Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 835
Filtrar
1.
Rev Med Virol ; 31(2): e2169, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33590566

RESUMEN

While high-risk human papillomavirus (HR-HPV) infection is related to the development of cervical, vulvar, anal, penile and oropharyngeal cancer, low-risk human papillomavirus (LR-HPV) infection is implicated in about 90% of genital warts, which rarely progress to cancer. The carcinogenic role of HR-HPV is due to the overexpression of HPV E5, E6 and E7 oncoproteins which target and modify cellular proteins implicated in cell proliferation, apoptosis and immortalization. LR-HPV proteins also target and modify some of these processes; however, their oncogenic potential is lower than that of HR-HPV. HR-HPVs have substantial differences with LR-HPVs such as viral integration into the cell genome, induction of p53 and retinoblastoma protein degradation, alternative splicing in HR-HPV E6-E7 open reading frames, among others. In addition, LR-HPV can activate the autophagy process in infected cells while HR-HPV infection deactivates it. However, in cancer HR-HPV might reactivate autophagy in advance stages. Autophagy is a catabolic process that maintains cell homoeostasis by lysosomal degradation and recycling of damaged macromolecules and organelles; nevertheless, depending upon cellular context autophagy may also induce cell death. Therefore, autophagy can contribute either as a promotor or as a suppressor of tumours. In this review, we focus on the role of HR-HPV and LR-HPV in autophagy during viral infection and cancer development. Additionally, we review key regulatory molecules such as microRNAs in HPV present during autophagy, and we emphasize the potential use of cancer treatments associated with autophagy in HPV-related cancers.


Asunto(s)
Alphapapillomavirus , Autofagia , MicroARNs/genética , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Infecciones por Papillomavirus , Humanos , Proteínas Oncogénicas , Proteínas Oncogénicas Virales/fisiología , Infecciones por Papillomavirus/complicaciones
2.
J Virol ; 93(12)2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-30971468

RESUMEN

Low-risk human papillomaviruses (LR-HPVs) are the causative agents of genital warts, which are a widespread sexually transmitted disease. How LR-HPVs affect autophagy and the specific proteins involved are unknown. In the current study, we investigated the impact of LR-HPV11 early protein 6 (E6) on the activity of the autophagy pathway. We transfected an HPV11 E6 (11E6) plasmid into HaCaT cells, H8 cells, and NHEK cells and established a stable cell line expressing the HPV11 E6 protein. The differences in autophagy activity and upstream regulatory pathways compared with those in the parent cell lines were investigated using a Western blot analysis of the total and phosphorylated protein levels and confocal microscopy of immunostained cells and cells transfected with an mCherry-green fluorescent protein-LC3 expression plasmid. We used short hairpin RNA (shRNA) to knock down 11E6 and showed that these effects require continued 11E6 expression. Compared with its expression in the control cells, the expression of HPV11 E6 in the cells activated the autophagy pathway. The increased autophagy activity was the result of the decreased phosphorylation levels of the canonical autophagy repressor mammalian target of rapamycin (mTOR) at its Ser2448 position (the mTOR complex 1 [mTORC1] phosphorylation site) and decreased AKT and Erk phosphorylation. Therefore, these results indicate that HPV11 E6 activates autophagy through the AKT/mTOR and Erk/mTOR pathways. Our findings provide novel insight into the relationship between LR-HPV infections and autophagy and could help elucidate the pathogenic mechanisms of LR-HPV.IMPORTANCE We transfected an HPV11 E6 plasmid into HaCaT cells, H8 cells, and NHEK cells and established a stable cell line expressing the HPV11 E6 protein. Then, we confirmed that HPV11 E6 induces autophagy by suppressing the AKT/mTOR and Erk/mTOR pathways. In contrast to the high-risk HPV E6 genes, HPV11 E6 did not affect the expression of p53. To the best of our knowledge, this study represents the first direct in-depth investigation of the relationship between the LR-HPV E6 gene and autophagy, which may help to reveal the pathogenesis of LR-HPV infection.


Asunto(s)
Autofagia/fisiología , Papillomavirus Humano 11/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Línea Celular , Papillomavirus Humano 11/genética , Humanos , Sistema de Señalización de MAP Quinasas/fisiología , Proteínas Oncogénicas Virales/fisiología , Infecciones por Papillomavirus/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo
3.
J Virol ; 93(4)2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30518656

RESUMEN

Human papillomaviruses (HPVs) are causative agents in ano-genital and oropharyngeal cancers. The virus must reprogram host gene expression to promote infection, and E6 and E7 contribute to this via the targeting of cellular transcription factors, including p53 and pRb, respectively. The HPV16 E2 protein regulates host gene expression in U2OS cells, and in this study, we extend these observations into telomerase reverse transcriptase (TERT) immortalized oral keratinocytes (NOKs) that are capable of supporting late stages of the HPV16 life cycle. We observed repression of innate immune genes by E2 that are also repressed by the intact HPV16 genome in NOKs. Transcriptome sequencing (RNA-seq) data identified 167 up- and 395 downregulated genes by E2; there was a highly significant overlap of the E2-regulated genes with those regulated by the intact HPV16 genome in the same cell type. Small interfering RNA (siRNA) targeting of E2 reversed the repression of E2-targeted genes. The ability of E2 to repress innate immune genes was confirmed in an ano-genital immortalized keratinocyte cell line, N/Tert-1. We present the analysis of data from The Cancer Genome Atlas (TCGA) for HPV16-positive and -negative head and neck cancers (HNC) suggesting that E2 plays a role in the regulation of the host genome in cancers. Patients with HPV16-positive HNC with a loss of E2 expression exhibited a worse clinical outcome, and we discuss how this could, at least partially, be related to the loss of E2 host gene regulation.IMPORTANCE Human papillomavirus 16 (HPV16)-positive tumors that retain expression of E2 have a better clinical outcome than those that have lost E2 expression. It has been suggested that this is due to a loss of E2 repression of E6 and E7 expression, but this is not supported by data from tumors where there is not more E6 and E7 expression in the absence of E2. Here we report that E2 regulates host gene expression and place this regulation in the context of the HPV16 life cycle and HPV16-positive head and neck cancers (the majority of which retain E2 expression). We propose that this E2 function may play an important part in the increased response of HPV16-positive cancers to radiation therapy. Therefore, host gene regulation by E2 may be important for promotion of the HPV16 life cycle and also for the response of HPV16-positive tumors to radiation therapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Línea Celular , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Expresión Génica , Regulación de la Expresión Génica/genética , Regulación Viral de la Expresión Génica/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/fisiología , Papillomaviridae/genética , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo
4.
J Virol ; 93(10)2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30842331

RESUMEN

The papillomavirus (PV) E2 protein coordinates viral transcription and genome replication. Following a strategy to identify amino acids in E2 that are posttranslationally modified, we reported that tyrosine kinase fibroblast growth factor receptor 3 (FGFR3) complexes with and phosphorylates E2, which inhibits viral DNA replication. Here, we present several lines of evidence indicating that tyrosine (Y) 138 of HPV-31 E2 is a substrate of FGFR3. The active form of FGFR3 bound to and phosphorylated the region of amino acids (aa) 107 to 175 in HPV-31 E2. The E2 phenylalanine (F) mutant Y138F displayed reduced FGFR3-induced phosphotyrosine. A constitutive kinase-active FGFR3 inhibited wild-type (WT) E2-induced E1-dependent DNA replication, while the 138F mutant retained activity. The tyrosine to glutamic acid (E) mutant Y138E, which can mimic phosphotyrosine, failed to induce transient DNA replication, although it maintained the ability to bind and localize the viral DNA helicase E1 to the viral origin. The bromodomain-containing protein 4 (Brd4) binds to E2 and is necessary for initiation of viral DNA synthesis. Interestingly, the Y138E protein coimmunoprecipitated with full-length Brd4 but was defective for association with its C-terminal domain (CTD). These results imply that the activity of the FGFR3 kinase in the infected epithelial cell restricts the HPV replication program through phosphorylation of E2 at Y138, which interferes with E2 binding to the Brd4 CTD, and that this interaction is required for initiation of viral DNA synthesis.IMPORTANCE Human papillomaviruses (HPVs) are highly infectious pathogens that commonly infect the oropharynx and uterine cervix. The idea that posttranslational modifications of viral proteins coordinates viral genome replication is less explored. We recently discovered that fibroblast growth factor receptor 3 (FGFR3) phosphorylates the viral E2 protein. The current study demonstrates that FGFR3 phosphorylates E2 at tyrosine 138, which inhibits association with the C-terminal peptide of Brd4. This study illustrates a novel regulatory mechanism of virus-host interaction and provides insight into the role of Brd4 in viral replication.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Factores de Transcripción/metabolismo , Replicación Viral/fisiología , Proteínas de Ciclo Celular/fisiología , ADN Helicasas/metabolismo , Replicación del ADN , ADN Viral/metabolismo , Proteínas de Unión al ADN/fisiología , Células HEK293 , Células HeLa , Humanos , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/fisiología , Papillomaviridae/metabolismo , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/metabolismo , Fosforilación , Unión Proteica , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/metabolismo , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Factores de Transcripción/fisiología , Tirosina/metabolismo , Replicación Viral/genética
5.
Exp Cell Res ; 381(2): 311-322, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31125556

RESUMEN

Human papilloma virus oncogenes and estradiol are major etiologic factors associated with cervical cancer. In order to understand the mechanism by which these two factors promote carcinogenesis, the role of the Hedgehog (Hh) signaling pathway was evaluated during the normal growth of cervical epithelium and in the presence of E6/E7 oncogenes and exogenous estradiol. Hh signaling activity was determined in live animals (i.e., Gli-Luc reporter levels) during the estrous cycle and was found to be higher in the cervical area during the major growth phases, proestrus-estrus, in comparison to the diestrus phase. The same pattern was observed in transgenic mice expressing the E6/E7 oncogenes, though with notably higher levels than in control mice. Adding estradiol also markedly increased Gli activity in the cervix and the skin. In agreement with the correlation between high bioluminescence and tissue growth in different context, cervical cell proliferation was reduced upon Hh signaling inhibition in mice. Treatment with itraconazole, a putative novel Hh inhibitor, at an early stage of cervical carcinogenesis, did not decrease Hh signaling but it did reduce growth. Therefore, Hh signaling likely contributes to cervical carcinogenesis and itraconazole is effective to reduce growth but by a mechanism involving additional signaling pathways.


Asunto(s)
Estradiol/farmacología , Proteínas Hedgehog/genética , Proteínas Oncogénicas Virales/fisiología , Proteínas E7 de Papillomavirus/fisiología , Neoplasias del Cuello Uterino/patología , Animales , Carcinogénesis/efectos de los fármacos , Carcinogénesis/genética , Modelos Animales de Enfermedad , Femenino , Células HeLa , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Ratones Transgénicos , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/metabolismo
6.
Radiographics ; 39(7): 2055-2068, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31603733

RESUMEN

During the past decade and a half, the most common cause of oropharyngeal squamous cell carcinoma (OPSCC) has shifted from tobacco and alcohol to the human papillomavirus (HPV). HPV-driven p16-positive OPSCC and tobacco-related OPSCC differ in their underlying molecular and genetic profiles, socioeconomic demographics, and response to treatment. HPV-related OPSCC tends to occur in younger patients and has a significantly better response to treatment and excellent prognosis. The stark contrast in prognosis-with around 90% overall 5-year survival for HPV-related p16-positive OPSCC and 40% for non-HPV-related p16-negative OPSCC-has prompted major changes in the eighth edition of the staging manual of the AJCC (American Joint Committee on Cancer). The past 10-15 years have also witnessed major advances in surgery, radiation therapy (RT), and systemic therapy. Minimally invasive surgery has come of age, with transoral robotic procedures and laser microsurgery. Intensity-modulated RT (IMRT) and more recently proton-beam RT have markedly improved the conformity of RT, with an ability to precisely target the cancer and cancer-bearing regions while sparing normal structures and significantly reducing long-term treatment-related morbidity. Progress in systemic therapy has come in the form of immunotherapy and targeted agents such as cetuximab. Owing to the better prognosis of HPV-driven OPSCC as well as the morbidity associated with treatment, de-escalation of therapy via multiple strategies is being explored. The article reviews the advances in diagnosis and multidisciplinary management of OPSCC in the HPV era.©RSNA, 2019.


Asunto(s)
Carcinoma de Células Escamosas/diagnóstico por imagen , Neoplasias Orofaríngeas/diagnóstico por imagen , Distribución por Edad , Consumo de Bebidas Alcohólicas/efectos adversos , Carcinoma de Células Escamosas/epidemiología , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/virología , Ciclo Celular , Quimioterapia Adyuvante , Ensayos Clínicos como Asunto , Terapia Combinada , Diagnóstico por Imagen/métodos , Manejo de la Enfermedad , Papillomavirus Humano 16/patogenicidad , Humanos , Inmunoterapia , Metástasis Linfática/diagnóstico por imagen , Estadificación de Neoplasias , Proteínas Oncogénicas Virales/fisiología , Procedimientos Quirúrgicos Orales , Neoplasias Orofaríngeas/epidemiología , Neoplasias Orofaríngeas/terapia , Neoplasias Orofaríngeas/virología , Proteínas E7 de Papillomavirus/fisiología , Infecciones por Papillomavirus/diagnóstico por imagen , Infecciones por Papillomavirus/epidemiología , Infecciones por Papillomavirus/virología , Pronóstico , Radioterapia Adyuvante , Proteínas Represoras/fisiología , Factores de Riesgo , Terapia Recuperativa , Fumar/efectos adversos
7.
Proc Natl Acad Sci U S A ; 113(33): 9333-8, 2016 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-27482104

RESUMEN

High-risk human papillomaviruses (HPVs) are causative agents of anogenital cancers and a fraction of head and neck cancers. The mechanisms involved in the progression of HPV neoplasias to cancers remain largely unknown. Here, we report that O-linked GlcNAcylation (O-GlcNAc) and O-GlcNAc transferase (OGT) were markedly increased in HPV-caused cervical neoplasms relative to normal cervix, whereas O-GlcNAcase (OGA) levels were not altered. Transduction of HPV16 oncogene E6 or E6/E7 into mouse embryonic fibroblasts (MEFs) up-regulated OGT mRNA and protein, elevated the level of O-GlcNAc, and promoted cell proliferation while reducing cellular senescence. Conversely, in HPV-18-transformed HeLa cervical carcinoma cells, inhibition of O-GlcNAc with a low concentration of a chemical inhibitor impaired the transformed phenotypes in vitro. We showed that E6 elevated c-MYC via increased protein stability attributable to O-GlcNAcylation on Thr58. Reduction of HPV-mediated cell viability by a high concentration of O-GlcNAc inhibitor was partially rescued by elevated c-MYC. Finally, knockdown of OGT or O-GlcNAc inhibition in HeLa cells or in TC-1 cells, a mouse cell line transformed by HPV16 E6/E7 and activated K-RAS, reduced c-MYC and suppressed tumorigenesis and metastasis. Thus, we have uncovered a mechanism for HPV oncoprotein-mediated transformation. These findings may eventually aid in the development of effective therapeutics for HPV-associated malignancies by targeting aberrant O-GlcNAc.


Asunto(s)
Carcinogénesis , N-Acetilglucosaminiltransferasas/fisiología , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras/fisiología , Neoplasias del Cuello Uterino/etiología , Animales , Línea Celular Tumoral , Femenino , Genes myc , Humanos , Ratones , Ratones Endogámicos C57BL , Proteínas E7 de Papillomavirus/fisiología , Neoplasias del Cuello Uterino/virología
8.
J Pathol ; 242(4): 448-462, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28497579

RESUMEN

In stratified epithelia such as the epidermis, homeostasis is maintained by the proliferation of cells in the lower epithelial layers and the concomitant loss of differentiated cells from the epithelial surface. These differentiating keratinocytes progressively stratify and form a self-regenerating multi-layered barrier that protects the underlying dermis. In such tissue, the continual loss and replacement of differentiated cells also limits the accumulation of oncogenic mutations within the tissue. Inactivating mutations in key driver genes, such as TP53 and NOTCH1, reduce the proportion of differentiating cells allowing for the long-term persistence of expanding mutant clones in the tissue. Here we show that through the expression of E6, HPV-16 prevents the early fate commitment of human keratinocytes towards differentiation and confers a strong growth advantage to human keratinocytes. When E6 is expressed either alone or with E7, it promotes keratinocyte proliferation at high cell densities, through the combined inactivation of p53 and Notch1. In organotypic raft culture, the activity of E6 is restricted to the basal layer of the epithelium and is enhanced during the progression from productive to abortive or transforming HPV-16 infection. Consistent with this, the expression of p53 and cleaved Notch1 becomes progressively more disrupted, and is associated with increased basal cell density and reduced commitment to differentiation. The expression of cleaved Notch1 is similarly disrupted also in HPV-16-positive cervical lesions, depending on neoplastic grade. When taken together, these data depict an important role of high-risk E6 in promoting the persistence of infected keratinocytes in the basal and parabasal layers through the inactivation of gene products that are commonly mutated in non-HPV-associated neoplastic squamous epithelia. © 2017 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/fisiología , Infecciones por Papillomavirus/metabolismo , Receptor Notch1/metabolismo , Proteínas Represoras/fisiología , Neoplasias del Cuello Uterino/virología , Diferenciación Celular/fisiología , División Celular , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Transformación Celular Viral/fisiología , Progresión de la Enfermedad , Femenino , Humanos , Queratinocitos/patología , Queratinocitos/virología , Clasificación del Tumor , Infecciones por Papillomavirus/patología , ARN Mensajero/genética , Receptor Notch1/deficiencia , Receptor Notch1/genética , Proteína p53 Supresora de Tumor/metabolismo , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología
9.
J Virol ; 90(23): 10629-10641, 2016 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-27654294

RESUMEN

The human papillomavirus (HPV) capsid protein L2 is essential for viral entry. To gain a deeper understanding of the role of L2, we searched for novel cellular L2-interacting proteins. A yeast two-hybrid analysis uncovered the actin-depolymerizing factor gelsolin, the membrane glycoprotein dysadherin, the centrosomal protein 68 (Cep68), and the cytoskeletal adaptor protein obscurin-like 1 protein (OBSL1) as putative L2 binding molecules. Pseudovirus (PsV) infection assays identified OBSL1 as a host factor required for gene transduction by three oncogenic human papillomavirus types, HPV16, HPV18, and HPV31. In addition, we detected OBSL1 expression in cervical tissue sections and noted the involvement of OBSL1 during gene transduction of primary keratinocytes by HPV16 PsV. Complex formation of HPV16 L2 with OBSL1 was demonstrated in coimmunofluorescence and coimmunoprecipitation studies after overexpression of L2 or after PsV exposure. We observed a strong colocalization of OBSL1 with HPV16 PsV and tetraspanin CD151 at the plasma membrane, suggesting a role for OBSL1 in viral endocytosis. Indeed, viral entry assays exhibited a reduction of viral endocytosis in OBSL1-depleted cells. Our results suggest OBSL1 as a novel L2-interacting protein and endocytosis factor in HPV infection. IMPORTANCE: Human papillomaviruses infect mucosal and cutaneous epithelia, and the high-risk HPV types account for 5% of cancer cases worldwide. As recently discovered, HPV entry occurs by a clathrin-, caveolin-, and dynamin-independent endocytosis via tetraspanin-enriched microdomains. At present, the cellular proteins involved in the underlying mechanism of this type of endocytosis are under investigation. In this study, the cytoskeletal adaptor OBSL1 was discovered as a previously unrecognized interaction partner of the minor capsid protein L2 and was identified as a proviral host factor required for HPV16 endocytosis into target cells. The findings of this study advance the understanding of a so far less well-characterized endocytic pathway that is used by oncogenic HPV subtypes.


Asunto(s)
Proteínas de la Cápside/fisiología , Proteínas del Citoesqueleto/fisiología , Papillomavirus Humano 16/fisiología , Proteínas Oncogénicas Virales/fisiología , Proteínas de la Cápside/genética , Línea Celular , Proteínas del Citoesqueleto/antagonistas & inhibidores , Proteínas del Citoesqueleto/genética , Endocitosis/fisiología , Técnicas de Silenciamiento del Gen , Células HeLa , Interacciones Huésped-Patógeno/fisiología , Papillomavirus Humano 16/genética , Humanos , Queratinocitos/fisiología , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/etiología , Técnicas del Sistema de Dos Híbridos , Internalización del Virus
10.
J Virol ; 89(5): 2857-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25540383

RESUMEN

UNLABELLED: Many of the small DNA tumor viruses encode transforming proteins that function by targeting critical cellular pathways involved in cell proliferation and survival. In this study, we have examined whether some of the functions of the polyomavirus small T antigens (ST) are shared by the E6 and E7 oncoproteins of two oncogenic papillomaviruses. Using three different assays, we have found that E7 can provide some simian virus 40 (SV40) or murine polyomavirus (PyV) ST functions. Both human papillomavirus 16 (HPV16) and bovine papillomavirus (BPV1) E7 proteins are capable of partially substituting for SV40 ST in a transformation assay that also includes SV40 large T antigen, the catalytic subunit of cellular telomerase, and oncogenic Ras. Like SV40 ST, HPV16 E7 has the ability to override a quiescence block induced by mitogen deprivation. Like PyV ST, it also has the ability to inhibit myoblast differentiation. At least two of these activities are dependent upon the interaction of HPV16 E7 with retinoblastoma protein family members. For small T antigens, interaction with PP2A is needed for each of these functions. Even though there is no strong evidence that E6 or E7 share the ability of small T to interact with PP2A, E7 provides these functions related to cellular transformation. IMPORTANCE: DNA tumor viruses have provided major insights into how cancers develop. Some viruses, like the human papillomaviruses, can cause cancer directly. Both the papillomaviruses and the polyomaviruses have served as tools for understanding pathways that are often perturbed in cancer. Here, we have compared the functions of transforming proteins from several DNA tumor viruses, including two papillomaviruses and two polyomaviruses. We tested the papillomavirus E6 and E7 oncoproteins in three functional assays and found that E7 can provide some or all of the functions of the SV40 small T antigen, another well-characterized oncoprotein, in two of these assays. In a third assay, papillomavirus E7 has the same effect as the murine polyomavirus small T protein. In summary, we report several new functions for the papillomavirus E7 proteins, which will contribute new insights into the roles of viruses in cancer and the cellular pathways they perturb in carcinogenesis.


Asunto(s)
Antígenos Transformadores de Poliomavirus/metabolismo , Transformación Celular Viral , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Proteínas Represoras/metabolismo , Prueba de Complementación Genética , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/fisiología , Virus 40 de los Simios/genética , Virus 40 de los Simios/fisiología
11.
PLoS Pathog ; 10(5): e1004102, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24788832

RESUMEN

Adenoviruses infect epithelial cells lining mucous membranes to cause acute diseases in people. They are also utilized as vectors for vaccination and for gene and cancer therapy, as well as tools to discover mechanisms of cancer due to their tumorigenic potential in experimental animals. The adenovirus E4-ORF1 gene encodes an oncoprotein that promotes viral replication, cell survival, and transformation by activating phosphatidylinositol 3-kinase (PI3K). While the mechanism of activation is not understood, this function depends on a complex formed between E4-ORF1 and the membrane-associated cellular PDZ protein Discs Large 1 (Dlg1), a common viral target having both tumor suppressor and oncogenic functions. Here, we report that in human epithelial cells, E4-ORF1 interacts with the regulatory and catalytic subunits of PI3K and elevates their levels. Like PI3K activation, PI3K protein elevation by E4-ORF1 requires Dlg1. We further show that Dlg1, E4-ORF1, and PI3K form a ternary complex at the plasma membrane. At this site, Dlg1 also co-localizes with the activated PI3K effector protein Akt, indicating that the ternary complex mediates PI3K signaling. Signifying the functional importance of the ternary complex, the capacity of E4-ORF1 to induce soft agar growth and focus formation in cells is ablated either by a mutation that prevents E4-ORF1 binding to Dlg1 or by a PI3K inhibitor drug. These results demonstrate that E4-ORF1 interacts with Dlg1 and PI3K to assemble a ternary complex where E4-ORF1 hijacks the Dlg1 oncogenic function to relocate cytoplasmic PI3K to the membrane for constitutive activation. This novel mechanism of Dlg1 subversion by adenovirus to dysregulate PI3K could be used by other pathogenic viruses, such as human papillomavirus, human T-cell leukemia virus type 1, and influenza A virus, which also target Dlg1 and activate PI3K in cells.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Membrana Celular/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Oncogénicas Virales/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Infecciones por Adenoviridae/metabolismo , Infecciones por Adenoviridae/patología , Transformación Celular Viral/genética , Células Cultivadas , Homólogo 1 de la Proteína Discs Large , Activación Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/virología , Células HeLa , Interacciones Huésped-Patógeno , Humanos , Proteínas Oncogénicas Virales/metabolismo , Unión Proteica , Transporte de Proteínas
12.
Carcinogenesis ; 36(11): 1397-406, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26354779

RESUMEN

Human papillomavirus (HPV) is the causative agent of a subgroup of head and neck cancer characterized by an intrinsic radiosensitivity. HPV initiates cellular transformation through the activity of E6 and E7 proteins. E6 and E7 expression is necessary but not sufficient to transform the host cell, as genomic instability is required to acquire the malignant phenotype in HPV-initiated cells. This study reveals a key role played by oxidative stress in promoting genomic instability and radiosensitivity in HPV-positive head and neck cancer. By employing an isogenic human cell model, we observed that expression of E6 and E7 is sufficient to induce reactive oxygen species (ROS) generation in head and neck cancer cells. E6/E7-induced oxidative stress is mediated by nicotinamide adenine dinucleotide phosphate oxidases (NOXs) and causes DNA damage and chromosomal aberrations. This mechanism for genomic instability distinguishes HPV-positive from HPV-negative tumors, as we observed NOX-induced oxidative stress in HPV-positive but not HPV-negative head and neck cancer cells. We identified NOX2 as the source of HPV-induced oxidative stress as NOX2 silencing significantly reduced ROS generation, DNA damage and chromosomal aberrations in HPV-positive cells. Due to their state of chronic oxidative stress, HPV-positive cells are more susceptible to DNA damage induced by ROS and ionizing radiation (IR). Furthermore, exposure to IR results in the formation of complex lesions in HPV-positive cells as indicated by the higher amount of chromosomal breakage observed in this group of cells. These results reveal a novel mechanism for sustaining genomic instability in HPV-positive head and neck tumors and elucidate its contribution to their intrinsic radiosensitivity.


Asunto(s)
Daño del ADN , Neoplasias de Cabeza y Cuello/virología , Glicoproteínas de Membrana/fisiología , NADPH Oxidasas/fisiología , Proteínas Oncogénicas Virales/fisiología , Estrés Oxidativo , Proteínas E7 de Papillomavirus/fisiología , Proteínas Represoras/fisiología , Carcinogénesis/metabolismo , Línea Celular Tumoral , Susceptibilidad a Enfermedades , Inestabilidad Genómica , Neoplasias de Cabeza y Cuello/genética , Humanos , NADPH Oxidasa 2 , Especies Reactivas de Oxígeno/metabolismo
13.
Biochem Biophys Res Commun ; 460(3): 555-60, 2015 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-25800874

RESUMEN

Recent cancer genomics studies have identified mutation patterns characteristic of APOBEC3B (A3B) in multiple cancers, including cervical cancer, which is caused by human papillomavirus (HPV) infection. A3B expression is upregulated by HPV E6/E7 oncoproteins, implying a crucial role for A3B upregulation in HPV-induced carcinogenesis. Here, we explored the molecular mechanisms underlying the activation of the A3B promoter by E6. Luciferase reporter assays with a series of deleted fragments of the human A3B promoter in normal immortalized human keratinocytes (NIKS) identified two functional regions in the promoter: the distal region (from -200 to -51), which is required for basal promoter activity, and the proximal region (from +1 to +45), which exerts an inhibitory effect on gene expression. Each promoter region was found to contain an E6-responsive element(s). Disruption of an AT-rich motif located between +10 and +16 abrogated the proximal-region-mediated activation of the A3B promoter by E6. DNA pull-down assays revealed that a cellular zinc-finger protein, ZNF384, binds to the AT-rich motif in the A3B promoter, and chromatin immunoprecipitation assays confirmed that ZNF384 binds to the A3B promoter in cells. ZNF384 knockdown reduced the A3B mRNA levels in NIKS expressing E6, but not in the parental NIKS, indicating that ZNF384 contributes to A3B upregulation by E6, but not to basal A3B expression. The exogenous expression of ZNF384 led to the activation of the A3B promoter in NIKS. Collectively, these results indicate that E6 activates the A3B promoter through the distal and proximal regions, and that ZNF384 is required for the proximal-region-mediated activation of A3B.


Asunto(s)
Citidina Desaminasa/genética , Proteínas Oncogénicas Virales/fisiología , Regiones Promotoras Genéticas , Proteínas Represoras/fisiología , Secuencia de Bases , Línea Celular Transformada , Citidina Desaminasa/metabolismo , ADN , Técnicas de Silenciamiento del Gen , Humanos , Antígenos de Histocompatibilidad Menor , Datos de Secuencia Molecular , ARN Interferente Pequeño , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores/genética , Transactivadores/metabolismo
14.
Biochem Biophys Res Commun ; 457(3): 295-9, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25576866

RESUMEN

Apolipoprotein B mRNA editing enzyme, catalytic polypeptide-like (APOBEC) proteins are cellular DNA/RNA-editing enzymes that play pivotal roles in the innate immune response to viral infection. APOBEC3 (A3) proteins were reported to hypermutate the genome of human papillomavirus 16 (HPV16), the causative agent of cervical cancer. However, hypermutation did not affect viral DNA maintenance, leaving the exact role of A3 against HPV infection elusive. Here we examine whether A3 proteins affect the virion assembly using an HPV16 pseudovirion (PsV) production system, in which PsVs are assembled from its capsid proteins L1/L2 encapsidating a reporter plasmid in 293FT cells. We found that co-expression of A3A or A3C in 293FT cells greatly reduced the infectivity of PsV. The reduced infectivity of PsV assembled in the presence of A3A, but not A3C, was attributed to the decreased copy number of the encapsidated reporter plasmid. On the other hand, A3C, but not A3A, efficiently bound to L1 in co-immunoprecipitation assays, which suggests that this physical interaction may lead to reduced infectivity of PsV assembled in the presence of A3C. These results provide mechanistic insights into A3s' inhibitory effects on the assembly phase of the HPV16 virion.


Asunto(s)
Citidina Desaminasa/fisiología , Papillomavirus Humano 16/patogenicidad , Proteínas/fisiología , Proteínas de la Cápside/fisiología , Citidina Desaminasa/genética , Femenino , Genoma Viral , Células HEK293 , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Proteínas Oncogénicas Virales/fisiología , Unión Proteica , Proteínas/genética , Virión/genética , Virión/patogenicidad , Virión/fisiología , Virulencia , Ensamble de Virus
15.
J Cell Mol Med ; 18(1): 181-6, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24286246

RESUMEN

High-risk human papillomavirus (HPV)-positive head and neck squamous cell carcinomas (HNSCCs) are highly invasive; however the identity of downstream effectors responsible for their aggressive phenotype remains underinvestigated. Here, we report that HPV-mediated up-regulation of heparanase enzyme can provide mechanistic explanation for augmented invasiveness of HPV-positive HNSCCs. Heparanase is the sole mammalian enzyme (endo-ß-d-glucuronidase) degrading heparan sulphate glycosaminoglycan, key polysaccharide of the extracellular matrix. Cleavage of heparan sulphate by heparanase leads to disassembly of extracellular barriers, enabling local invasion and metastatic spread of the tumour, and releases heparan sulphate-bound growth factors from the extracellular depots. Heparanase is tightly implicated in head and neck cancer progression; yet, molecular mechanisms underlying transcriptional activation of the heparanase gene in HNSCC are largely unknown. We found that HPV16 oncogene E6 is capable of inducing overexpression of heparanase in HNSCC. Notably, radiation treatment dose-dependently suppresses E6-induced heparanase expression in vitro. Our results provide the first evidence for a functional involvement of HPV in heparanase induction in head and neck tumourigenesis and, given ongoing clinical testing of several heparanase-inhibiting compounds, offer important avenue for future therapeutic exploration in HNSCC, as well as other HPV-associated malignancies (i.e. cervical carcinoma).


Asunto(s)
Carcinoma de Células Escamosas/enzimología , Glucuronidasa/genética , Neoplasias de Cabeza y Cuello/enzimología , Papillomavirus Humano 16/enzimología , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras/fisiología , Carcinoma de Células Escamosas/virología , Línea Celular Tumoral , Inducción Enzimática , Glucuronidasa/metabolismo , Neoplasias de Cabeza y Cuello/virología , Interacciones Huésped-Patógeno , Humanos , Transcripción Genética
16.
Mol Carcinog ; 53(8): 648-58, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23475592

RESUMEN

A large number of epidemiological studies have linked a common single-nucleotide polymorphism (SNP) in the human p53 gene to risk for developing a variety of cancers. This SNP encodes either an arginine or proline at position 72 (R72P) of the p53 protein, which can alter the apoptotic activity of p53 via transcriptional and non-transcriptional mechanisms. This SNP has also been reported to modulate the development of human papilloma virus (HPV)-driven cancers through differential targeting of the p53 variant proteins by the E6 viral oncoprotein. Mouse models for the p53 R72P polymorphism have recently been developed but a role for this SNP in modifying cancer risk in response to viral and chemical carcinogens has yet to be established experimentally. Here, we demonstrate that the p53 R72P polymorphism modulates the hyperprolferative, apoptotic and inflammatory phenotypes caused by expression of the HPV16 E6 and E7 oncoproteins. Moreover, the R72P SNP also modifies the carcinogenic response to the chemical carcinogen 4NQO, in the presence and absence of the HPV16 transgene. Our findings confirm several human epidemiological studies associating the codon 72 proline variant with increased risk for certain cancers but also suggest that there are tissue-specific differences in how the R72P polymorphism influences the response to environmental carcinogens.


Asunto(s)
4-Nitroquinolina-1-Óxido/toxicidad , Neoplasias Esofágicas/etiología , Interacción Gen-Ambiente , Inflamación/etiología , Neoplasias de la Boca/etiología , Infecciones por Papillomavirus/etiología , Polimorfismo de Nucleótido Simple/genética , Proteína p53 Supresora de Tumor/fisiología , Animales , Western Blotting , Carcinógenos/toxicidad , Células Cultivadas , ADN Viral/genética , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/patología , Femenino , Humanos , Técnicas para Inmunoenzimas , Inflamación/metabolismo , Inflamación/patología , Queratinocitos/citología , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Ratones , Ratones Transgénicos , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Proteínas Oncogénicas Virales/fisiología , Papillomaviridae/genética , Proteínas E7 de Papillomavirus/fisiología , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/patología , Fenotipo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Biochim Biophys Acta ; 1819(7): 820-5, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22306660

RESUMEN

Persistent viruses need mechanisms to protect their genomes from cellular defenses and to ensure that they are efficiently propagated to daughter host cells. One mechanism by which papillomaviruses achieve this is through the association of viral genomes with host chromatin, mediated by the viral E2 tethering protein. Association of viral DNA with regions of active host chromatin ensures that the virus remains transcriptionally active and is not relegated to repressed heterochromatin. In addition, viral genomes are tethered to specific regions of host mitotic chromosomes to efficiently partition their DNA to daughter cells. Vegetative viral DNA replication also initiates at specific regions of host chromatin, where the viral E1 and E2 proteins initiate a DNA damage response that recruits cellular DNA damage and repair proteins to viral replication foci for efficient viral DNA synthesis. Thus, these small viruses have capitalized on interactions with chromatin to efficiently target their genomes to beneficial regions of the host nucleus. This article is part of a Special Issue entitled: Chromatin in time and space.


Asunto(s)
Alphapapillomavirus/fisiología , Cromatina/genética , Infecciones por Papillomavirus/virología , Alphapapillomavirus/genética , Animales , Segregación Cromosómica , Cromosomas Humanos , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/fisiología , Genoma Viral , Interacciones Huésped-Patógeno , Humanos , Inmunidad Innata , Proteínas Oncogénicas Virales/metabolismo , Proteínas Oncogénicas Virales/fisiología , Infecciones por Papillomavirus/inmunología , Internalización del Virus , Replicación Viral
18.
Biochem Biophys Res Commun ; 435(3): 339-44, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23628416

RESUMEN

Human papillomavirus (HPV) 16 is among the most important etiological factors in many human cancers, including head and neck squamous cell carcinomas (HNSCCs) not associated with alcohol or tobacco use. HPV16-E6 and E7 oncoproteins target intracellular signaling networks, altering key molecular and cellular events during tumor progression. The present study investigates the role of HPV16-E6 and E7 oncogenes on the epithelial-mesenchymal transition (EMT), a cellular process thought to be critical for tumor cell invasion and metastasis. Using the epithelial MDCK cell line as an in vitro model, we show that the stable expression of HPV16-E6 or E7 induces morphological conversion from cobblestone-shaped epithelium to spindle-shaped mesenchyme-like phenotype. Consistent with these morphological changes, both E6 and E7 induce expression of the EMT-activating transcriptional factors Slug, Twist, ZEB1 and ZEB2, especially ZEBs, accompanied with switch from epithelial to mesenchymal markers. Importantly, E6 and E7 expression results in induction of the migratory and invasive potential, a functional hallmark of EMT. When we examined the association between HPV16 and the EMT signature in HNSCC cell lines derived from head and neck cancer patients, we found a correlation between HPV16 positivity and the expression of EMT transcription factor ZEB1. Taken together, our findings suggest HPV16 induces EMT-like processes via induction of the EMT transcription factors which may contribute to tumor progression and metastasis.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Papillomavirus Humano 16/patogenicidad , Proteínas Oncogénicas Virales/fisiología , Proteínas E7 de Papillomavirus/fisiología , Proteínas Represoras/fisiología , Animales , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Perros , Neoplasias de Cabeza y Cuello/etiología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/genética , Papillomavirus Humano 16/fisiología , Humanos , Invasividad Neoplásica/genética , Invasividad Neoplásica/patología , Proteínas Oncogénicas/genética , Proteínas Oncogénicas/fisiología , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Proteínas Represoras/genética , Carcinoma de Células Escamosas de Cabeza y Cuello
19.
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
20.
Nat Rev Cancer ; 2(1): 59-65, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11902586

RESUMEN

Tumour-associated viruses produce antigens that, on the face of it, are ideal targets for immunotherapy. Unfortunately, these viruses are experts at avoiding or subverting the host immune response. Cervical-cancer-associated human papillomavirus (HPV) has a battery of immune-evasion mechanisms at its disposal that could confound attempts at HPV-directed immunotherapy. Other virally associated human cancers might prove similarly refractive to immuno-intervention unless we learn how to circumvent their strategies for immune evasion.


Asunto(s)
Papillomaviridae/inmunología , Infecciones por Papillomavirus/inmunología , Neoplasias del Cuello Uterino/inmunología , Animales , Presentación de Antígeno , Células Presentadoras de Antígenos/inmunología , Antígenos Virales/inmunología , Cápside/inmunología , Transformación Celular Viral , Codón/genética , Células Dendríticas/inmunología , Femenino , Regulación Viral de la Expresión Génica , Código Genético , Humanos , Tolerancia Inmunológica , Vigilancia Inmunológica , Interferones/fisiología , Queratinocitos/virología , Ratones , Ratones Transgénicos , Imitación Molecular , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/fisiología , Papillomaviridae/genética , Neoplasias del Cuello Uterino/virología , Proteínas Virales/genética , Proteínas Virales/inmunología , Proteínas Virales/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA