally differentiated epithelium is known to support EBV genome amplification and virion morphogenesis following infection. The contribution of the cell cycle in differentiated tissues to efficient EBV replication is not understood. Using organotypic epithelial raft cultures and genetic interference, we can identify factors required for EBV replication in quiescent cells. Here, we phenocopied HPV16 E7 inhibition of EBV replication through knockdown of pRb. Loss of pRb was found to reduce EBV early gene expression and viral replication. Interruption of the viral life cycle was accompanied by increased S-phase gene expression in postmitotic keratinocytes, a process also observed in E7-positive epithelia, and deregulation of other pocket proteins. Together, these findings provide evidence of a global requirement for pRb in EBV lytic replication and provide a mechanistic framework for how HPV E7 may facilitate a latent EBV infection through its mediated degradation of pRb in copositive epithelia.


Asunto(s)
Linfoma de Burkitt , Infecciones por Virus de Epstein-Barr , Proteína de Retinoblastoma , Replicación Viral , Humanos , Linfoma de Burkitt/virología , Diferenciación Celular , Epitelio/virología , Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Infecciones por Papillomavirus , Proteína de Retinoblastoma/metabolismo
4.
J Virol ; 94(3)2020 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-31748387

RESUMEN

It is established that the host cell transcriptomes of natural lesions, organotypic rafts, and human papillomavirus (HPV)-immortalized keratinocytes are altered in the presence of HPV genomes. However, the establishment of HPV-harboring cell lines requires selection and immortalization, which makes it impossible to distinguish between alterations directly induced by HPV or indirectly by the need for immortalization or selection. To address direct effects of HPV infection on the host cell transcriptome, we have used our recently established infection model that allows efficient infection of primary keratinocytes with HPV16 virions. We observed only a small set of genes to be deregulated at the transcriptional level at 7 days postinfection (dpi), most of which fall into the category regulated by pocket proteins pRb, p107, and p130. Furthermore, cell cycle genes were not deregulated in cells infected with a virus lacking E7 despite the presence of episomal genome and viral transcripts. These findings imply that the majority of transcriptional changes are due to the E7 protein impairing pocket protein function. Additional pathways, such as the Fanconi anemia-BRCA pathway, became perturbed only after long-term culturing of infected cells. When grown as organotypic raft cultures, keratinocytes infected with wild-type but not E7 mutant virus had perturbed transcriptional regulation of pathways previously identified in natural lesions and in rafts derived from immortalized keratinocytes. We conclude that the HPV infection model provides a valuable tool to distinguish immediate transcriptional alterations from those induced by persistent infection and the need for selection and immortalization.IMPORTANCE To establish infection and complete the viral life cycle, human papillomavirus (HPV) needs to alter the transcriptional program of host cells. Until recently, studies were restricted to keratinocyte-derived cell lines immortalized by HPV due to the lack of experimental systems to efficiently infect primary keratinocytes. Need for selection and immortalization made it impossible to distinguish between alterations induced by HPV and secondary adaptation due to selection and immortalization. With our recent establishment of an extracellular matrix (ECM)-to-cell transfer system allowing efficient infection of primary keratinocytes, we were able to identify transcriptional changes attributable to HPV16 infection. Most perturbed genes fall into the class of S-phase genes, which are regulated by pocket proteins. Indeed, infection with viruses lacking E7 abrogated most transcriptional changes. It is important to note that many transcriptional alterations thought to be important for the HPV life cycle are actually late events that may reflect immortalization and, possibly, disease progression.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Papillomavirus Humano 16/fisiología , Queratinocitos/virología , Papillomaviridae/genética , Papillomaviridae/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Ciclo Celular/genética , Línea Celular , Regulación Viral de la Expresión Génica , Humanos , Queratinocitos/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/virología , Transcriptoma
5.
PLoS Pathog ; 15(2): e1007590, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30802273

RESUMEN

Subnuclear promyelocytic leukemia (PML) nuclear bodies (NBs) are targeted by many DNA viruses after nuclear delivery. PML protein is essential for formation of PML NBs. Sp100 and Small Ubiquitin-Like Modifier (SUMO) are also permanently residing within PML NBs. Often, large DNA viruses disassemble and reorganize PML NBs to counteract their intrinsic antiviral activity and support establishment of infection. However, human papillomavirus (HPV) requires PML protein to retain incoming viral DNA in the nucleus for subsequent efficient transcription. In contrast, Sp100 was identified as a restriction factor for HPV. These findings suggested that PML NBs are important regulators of early stages of the HPV life cycle. Nuclear delivery of incoming HPV DNA requires mitosis. Viral particles are retained within membrane-bound transport vesicles throughout mitosis. The viral genome is released from transport vesicles by an unknown mechanism several hours after nuclear envelope reformation. The minor capsid protein L2 mediates intracellular transport by becoming transmembranous in the endocytic compartment. Herein, we tested our hypothesis that PML protein is recruited to incoming viral genome prior to egress from transport vesicles. High-resolution microscopy revealed that PML protein, SUMO-1, and Sp100 are recruited to incoming viral genomes, rather than viral genomes being targeted to preformed PML NBs. Differential immunofluorescent staining suggested that PML protein and SUMO-1 associated with transport vesicles containing viral particles prior to egress, implying that recruitment is likely mediated by L2 protein. In contrast, Sp100 recruitment to HPV-harboring PML NBs occurred after release of viral genomes from transport vesicles. The delayed recruitment of Sp100 is specific for HPV-associated PML NBs. These data suggest that the virus continuously resides within a protective environment until the transport vesicle breaks down in late G1 phase and imply that HPV might modulate PML NB assembly to achieve establishment of infection and the shift to viral maintenance.


Asunto(s)
Papillomaviridae/genética , Papillomaviridae/metabolismo , Proteína de la Leucemia Promielocítica/metabolismo , Antígenos Nucleares/metabolismo , Autoantígenos/metabolismo , Proteínas de la Cápside , Núcleo Celular , Genoma Viral , Humanos , Cuerpos de Inclusión Intranucleares , Espacio Intranuclear , Proteínas Nucleares , Papillomaviridae/patogenicidad , Proteína de la Leucemia Promielocítica/fisiología , Proteína SUMO-1/metabolismo , Factores de Transcripción , Proteínas Supresoras de Tumor , Replicación Viral
6.
PLoS Pathog ; 14(3): e1006846, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29494681

RESUMEN

Herein, we describe a novel infection model that achieves highly efficient infection of primary keratinocytes with human papillomavirus type 16 (HPV16). This cell culture model does not depend on immortalization and is amenable to extensive genetic analyses. In monolayer cell culture, the early but not late promoter was active and yielded a spliced viral transcript pattern similar to HPV16-immortalized keratinocytes. However, relative levels of the E8^E2 transcript increased over time post infection suggesting the expression of this viral repressor is regulated independently of other early proteins and that it may be important for the shift from the establishment to the maintenance phase of the viral life cycle. Both the early and the late promoter were strongly activated when infected cells were subjected to differentiation by growth in methylcellulose. When grown as organotypic raft cultures, HPV16-infected cells expressed late E1^E4 and L1 proteins and replication foci were detected, suggesting that they supported the completion of the viral life cycle. As a proof of principle that the infection system may be used for genetic dissection of viral factors, we analyzed E1, E6 and E7 translation termination linker mutant virus for establishment of infection and genome maintenance. E1 but not E6 and E7 was essential to establish infection. Furthermore, E6 but not E7 was required for episomal genome maintenance. Primary keratinocytes infected with wild type HPV16 immortalized, whereas keratinocytes infected with E6 and E7 knockout virus began to senesce 25 to 35 days post infection. The novel infection model provides a powerful genetic tool to study the role of viral proteins throughout the viral life cycle but especially for immediate early events and enables us to compare low- and high-risk HPV types in the context of infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Papillomavirus Humano 16/patogenicidad , Queratinocitos/virología , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/virología , Replicación Viral , Células Cultivadas , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología
7.
Proc Natl Acad Sci U S A ; 113(22): 6289-94, 2016 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-27190090

RESUMEN

During the entry process, the human papillomavirus (HPV) capsid is trafficked to the trans-Golgi network (TGN), whereupon it enters the nucleus during mitosis. We previously demonstrated that the minor capsid protein L2 assumes a transmembranous conformation in the TGN. Here we provide evidence that the incoming viral genome dissociates from the TGN and associates with microtubules after the onset of mitosis. Deposition onto mitotic chromosomes is L2-mediated. Using differential staining of an incoming viral genome by small molecular dyes in selectively permeabilized cells, nuclease protection, and flotation assays, we found that HPV resides in a membrane-bound vesicle until mitosis is completed and the nuclear envelope has reformed. As a result, expression of the incoming viral genome is delayed. Taken together, these data provide evidence that HPV has evolved a unique strategy for delivering the viral genome to the nucleus of dividing cells. Furthermore, it is unlikely that nuclear vesicles are unique to HPV, and thus we may have uncovered a hitherto unrecognized cellular pathway that may be of interest for future cell biological studies.


Asunto(s)
Vesículas Citoplasmáticas/metabolismo , Genoma Viral , Papillomavirus Humano 16/fisiología , Mitosis/fisiología , Infecciones por Papillomavirus/virología , Virión , Internalización del Virus , Núcleo Celular/metabolismo , Núcleo Celular/virología , Vesículas Citoplasmáticas/virología , Endosomas/metabolismo , Endosomas/virología , Células HeLa , Humanos , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Transporte de Proteínas , Red trans-Golgi/metabolismo , Red trans-Golgi/virología
8.
J Virol ; 91(16)2017 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-28566382

RESUMEN

During infectious entry, acidification within the endosome triggers uncoating of the human papillomavirus (HPV) capsid, whereupon host cyclophilins facilitate the release of most of the major capsid protein, L1, from the minor capsid protein L2 and the viral genome. The L2/DNA complex traffics to the trans-Golgi network (TGN). After the onset of mitosis, HPV-harboring transport vesicles bud from the TGN, followed by association with mitotic chromosomes. During this time, the HPV genome remains in a vesicular compartment until the nucleus has completely reformed. Recent data suggest that while most of L1 protein dissociates and is degraded in the endosome, some L1 protein remains associated with the viral genome. The L1 protein has DNA binding activity, and the L2 protein has multiple domains capable of interacting with L1 capsomeres. In this study, we report that some L1 protein traffics with L2 and viral genome to the nucleus. The accompanying L1 protein is mostly full length and retains conformation-dependent epitopes, which are recognized by neutralizing antibodies. Since more than one L1 molecule contributes to these epitopes and requires assembly into capsomeres, we propose that L1 protein is present in the form of pentamers. Furthermore, we provide evidence that the L1 protein interacts directly with viral DNA within the capsid. Based on our findings, we propose that the L1 protein, likely arranged as capsomeres, stabilizes the viral genome within the subviral complex during intracellular trafficking.IMPORTANCE After internalization, the nonenveloped human papillomavirus virion uncoats in the endosome, whereupon conformational changes result in a dissociation of a subset of the major capsid protein L1 from the minor capsid protein L2, which remains in complex with the viral DNA. Recent data suggest that some L1 protein may accompany the viral genome beyond the endosomal compartment. We demonstrate that conformationally intact L1 protein, likely still arranged as capsomeres, remains associated with the incoming viral genome throughout mitosis and transiently resides in the nucleus until after the viral DNA is released from the transport vesicle.

10.
Cell Microbiol ; 19(5)2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27860076

RESUMEN

Human papillomaviruses (HPVs) target promyelocytic leukemia (PML) nuclear bodies (NBs) during infectious entry and PML protein is important for efficient transcription of incoming viral genome. However, the transcriptional down regulation was shown to be promoter-independent in that heterologous promoters delivered by papillomavirus particles were also affected. To further investigate the role of PML protein in HPV entry, we used small hairpin RNA to knockdown PML protein in HaCaT keratinocytes. Confirming previous findings, PML knockdown in HaCaT cells reduced HPV16 transcript levels significantly following infectious entry without impairing binding and trafficking. However, when we quantified steady-state levels of pseudogenomes in interphase cells, we found strongly reduced genome levels compared with parental HaCaT cells. Because nuclear delivery was comparable in both cell lines, we conclude that viral pseudogenome must be removed after successful nuclear delivery. Transcriptome analysis by gene array revealed that PML knockdown in clonal HaCaT cells was associated with a constitutive interferon response. Abrogation of JAK1/2 signaling prevented genome loss, however, did not restore viral transcription. In contrast, knockdown of PML protein in HeLa cells did not affect HPV genome delivery and transcription. HeLa cells are transformed by HPV18 oncogenes E6 and E7, which have been shown to interfere with the JAK/Stat signaling pathway. Our data imply that PML NBs protect incoming HPV genomes. Furthermore, they provide evidence that PML NBs are key regulators of the innate immune response in keratinocytes. IMPORTANCE: Promyelocytic leukemia nuclear bodies (PML NBs) are important for antiviral defense. Many DNA viruses target these subnuclear structures and reorganize them. Reorganization of PML NBs by viral proteins is important for establishment of infection. In contrast, HPVs require the presence of PML protein for efficient transcription of incoming viral genome. Our finding that PML protein prevents the loss of HPV genome following infection implies that the host cell may be able to recognize chromatinized HPV genome or the associated capsid proteins. A constitutively active interferon response in absence of PML protein suggests that PML NBs are key regulators of the innate immune response in keratinocytes.


Asunto(s)
Papillomavirus Humano 16/genética , Queratinocitos/virología , Infecciones por Papillomavirus/virología , Proteína de la Leucemia Promielocítica/metabolismo , Línea Celular , Núcleo Celular/virología , Expresión Génica , Técnicas de Silenciamiento del Gen , Genoma Viral , Papillomavirus Humano 16/fisiología , Humanos , Inmunidad Innata , Interferones/genética , Interferones/metabolismo , Proteína de la Leucemia Promielocítica/genética , Internalización del Virus
11.
J Virol ; 90(18): 8032-5, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27412595

RESUMEN

Incoming human papillomavirus (HPV) utilize vesicular transport to traffic from the plasma membrane to the trans-Golgi network. Following nuclear envelope breakdown during mitosis, the viral DNA associates with condensed chromosomes utilizing spindle microtubules for delivery. Most intriguingly, the viral DNA resides in a transport vesicle until mitosis is completed and the nuclear envelope has reformed. This finding provides support for the transient existence of nuclear membrane-bound vesicles. Due to their transient nature, it also points to the existence of a cell pathway for the disposal of vesicles ending up fortuitously or purposefully in the nucleus.


Asunto(s)
Papillomaviridae/fisiología , Internalización del Virus , Vesículas Citoplasmáticas/virología , Endocitosis , Humanos
12.
J Virol ; 89(20): 10442-52, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26246568

RESUMEN

UNLABELLED: The human papillomavirus (HPV) capsid is composed of the major capsid protein L1 and the minor capsid protein L2. During entry, the HPV capsid undergoes numerous conformational changes that result in endosomal uptake and subsequent trafficking of the L2 protein in complex with the viral DNA to the trans-Golgi network. To facilitate this transport, the L2 protein harbors a number of putative motifs that, if capable of direct interaction, would interact with cytosolic host cell factors. These data imply that a portion of L2 becomes cytosolic during infection. Using a low concentration of digitonin to selectively permeabilize the plasma membrane of infected cells, we mapped the topography of the L2 protein during infection. We observed that epitopes within amino acid residues 64 to 81 and 163 to 170 and a C-terminal tag of HPV16 L2 are exposed on the cytosolic side of intracellular membranes, whereas an epitope within residues 20 to 38, which are upstream of a putative transmembrane region, is luminal. Corroborating these findings, we also found that L2 protein is sensitive to trypsin digestion during infection. These data demonstrate that the majority of the L2 protein becomes accessible on the cytosolic side of intracellular membranes in order to interact with cytosolic factors to facilitate vesicular trafficking. IMPORTANCE: In order to complete infectious entry, nonenveloped viruses have to pass cellular membranes. This is often achieved through the viral capsid protein associating with or integrating into intracellular membrane. Here, we determine the topography of HPV L2 protein in the endocytic vesicular compartment, suggesting that L2 becomes a transmembrane protein with a short luminal portion and with the majority facing the cytosolic side for interaction with host cell transport factors.


Asunto(s)
Proteínas de la Cápside/química , Cápside/química , Papillomavirus Humano 16/química , Queratinocitos/virología , Proteínas Oncogénicas Virales/química , Internalización del Virus , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Línea Celular Transformada , Permeabilidad de la Membrana Celular/efectos de los fármacos , Digitonina/farmacología , Endosomas/química , Endosomas/metabolismo , Endosomas/virología , Células HEK293 , Células HeLa , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/química , Papillomavirus Humano 18/metabolismo , Humanos , Membranas Intracelulares/química , Membranas Intracelulares/metabolismo , Membranas Intracelulares/virología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Dominios y Motivos de Interacción de Proteínas , Transporte de Proteínas , Proteolisis , Tripsina/química , Virión/química , Virión/metabolismo
13.
J Virol ; 87(21): 11426-37, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23966387

RESUMEN

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.


Asunto(s)
Proteínas de la Cápside/metabolismo , Heparitina Sulfato/metabolismo , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/metabolismo , Internalización del Virus , Sitios de Unión , Proteínas de la Cápside/genética , Línea Celular , Análisis Mutacional de ADN , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Proteínas Oncogénicas Virales/genética
14.
J Virol ; 87(13): 7765-73, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23616662

RESUMEN

Human papillomavirus type 18 (HPV18), one of the HPVs with malignant potential, enters cells by an unknown endocytic mechanism. The key cellular requirements for HPV18 endocytosis were tested in comparison to those for HPV16 and -31 endocytoses. HPV18 (like HPV16 and -31) entry was independent of clathrin, caveolin, dynamin, and lipid rafts but required actin polymerization and tetraspanin CD151, and the viruses were routed to the same LAMP-1-positive compartment. Hence, the viruses shared similar cellular requirements for endocytic entry.


Asunto(s)
Endocitosis/fisiología , Papillomavirus Humano 16/fisiología , Papillomavirus Humano 18/fisiología , Papillomavirus Humano 31/fisiología , Internalización del Virus , Actinas/metabolismo , Dinamina II , Electroforesis en Gel de Poliacrilamida , Células HeLa , Humanos , Proteínas de Membrana de los Lisosomas/metabolismo , Microdominios de Membrana , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Polimerizacion , Tetraspanina 24/metabolismo
15.
J Virol ; 86(18): 9875-87, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22761365

RESUMEN

Human papillomaviruses (HPV) are composed of the major and minor capsid proteins, L1 and L2, that encapsidate a chromatinized, circular double-stranded DNA genome. At the outset of infection, the interaction of HPV type 16 (HPV16) (pseudo)virions with heparan sulfate proteoglycans triggers a conformational change in L2 that is facilitated by the host cell chaperone cyclophilin B (CyPB). This conformational change results in exposure of the L2 N terminus, which is required for infectious internalization. Following internalization, L2 facilitates egress of the viral genome from acidified endosomes, and the L2/DNA complex accumulates at PML nuclear bodies. We recently described a mutant virus that bypasses the requirement for cell surface CyPB but remains sensitive to cyclosporine for infection, indicating an additional role for CyP following endocytic uptake of virions. We now report that the L1 protein dissociates from the L2/DNA complex following infectious internalization. Inhibition and small interfering RNA (siRNA)-mediated knockdown of CyPs blocked dissociation of L1 from the L2/DNA complex. In vitro, purified CyPs facilitated the dissociation of L1 pentamers from recombinant HPV11 L1/L2 complexes in a pH-dependent manner. Furthermore, CyPs released L1 capsomeres from partially disassembled HPV16 pseudovirions at slightly acidic pH. Taken together, these data suggest that CyPs mediate the dissociation of HPV L1 and L2 capsid proteins following acidification of endocytic vesicles.


Asunto(s)
Proteínas de la Cápside/fisiología , Ciclofilinas/fisiología , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/fisiología , Sustitución de Aminoácidos , Proteínas de la Cápside/química , Proteínas de la Cápside/genética , Línea Celular , Ciclofilina A/antagonistas & inhibidores , Ciclofilina A/genética , Ciclofilina A/fisiología , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , ADN Viral/química , ADN Viral/genética , ADN Viral/metabolismo , Endosomas/fisiología , Endosomas/virología , Técnicas de Silenciamiento del Gen , Genoma Viral , Células HEK293 , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Humanos , Concentración de Iones de Hidrógeno , Sustancias Macromoleculares , Mutagénesis Sitio-Dirigida , Proteínas Oncogénicas Virales/química , Proteínas Oncogénicas Virales/genética , Conformación Proteica , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Internalización del Virus
16.
J Biol Chem ; 286(4): 2617-24, 2011 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-21115492

RESUMEN

High risk human papillomavirus types 16 (HPV16) and 18 (HPV18) can cause cervical cancer. Efficient infection by HPV16 and HPV18 pseudovirions requires interactions of particles with cell-surface receptor heparan sulfate oligosaccharide. To understand the virus-receptor interactions for HPV infection, we determined the crystal structures of HPV16 and HPV18 capsids bound to the oligosaccharide receptor fragment using oligomeric heparin. The HPV-heparin structures revealed multiple binding sites for the highly negatively charged oligosaccharide fragment on the capsid surface, which is different from previously reported virus-receptor interactions in which a single type of binding pocket is present for a particular receptor. We performed structure-guided mutagenesis to generate mutant viruses, and cell binding and infectivity assays demonstrated the functional role of viral residues involved in heparin binding. These results provide a basis for understanding virus-heparan sulfate receptor interactions critical for HPV infection and for the potential development of inhibitors against HPV infection.


Asunto(s)
Heparitina Sulfato/química , Papillomavirus Humano 16/química , Papillomavirus Humano 18/química , Sitios de Unión , Cristalografía por Rayos X , Heparitina Sulfato/genética , Heparitina Sulfato/metabolismo , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/genética , Papillomavirus Humano 18/metabolismo , Humanos , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/metabolismo , Relación Estructura-Actividad
17.
Med Microbiol Immunol ; 201(4): 437-48, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22972234

RESUMEN

Papillomaviruses infect skin and mucosa where they induce warts and cancers. For entry to occur, they sequentially engage numerous host proteins, allowing them to deliver their genetic information into target cells. This multistep process starts with initial binding via its L1 major capsid protein, followed by structural changes of the capsid on the cell surface, engagement of different receptors, and endocytosis. The post-entry phase includes capsid disassembly, endosomal escape of a complex of the minor capsid protein L2 and the viral genome, its transport into the nucleus, and accumulation at nuclear substructures. This review summarizes the current knowledge of the papillomavirus entry pathway and the role of cellular proteins involved in this course of events.


Asunto(s)
Interacciones Huésped-Patógeno , Papillomaviridae/fisiología , Internalización del Virus , Células Epiteliales/virología , Humanos
18.
PLoS Pathog ; 5(7): e1000524, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19629175

RESUMEN

Following attachment to primary receptor heparan sulfate proteoglycans (HSPG), human papillomavirus type 16 (HPV16) particles undergo conformational changes affecting the major and minor capsid proteins, L1 and L2, respectively. This results in exposure of the L2 N-terminus, transfer to uptake receptors, and infectious internalization. Here, we report that target cell cyclophilins, peptidyl-prolyl cis/trans isomerases, are required for efficient HPV16 infection. Cell surface cyclophilin B (CyPB) facilitates conformational changes in capsid proteins, resulting in exposure of the L2 N-terminus. Inhibition of CyPB blocked HPV16 infection by inducing noninfectious internalization. Mutation of a putative CyP binding site present in HPV16 L2 yielded exposed L2 N-terminus in the absence of active CyP and bypassed the need for cell surface CyPB. However, this mutant was still sensitive to CyP inhibition and required CyP for completion of infection, probably after internalization. Taken together, these data suggest that CyP is required during two distinct steps of HPV16 infection. Identification of cell surface CyPB will facilitate the study of the complex events preceding internalization and adds a putative drug target for prevention of HPV-induced diseases.


Asunto(s)
Ciclofilinas/metabolismo , Papillomavirus Humano 16/fisiología , Infecciones por Papillomavirus/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Línea Celular Tumoral , Ciclofilinas/antagonistas & inhibidores , Ciclofilinas/genética , Técnicas de Silenciamiento del Gen , Células HeLa , Papillomavirus Humano 16/metabolismo , Papillomavirus Humano 18/metabolismo , Humanos , Microscopía Fluorescente , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/prevención & control , Infecciones por Papillomavirus/virología , Unión Proteica , Estructura Terciaria de Proteína , ARN Interferente Pequeño , Acoplamiento Viral , Internalización del Virus
20.
Microorganisms ; 9(10)2021 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-34683397

RESUMEN

Papillomaviruses are a diverse viral species, but several types such as HPV16 are given special attention due to their contribution towards the pathogenesis of several major cancers. In this review, we will summarize how the knowledge of HPV16 entry has expanded since the last comprehensive HPV16 entry review our lab published in 2017.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA