Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Oncogene ; 34(7): 868-77, 2015 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-24608431

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common malignancy worldwide, and patient outcomes using current treatments remain poor. Tumor development is etiologically associated with tobacco or alcohol use and/or human papillomavirus (HPV) infection. HPV-positive HNSCCs, which frequently harbor wild-type p53, carry a more favorable prognosis and are a biologically distinct subgroup when compared with their HPV-negative counterparts. HPV E7 induces expression of the human DEK gene, both in vitro and in vivo. In keratinocytes, DEK overexpression is sufficient for causing oncogenic phenotypes in the absence of E7. Conversely, DEK loss results in cell death in HPV-positive cervical cancer cells at least in part through p53 activation, and Dek knockout mice are relatively resistant to the development of chemically induced skin papillomas. Despite the established oncogenic role of DEK in HPV-associated cervical cancer cell lines and keratinocytes, a functional role of DEK has not yet been explored in HNSCC. Using an established transgenic mouse model of HPV16 E7-induced HNSCC, we demonstrate that Dek is required for optimal proliferation of E7-transgenic epidermal cells and for the growth of HNSCC tumors. Importantly, these studies also demonstrate that DEK protein is universally upregulated in both HPV-positive and -negative human HNSCC tumors relative to adjacent normal tissue. Furthermore, DEK knockdown inhibited the proliferation of HPV-positive and -negative HNSCC cells, establishing a functional role for DEK in human disease. Mechanistic studies reveal that attenuated HNSCC cell growth in response to DEK loss was associated with reduced expression of the oncogenic p53 family member, ΔNp63. Exogenous ΔNp63 expression rescued the proliferative defect in the absence of DEK, thereby establishing a functional DEK-ΔNp63 oncogenic pathway that promotes HNSCC. Taken together, our data demonstrate that DEK stimulates HNSCC cellular growth and identify ΔNp63 as a novel DEK effector.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Proliferación Celular , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Papillomavirus Humano 16/metabolismo , Proteínas Oncogénicas/metabolismo , Proteínas E7 de Papillomavirus/metabolismo , Infecciones por Papillomavirus/metabolismo , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/genética , Neoplasias de Cabeza y Cuello , Papillomavirus Humano 16/genética , Humanos , Queratinocitos/metabolismo , Queratinocitos/patología , Ratones , Ratones Noqueados , Proteínas Oncogénicas/genética , Proteínas E7 de Papillomavirus/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
2.
Oncogene ; 33(26): 3383-91, 2014 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-24013229

RESUMEN

Fanconi anemia (FA) is a rare genetic disorder caused by defects in a DNA damage repair system, the FA pathway. FA patients frequently develop squamous cell carcinoma (SCC) at sites that are associated with human papillomavirus (HPV)-driven cancer including the female reproductive tract. To assess experimentally whether FA deficiency increases susceptibility to HPV-associated cervical/vaginal cancer, we monitored cancer incidence in the female lower reproductive tract of FA-deficient mice expressing HPV16 oncogenes, E6 and/or E7. FA deficiency specifically increased the incidence of cancers in mice expressing E7; but this effect was not observed in mice just expressing E6. We also observed that E7, but not E6, induced DNA damage as scored by induction of γ-H2AX and 53BP1 (p53 binding protein 1) nuclear foci, and this induction was heightened in FA-deficient tissue. Finally, we discovered that this induction of DNA damage responses was recapitulated in mice deficient in expression of 'pocket' proteins, pRb, p107 and p130, which are established targets of E7. Our findings support the hypothesis that E7 induces cancer by causing DNA damage at least in part through the inactivation of pocket proteins. This hypothesis explains why a deficiency in DNA damage repair would increase susceptibility to E7-driven cancer.


Asunto(s)
Proteínas del Grupo de Complementación de la Anemia de Fanconi/genética , Papillomavirus Humano 16/genética , Proteínas Oncogénicas Virales/genética , Proteínas E7 de Papillomavirus/genética , Proteínas Represoras/genética , Neoplasias del Cuello Uterino/genética , Animales , Carcinoma de Células Escamosas/genética , Proliferación Celular , Daño del ADN/genética , Anemia de Fanconi/genética , Femenino , Predisposición Genética a la Enfermedad , Histonas/biosíntesis , Ratones , Ratones Noqueados , Proteínas Oncogénicas Virales/biosíntesis , Proteínas E7 de Papillomavirus/biosíntesis , Infecciones por Papillomavirus/genética , Proteínas Represoras/biosíntesis , Proteína de Retinoblastoma/genética , Proteína p107 Similar a la del Retinoblastoma/genética , Proteína p130 Similar a la del Retinoblastoma/genética , Proteína p53 Supresora de Tumor/biosíntesis
3.
Virology ; 433(2): 337-45, 2012 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-22980503

RESUMEN

HPV16 E7 oncoprotein expression in K14E7 transgenic mice induces cervical cancer after 6 months of treatment with the co-carcinogen 17ß-estradiol. In untreated mice, E7 also induces skin tumors late in life albeit at low penetrance. These findings indicate that E7 alters cellular functions in cervix and skin so as to predispose these organs to tumorigenesis. Using microarrays, we determined the global genes expression profile in cervical and skin tissue of young adult K14E7 transgenic mice without estrogen treatment. In these tissues, the E7 oncoprotein altered the transcriptional pattern of genes involved in several biological processes including signal transduction, transport, metabolic process, cell adhesion, apoptosis, cell differentiation, immune response and inflammatory response. Among the E7-dysregulated genes were ones not previously known to be involved in cervical neoplasia including DMBT1, GLI1 and 17ßHSD2 in cervix, as well as MMP2, 12, 14, 19 and 27 in skin.


Asunto(s)
Papillomavirus Humano 16/genética , Papillomavirus Humano 16/patogenicidad , Proteínas E7 de Papillomavirus/genética , Proteínas E7 de Papillomavirus/fisiología , Animales , Secuencia de Bases , Cuello del Útero/metabolismo , Cuello del Útero/virología , Modelos Animales de Enfermedad , Femenino , Genes Virales , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/fisiología , Papillomavirus Humano 16/fisiología , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Infecciones por Papillomavirus/etiología , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/virología , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Viral/genética , ARN Viral/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Piel/metabolismo , Piel/virología , Transcriptoma , Neoplasias del Cuello Uterino/etiología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/virología
4.
Oncogene ; 29(23): 3435-45, 2010 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-20383192

RESUMEN

Persistent infection with high-risk human papillomaviruses (HPVs), especially type 16 has been undeniably linked to cervical cancer. The Asian-American (AA) variant of HPV16 is more common in the Americas than the prototype in cervical cancer. The different prevalence is based on three amino acid changes within the E6 protein denoted Q14H/H78Y/L83V. To investigate the mechanism(s) behind this observation, both E6 proteins, in the presence of E7, were evaluated for their ability to extend the life span of and transform primary human foreskin keratinocytes (PHFKs). Long-term cell culture studies resulted in death at passage 9 of vector-transduced PHFKs (negative control), but survival of both E6 PHFKs to passage 65 (and beyond). Compared with E6/E7 PHFKs, AA/E7 PHFKs were significantly faster dividing, developed larger cells in monolayer cultures, showed double the epithelial thickness and expressed cytokeratin 10 when grown as organotypic raft cultures. Telomerase activation and p53 inactivation, two hallmarks of immortalization, were not significantly different between the two populations. Both were resistant to anoikis at later passages, but only AA/E7 PHFKs acquired the capacity for in vitro transformation. Proteomic analysis revealed markedly different protein patterns between E6/E7 and AA/E7, particularly with respect to key cellular metabolic enzymes. Our results provide new insights into the reasons underlying the greater prevalence of the AA variant in cervical cancer as evidenced by characteristics associated with higher oncogenic potential.


Asunto(s)
Transformación Celular Neoplásica , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Neoplasias del Cuello Uterino/virología , Anoicis , Diferenciación Celular , Línea Celular Tumoral , Proliferación Celular , Daño del ADN , Activación Enzimática , Femenino , Humanos , Queratinocitos/metabolismo , Proteínas E7 de Papillomavirus/genética , Telomerasa/metabolismo , Proteína p53 Supresora de Tumor/fisiología
5.
Oncogene ; 26(23): 3321-8, 2007 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-17130828

RESUMEN

High-risk human papillomaviruses are the causative agents of cervical and other anogenital cancers. In these cancers, two viral oncogenes, E6 and E7, are expressed. E6 is best known for its ability to inactivate the tumor suppressor p53, which is thought to arise through ubiquitin-mediated degradation of p53 and involve a ternary complex between E6, p53 and the E3 ligase, E6AP. In mice transgenic for wild-type HPV16 E6, its expression leads to epithelial hyperplasia and an abrogation of normal cellular responses to DNA damage. Whereas only the latter phenotype is dependent upon E6's inactivation of p53, both are reduced in transgenic mice expressing an E6 mutant severely reduced in its binding to E6AP and other cellular proteins that bind E6 through a shared alpha-helix motif. Here, we investigated whether E6AP is required for the induction of the above phenotypes through the use of both E6AP-mutant and E6AP-null mice. E6, in the absence of E6AP retains an ability to induce epithelial hyperplasia, abrogate DNA damage responses and inhibit the induction of p53 protein following exposure to ionizing radiation. We conclude that E6 is able to induce both p53-dependent and p53-independent phenotypes through E6AP-independent pathways in the mouse.


Asunto(s)
Epidermis/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Fenotipo , Proteínas Represoras/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Daño del ADN , Lisina/genética , Lisina/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Oncogénicas Virales/genética , Proteínas Represoras/genética , Ubiquitina-Proteína Ligasas/metabolismo
6.
J Virol ; 75(16): 7712-6, 2001 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-11462043

RESUMEN

Primary human keratinocytes with ectopic expression of high-risk human papillomavirus (HPV) E6 and E7 oncoproteins display abnormal centrosome numbers, multipolar mitoses, and aneusomy. However, it has not been explored whether these abnormalities can occur in cells containing HPV episomes where E6 and E7 expression is under viral transcriptional control. Here, we demonstrate that centrosome abnormalities and genomic instability occur in organotypic raft cultures of human keratinocytes with episomal HPV-16 even at low copy numbers. We conclude that HPV-16 DNA, when maintained as an episome, can disturb centrosome homeostasis and subvert genomic integrity of the host cell during early stages of the viral infection.


Asunto(s)
Centrómero/genética , Fragilidad Cromosómica , Queratinocitos/virología , Papillomaviridae , Infecciones por Papillomavirus/genética , Infecciones Tumorales por Virus/genética , Células Cultivadas , Centrómero/patología , Humanos , Queratinocitos/fisiología , Plásmidos
7.
J Virol ; 75(4): 1736-43, 2001 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11160671

RESUMEN

The human immunodeficiency virus type 1 (HIV-1) Tat protein has been reported to transactivate several cellular genes, including the potent chemotactic factor interleukin-8 (IL-8). Consistent with these in vitro assays, elevated levels of IL-8 protein are found in the serum of HIV-infected individuals. We now extend these observations by demonstrating that Tat induction of IL-8 is linked to the cell cycle. Cells that constitutively express the Tat(1-86) protein (eTat) and control cells (pCEP) were reversibly blocked at the G(1)/S border with hydroxyurea or thymidine. The cells were subsequently released, and IL-8 expression was monitored by RNase protection assays and enzyme-linked immunosorbent assay (ELISA). RNase protection assays demonstrated that IL-8 mRNA expression is transiently induced, approximately fourfold, as the Tat-expressing cells enter S phase. Consistent with the RNase protection assay, an increase in IL-8 protein was observed in the cell supernatant using an IL-8 ELISA. Similar experiments were performed following a reversible block at the G(2)/M border with nocodazole and release into G(1). Using the RNase protection assay and ELISA, little or no increase in IL-8 expression was observed during G(1). Using gel shift as well as an immobilized DNA binding assay, we demonstrate that the increase in IL-8 gene expression correlates with a specific increase in p65 NF-kappa B binding activity only in the nucleus of the Tat-expressing cells. Moreover, the CREB-binding protein coactivator is present in the complex in the Tat cell line. Finally, we demonstrate that the presence of the proteasome inhibitor MG-132 inhibits the induction of NF-kappa B binding, as well as IL-8 expression, supporting the role of NF-kappa B.


Asunto(s)
Ciclo Celular/fisiología , Regulación de la Expresión Génica , Productos del Gen tat/metabolismo , VIH-1/metabolismo , Interleucina-8/biosíntesis , Interleucina-8/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Productos del Gen tat/genética , VIH-1/genética , Células HeLa , Humanos , Hidroxiurea/farmacología , Leupeptinas/farmacología , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Fase S , Transcripción Genética , Transfección , Productos del Gen tat del Virus de la Inmunodeficiencia Humana
9.
Virology ; 276(2): 304-14, 2000 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-11040122

RESUMEN

The minor capsid protein L2 of papillomaviruses (PVs) likely plays a role in the selective encapsidation of PV DNA in viral capsids and in the infectivity of PV virions. The L2 protein also can cause the relocalization of the PV early protein, E2TA, to nuclear subdomains known as promyelocytic leukemia oncogenic domains (PODs) in which it is localized. E2TA is a transcriptional transactivator that also plays a critical role in viral DNA replication. In this study, we investigated whether L2, in causing the relocalization of E2TA, alters the activities of E2TA. We provide evidence that L2 inhibits the transcriptional transactivation function of E2, but it does not specifically inhibit the capacity of E2 to support viral DNA replication. We also investigated whether the colocalization of E2 and L2 to PODs and the ability of L2 to inhibit the transcriptional transactivation activity of E2TA might be mediated through a direct interaction between these two proteins. Using an in vitro protein-protein association assay, we found that L2 binds to E2TA. Two regions in E2TA were found to mediate this interaction. One of those domains is present in an alternative E2 gene product, E2TR, which is an antagonist to E2TA. Here we show that the L2 protein also relocalizes the E2 transcriptional repressor, E2TR, to the nuclear subdomains. These data suggest that the ability of L2 to relocalize E2 proteins to PODs is mediated through a direct interaction with L2.


Asunto(s)
Papillomavirus Bovino 1/metabolismo , Proteínas de la Cápside , Cápside/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Virales/metabolismo , Animales , Núcleo Celular/metabolismo , Núcleo Celular/virología , Células Cultivadas , Replicación del ADN , ADN Viral/biosíntesis , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Represoras/metabolismo , Transactivadores/metabolismo , Activación Transcripcional
10.
J Virol ; 74(15): 6866-74, 2000 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10888626

RESUMEN

We have analyzed the functional activity of the p53 tumor suppressor in human T-cell lymphotropic virus type 2 (HTLV-2)-transformed cells. Abundant levels of the p53 protein were detected in both HTLV-2A and -2B virus-infected cell lines. The p53 was functionally inactive, however, both in transient-transfection assays using a p53 reporter plasmid and in induction of p53-responsive genes in response to gamma irradiation. We further investigated HTLV-2A Tax and HTLV-2B Tax effects on p53 activity. Interestingly, although Tax-2A and -2B inactivate p53, the Tax-2A protein appears to inhibit p53 function less efficiently than either Tax-1 or Tax-2B. In transient-cotransfection assays, Tax-1 and Tax-2B inactivated p53 by 80%, while Tax2A reduced p53 activity by 20%. In addition, Tax-2A does not increase the steady-state level of cellular p53 as well as Tax-1 or -2B does in the same assays. Cotransfection assays demonstrated that Tax-2A could efficiently transactivate CREB-responsive promoters to the same level as Tax-1 and Tax-2B, indicating that the protein was functional. This report provides evidence of the first functional difference between the HTLV-2A and -2B subtypes. This comparison of the action of HTLV-1 and HTLV-2 Tax proteins on p53 function will provide important insights into the mechanism of HTLV transformation.


Asunto(s)
Transformación Celular Viral , Productos del Gen tax/metabolismo , Virus Linfotrópico T Tipo 1 Humano/fisiología , Virus Linfotrópico T Tipo 2 Humano/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Línea Celular Transformada , Rayos gamma , Regulación de la Expresión Génica , Productos del Gen tax/genética , Infecciones por HTLV-II/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 2 Humano/genética , Humanos , Células Jurkat , Fosforilación , Linfocitos T/virología , Transcripción Genética , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/efectos de la radiación
11.
J Virol ; 74(14): 6622-31, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10864676

RESUMEN

The production of the human papillomavirus type 16 (HPV-16) is intimately tied to the differentiation of the host epithelium that it infects. Infection occurs in the basal layer of the epithelium at a site of wounding, where the virus utilizes the host DNA replication machinery to establish itself as a low-copy-number episome. The productive stage of the HPV-16 life cycle occurs in the postmitotic suprabasal layers of the epithelium, where the virus amplifies its DNA to high copy number, synthesizes the capsid proteins (L1 and L2), encapsidates the HPV-16 genome, and releases virion particles as the upper layer of the epithelium is shed. Papillomaviruses are hypothesized to possess a mechanism to overcome the block in DNA synthesis that occurs in the differentiated epithelial cells, and the HPV-16 E7 oncoprotein has been suggested to play a role in this process. To determine whether E7 plays a role in the HPV-16 life cycle, an E7-deficient HPV-16 genome was created by inserting a translational termination linker (TTL) in the E7 gene of the full HPV-16 genome. This DNA was transfected into an immortalized human foreskin keratinocyte cell line shown previously to support the HPV-16 life cycle, and stable cell lines were obtained that harbored the E7-deficient HPV-16 genome episomally, the state of the genome found in normal infections. By culturing these cells under conditions which promote the differentiation of epithelial cells, we found E7 to be necessary for the productive stage of the HPV-16 life cycle. HPV-16 lacking E7 failed to amplify its DNA and expressed reduced amounts of the capsid protein L1, which is required for virus production. E7 appears to create a favorable environment for HPV-16 DNA synthesis by perturbing the keratinocyte differentiation program and inducing the host DNA replication machinery. These data demonstrate that E7 plays an essential role in the papillomavirus life cycle.


Asunto(s)
Proteínas de la Cápside , Proteínas Nucleares , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/crecimiento & desarrollo , Replicación Viral/fisiología , Apoptosis , Southern Blotting , Cápside/biosíntesis , Diferenciación Celular , Células Cultivadas , ADN Viral/biosíntesis , Células Epiteliales/virología , Humanos , Inmunohistoquímica , Hibridación in Situ , Queratinocitos/virología , Proteínas Oncogénicas Virales/biosíntesis , Proteínas Oncogénicas Virales/genética , Papillomaviridae/metabolismo , Papillomaviridae/fisiología , Proteínas E7 de Papillomavirus , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2 , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo
12.
Cancer Res ; 60(11): 2810-5, 2000 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-10850419

RESUMEN

The E7 oncoprotein of human papillomavirus 16 functions as a tumor-specific antigen in transformed epithelial cells of the uterine cervix to which immunotherapeutic strategies aimed at CTL induction may be directed. We previously have shown in mice transgenic for the E7 gene driven off an epithelial specific (keratin-14) promoter, that expression of E7 protein in peripheral epithelium is sufficient to tolerize E7-directed CTL precursors (pCTL; Doan et al, J. Virol., 73: 6166-1670, 1999). Here we show that E7 is presented to T cells for tolerization by cells of bone marrow origin ("cross-tolerization"). We demonstrate that tolerization of E7-directed pCTLs occurs within 2 weeks of exposure to E7 in epithelium. It is maintained in the near absence of CD4+ cells and in the absence of the thymus, and is independent of a coexisting E7-directed Th2-type antibody response. Tolerance was broken by immunization with E7 CTL epitope-pulsed dendritic cells. These findings have implications for immunotherapy of patients with human papillomavirus 16-associated cervical carcinoma, whose immune systems may have experienced long-term exposure to E7-expressing epithelial cells.


Asunto(s)
Células Dendríticas/inmunología , Tolerancia Inmunológica , Proteínas Oncogénicas Virales/inmunología , Proteínas Oncogénicas Virales/metabolismo , Células Th2/inmunología , Traslado Adoptivo , Animales , Antígenos de Neoplasias/metabolismo , Linfocitos T CD4-Positivos/inmunología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales , Epítopos , Femenino , Interferón gamma/inmunología , Masculino , Ratones , Proteínas E7 de Papillomavirus , Bazo/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Células Th2/metabolismo , Timo/metabolismo , Factores de Tiempo
13.
J Clin Microbiol ; 38(2): 688-95, 2000 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-10655368

RESUMEN

Many human papillomavirus (HPV) genotypes are associated with cervical carcinoma. We demonstrate the utility of an innovative technique for genotyping of HPV in cervical tissue samples. This method provides an accurate means of identification of the specific HPV genotypes present in clinical specimens. By using the MY09-MY11 and the GP5(+)-GP6(+) consensus primer pairs, HPV sequences were amplified by nested PCR from DNA isolated from cervical smear samples. This led to the production of an approximately 140-bp PCR product from the L1 (major capsid) gene of any of the HPVs present in the sample. PCR was performed with a deoxynucleoside triphosphate mixture which resulted in the incorporation of deoxyuridine into the amplified DNA product at positions where deoxythymidine would normally be incorporated at a frequency of about once or twice per strand. Following the PCR, the product was treated with an enzyme mix that contains uracil N-glycosylase (UNG) and endonuclease IV. UNG removes the uracil base from the nucleotide, and endonuclease IV cleaves the phosphodiester bond at this newly formed abasic site, producing fragments of various sizes. By having end labeled one of the amplification primers, a DNA ladder which is analogous to a "T-sequencing ladder" was produced upon electrophoresis of the products. By comparing this T-sequencing ladder to the known sequences of HPVs, the genotypes of unknown HPV isolates in samples were assigned. Data showing the utility of this technique for the rapid analysis of clinical samples are presented.


Asunto(s)
ADN Glicosilasas , Papillomaviridae/clasificación , Papillomaviridae/genética , Infecciones por Papillomavirus/virología , Reacción en Cadena de la Polimerasa/métodos , Infecciones Tumorales por Virus/virología , Secuencia de Bases , Cápside/genética , Liasas de Carbono-Oxígeno/metabolismo , Cuello del Útero/virología , Cartilla de ADN , ADN Viral/análisis , ADN Viral/aislamiento & purificación , ADN-(Sitio Apurínico o Apirimidínico) Liasa , Desoxirribonucleasa IV (Fago T4-Inducido) , Femenino , Genotipo , Humanos , Datos de Secuencia Molecular , N-Glicosil Hidrolasas/metabolismo , Radioisótopos de Fósforo/metabolismo , Juego de Reactivos para Diagnóstico , Análisis de Secuencia de ADN , Uracil-ADN Glicosidasa
14.
Virology ; 267(2): 141-50, 2000 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-10662610

RESUMEN

High-risk human papillomaviruses (HPVs) are etiologically implicated in human cervical cancer. Two viral genes, E6 and E7, are commonly found expressed in these cancer cells. We have previously shown that mice transgenic for the HPV-16 E6 gene or E7 gene, in which the E6 or E7 was expressed in the basal layer of epithelia, developed skin tumors. The spectrum of tumors derived from E6 and E7 mice differed, however; although most tumors derived from the E7-transgenic mice were benign, the majority of the tumors from the E6-transgenic mice were malignant. These findings led us to hypothesize that E6 and E7 play different roles in carcinogenesis. To assess at what stages in carcinogenesis E6 and E7 act, we treated the skin of K14E6- and K14E7-transgenic mice with chemical carcinogens known to contribute to distinct stages in carcinogenesis. Both E6 and E7 were found to synergize with chemical carcinogens in causing tumor formation. E6 was found to act weakly at the promotion stage of carcinogenesis in the formation of benign tumors but strongly at the progression stage which involves the malignant conversion of benign tumors. In contrast, E7 primarily affected the promotion stage of carcinogenesis. These results provide direct evidence that E6 and E7 contribute differently to carcinogenesis; E7 promotes the formation of benign tumors, and E6 acts primarily to accelerate progression of these benign tumors to the malignant stage. Consistent with this model, we found E6 and E7 to cooperate in inducing tumor formation in mice expressing both oncogenes.


Asunto(s)
Carcinógenos/efectos adversos , Proteínas Oncogénicas Virales/fisiología , Proteínas Represoras , 9,10-Dimetil-1,2-benzantraceno/efectos adversos , Animales , Cocarcinogénesis , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes ras/genética , Humanos , Ratones , Ratones Transgénicos , Mutación , Proteínas Oncogénicas Virales/genética , Papiloma/etiología , Papiloma/genética , Papiloma/patología , Proteínas E7 de Papillomavirus , Índice de Severidad de la Enfermedad , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología , Acetato de Tetradecanoilforbol/efectos adversos
15.
Am J Pathol ; 155(4): 1121-7, 1999 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-10514395

RESUMEN

Different parts of the skin respond to ionizing radiation with different sensitivities. To examine the mechanisms underlying these different responses, we investigated various cellular parameters in the skin after exposure of mice to 5 Gy of ionizing radiation. Epidermal cells responded to radiation by undergoing growth arrest, whereas the cells in the matrix of hair follicles underwent apoptosis but not growth arrest. These distinct responses correlated with differential increases in p53 and p21 proteins in these two populations of cells; whereas an increase in p53 protein levels was observed in both epidermis and hair follicular matrix, especially in the latter, the induction of p21 was strong in the epidermis but absent in the follicular matrical cells. Studies using p53-null and p21-null mice demonstrated that the radiation-induced apoptosis in the hair follicles was fully dependent on p53, and growth arrest in the epidermis was only partially dependent on p53 but fully dependent on p21. These results indicate that two epithelial cell types respond to radiation by different pathways that are governed in part by the differential p53- and p21-dependent responses of these cells; high-level induction of p53 in the absence of p21 induction led to apoptosis, whereas intermediate induction of both p53 and p21 led to growth arrest.


Asunto(s)
Ciclinas/biosíntesis , Epidermis/efectos de la radiación , Folículo Piloso/efectos de la radiación , Proteína p53 Supresora de Tumor/biosíntesis , Animales , Apoptosis , Diferenciación Celular , División Celular/efectos de la radiación , Inhibidor p21 de las Quinasas Dependientes de la Ciclina , Ciclinas/genética , Células Epidérmicas , Epidermis/metabolismo , Rayos gamma , Regulación de la Expresión Génica/efectos de la radiación , Folículo Piloso/citología , Folículo Piloso/metabolismo , Inmunohistoquímica , Etiquetado Corte-Fin in Situ , Ratones , Ratones Endogámicos , Ratones Noqueados , Factores de Tiempo , Proteína p53 Supresora de Tumor/genética
16.
Virology ; 262(2): 344-54, 1999 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-10502513

RESUMEN

The study of human papillomaviruses (HPVs) in cell culture has been hindered because of the difficulty in recreating the three-dimensional structure of the epithelium on which the virus depends to complete its life cycle. Additionally, the study of genetic mutations in the HPV genome and its effects on the viral life cycle are difficult using the current method of transfecting molecularly cloned HPV genomes into early-passage human foreskin keratinocytes (HFKs) because of the limited life span of these cells. Unless the HPV genome transfected into the early-passage HFK extends the life span of the cell, analysis of stable transfectants becomes difficult. In this study, we have used BC-1-Ep/SL cells, an immortalized human foreskin keratinocyte cell line, to recreate the HPV-16 life cycle. This cell line exhibits many characteristics of the early-passage HFKs including the ability to stratify and terminally differentiate in an organotypic raft culture system. Because of their similarity to early-passage HFKs, these cells were tested for their ability to support the HPV-16 life cycle. The BC-1-Ep/SL cells could stably maintain two HPV genotypes, HPV-16 and HPV-31b, episomally. Additionally, when the BC-1-Ep/SL cell line was stably transfected with HPV-16 and cultured using the organotypic raft culture system (rafts), it sustained the HPV-16 life cycle. Evidence for the productive stage of the HPV-16 life cycle was provided by: DNA in situ hybridization demonstrating HPV-16 DNA amplification in the suprabasal layers of the rafts, immunohistochemical staining for L1 showing the presence of capsid protein in the suprabasal layers of the rafts, and electron microscopy indicating the presence of virus like particles (VLPs) in nuclei from cells in the differentiated layers of the rafts.


Asunto(s)
Proteínas de la Cápside , Queratinocitos/virología , Papillomaviridae/crecimiento & desarrollo , Plásmidos/genética , Técnicas de Cultivo de Célula , Diferenciación Celular , Línea Celular , Núcleo Celular/virología , Clonación Molecular , ADN Viral/análisis , ADN Viral/genética , Proteínas Filagrina , Genoma Viral , Humanos , Inmunohistoquímica , Hibridación in Situ , Proteínas de Filamentos Intermediarios/análisis , Queratina-10 , Queratinocitos/citología , Queratinocitos/metabolismo , Queratinocitos/ultraestructura , Queratinas/análisis , Microscopía Electrónica , Datos de Secuencia Molecular , Proteínas Oncogénicas Virales/análisis , Papillomaviridae/genética , Papillomaviridae/aislamiento & purificación , Papillomaviridae/ultraestructura , Pase Seriado , Transfección
17.
Intervirology ; 42(1): 43-50, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10393503

RESUMEN

One of three lines of mice transgenic for the E6 and E7 genes of human papillomavirus type 16 (HPV16) expressed from an alphaA-crystallin promoter also expresses the transgene ectopically in the skin. This line, designated alphaACE6E7#19, develops skin disease from 3 months of age, characterised by epidermal hyperplasia and eventual skin loss. Administration of complete Freund's adjuvant (CFA) to alphaACE6E7#19 mice, but not to non-transgenic littermate controls, induced local epidermal hyperplasia which was histologically similar to the spontaneously arising skin pathology. Local application of 2,4-dinitrochlorobenzene (DNCB) to DNCB-sensitised alphaACE6E7#19 mice, but not DNCB-sensitised controls, also induced hyperplasia. Treatment with cyclosporin A (CsA) or systemic depletion of CD4+ cells significantly reduced the incidence of skin disease. These data suggest that local inflammation, and cytokines produced by T helper cells, contribute to the induction of hyperplastic skin disease in alphaACE6E7#19 mice. Spontaneous skin disease with similar histological appearance, frequency, age of onset and severity in alphaACE6E7#19 mice was observed in scid-/- alphaACE6E7#19 mice, despite immune paresis. Antigen-specific immune responses and T-cell cytokines are therefore not necessary for the induction of skin disease. We propose that epidermal hyperplasia associated with HPV16 E6 and E7 expression in skin is accelerated by local secretion of pro-inflammatory cytokines, whose production can be enhanced by activated CD4+ T cells.


Asunto(s)
Ratones Transgénicos/genética , Proteínas Represoras , Enfermedades de la Piel/inmunología , Piel/patología , Linfocitos T/inmunología , Factores de Edad , Animales , Ciclosporina/uso terapéutico , Dinitroclorobenceno , Femenino , Humanos , Hiperplasia/inducido químicamente , Hiperplasia/inmunología , Hiperplasia/patología , Inmunidad Celular , Inmunosupresores/uso terapéutico , Irritantes , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Ratones SCID , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Proteínas E7 de Papillomavirus , Piel/efectos de los fármacos , Piel/inmunología , Piel/virología , Enfermedades de la Piel/inducido químicamente , Enfermedades de la Piel/tratamiento farmacológico , Organismos Libres de Patógenos Específicos
18.
J Virol ; 73(7): 5887-93, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10364340

RESUMEN

High-risk human papillomaviruses (HPVs) are the causative agents of certain human cancers. HPV type 16 (HPV16) is the papillomavirus most frequently associated with cervical cancer in women. The E6 and E7 genes of HPV are expressed in cells derived from these cancers and can transform cells in tissue culture. Animal experiments have demonstrated that E6 and E7 together cause tumors. We showed previously that E6 and E7 together or E7 alone could induce skin tumors in mice when these genes were expressed in the basal epithelia of the skin. In this study, we investigated the role that the E6 gene plays in carcinogenesis. We generated K14E6 transgenic mice, in which the HPV16 E6 gene was directed in its expression by the human keratin 14 promoter (hK14) to the basal layer of the epidermis. We found that E6 induced cellular hyperproliferation and epidermal hyperplasia and caused skin tumors in adult mice. Interestingly, the tumors derived from E6 were mostly malignant, as opposed to the tumors from E7 mice, which were mostly benign. This result leads us to hypothesize that E6 may contribute differently than E7 to HPV-associated carcinogenesis; whereas E7 primarily contributes to the early stages of carcinogenesis that lead to the formation of benign tumors, E6 primarily contributes to the late stages of carcinogenesis that lead to malignancy.


Asunto(s)
Transformación Celular Neoplásica , Transformación Celular Viral , Proteínas Oncogénicas Virales/fisiología , Papillomaviridae/fisiología , Proteínas Represoras , Neoplasias Cutáneas/virología , Animales , Daño del ADN , Genes ras , Humanos , Hiperplasia/patología , Hiperplasia/virología , Ratones , Ratones Transgénicos , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Fenotipo , Neoplasias Cutáneas/patología
19.
Invest Ophthalmol Vis Sci ; 39(13): 2723-32, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9856783

RESUMEN

PURPOSE: Forty percent of cases of retinoblastoma, a childhood malignancy of the retina, are linked to the inheritance of a mutant allele of the retinoblastoma susceptibility gene Rb1. Tumor penetrance varies among carriers in different family pedigrees, indicating that other genetic factors may modify risk for occurrence of retinoblastoma. This study was undertaken to determine whether multiple genetic loci modify the risk for retinoblastoma in mice. METHODS: A line of alphaAcry-HPV16E6/E7 transgenic mice expressing the human papillomavirus type 16 E6 and E7 oncogenes (HPV-16 E6 and E7) ectopically in the retina was characterized. E6 and E7 proteins bind to and inactivate the cellular tumor suppressor proteins p53 and Rb, respectively. RESULTS: Retinoblastomas developed rarely when the alphaAcry-HPV16E6/E7 transgene was maintained on the FVB background, but tumors arose with high frequency on C57BL/6 X FVB and C3H x FVB F1 hybrid backgrounds. The incidence of retinoblastoma in the LHbeta-TAG transgenic mice, which express simian virus 40 large tumor antigen (SV40 T-ag), was also influenced by the FVB and C57BL/6 backgrounds. Resistance of the alphaAcry-HPV16E6/E7 FVB mice to retinoblastoma mapped in part to the retinal degeneration (rd) locus. However, multiple genetic experiments indicate that resistance to retinoblastoma depends on additional loci in FVB mice. CONCLUSIONS: Multiple cellular genes can modify risk for retinoblastoma in mice.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Oncogénicas Virales/genética , Papillomaviridae/genética , Retina/patología , Neoplasias de la Retina/genética , Retinoblastoma/genética , Animales , Antígenos Transformadores de Poliomavirus/genética , Cartilla de ADN/química , Femenino , Genotipo , Hibridación in Situ , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas E7 de Papillomavirus , Retina/virología , Neoplasias de la Retina/patología , Retinoblastoma/patología , Proteína de Retinoblastoma/genética , Factores de Riesgo , Proteína p53 Supresora de Tumor/genética
20.
J Biol Chem ; 273(49): 33048-53, 1998 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-9830059

RESUMEN

p53 exerts its cell cycle regulatory effects through its ability to function as a sequence-specific DNA binding transcription factor. CREB-binding protein (CBP)/p300, through its interaction with the N terminus of p53, acts as a coactivator for p53 and increases the sequence-specific DNA-binding activity of p53 by acetylating its C terminus. The same N-terminal domain of p53 has recently been shown to be phosphorylated at Ser15 in response to gamma-irradiation. Remarkably, we now demonstrate that phosphorylation of p53 at Ser15 increases its ability to recruit CBP/p300. The increase in CBP/p300 binding was followed by an increase in the overall level of acetylation of the C terminus of p53. These results provide a mechanism for the activation of p53-regulated genes following DNA damage, through a signaling pathway linking p53 N-terminal kinase and C-terminal acetyltransferase activities.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Nucleares/metabolismo , Serina/metabolismo , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Acetilación , Proteína de Unión a CREB , Proteína Quinasa Activada por ADN , Células HeLa , Humanos , Mutagénesis Sitio-Dirigida , Fosforilación , Unión Proteica , Proteínas Serina-Treonina Quinasas/metabolismo , Radiación Ionizante , Proteína p53 Supresora de Tumor/química , Proteína p53 Supresora de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA