Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Más filtros

Intervalo de año de publicación
1.
J Virol ; 96(4): e0186521, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34878887

RESUMEN

Etiologically, 5% of all cancers worldwide are caused by the high-risk human papillomaviruses (hrHPVs). These viruses encode two oncoproteins (E6 and E7) whose expression is required for cancer initiation and maintenance. Among their cellular targets are the p53 and the retinoblastoma tumor suppressor proteins. Inhibition of the hrHPV E6-mediated ubiquitylation of p53 through the E6AP ubiquitin ligase results in the stabilization of p53, leading to cellular apoptosis. We utilized a live cell high-throughput screen to determine whether exogenous microRNA (miRNA) transfection had the ability to stabilize p53 in hrHPV-positive cervical cancer cells expressing a p53-fluorescent protein as an in vivo reporter of p53 stability. Among the miRNAs whose transfection resulted in the greatest p53 stabilization was 375-3p, which has previously been reported to stabilize p53 in HeLa cells, providing validation of the screen. The top 32 miRNAs, in addition to 375-3p, were further assessed using a second cell-based p53 stability reporter system, as well as in nonreporter HeLa cells to examine their effects on endogenous p53 protein levels, resulting in the identification of 23 miRNAs whose transfection increased p53 levels in HeLa cells. While a few miRNAs that stabilized p53 led to decreases in E6AP protein levels, all targeted HPV oncoprotein expression. We further examined subsets of these miRNAs for their abilities to induce apoptosis and determined whether it was p53-mediated. The introduction of specific miRNAs revealed surprisingly heterogeneous responses in different cell lines. Nonetheless, some of the miRNAs described here have potential as therapeutics for treating HPV-positive cancers. IMPORTANCE Human papillomaviruses cause approximately 5% of all cancers worldwide and encode genes that contribute to both the initiation and maintenance of these cancers. The viral oncoprotein E6 is expressed in all HPV-positive cancers and functions by targeting the degradation of p53 through the engagement of the cellular ubiquitin ligase E6AP. Inhibiting the degradation of p53 leads to apoptosis in HPV-positive cancer cells. Using a high-throughput live cell assay, we identified several miRNAs whose transfection stabilize p53 in HPV-positive cells. These miRNAs have the potential to be used in the treatment of HPV-positive cancers.


Asunto(s)
Alphapapillomavirus/metabolismo , MicroARNs/genética , Proteína p53 Supresora de Tumor/metabolismo , Alphapapillomavirus/genética , Apoptosis , Línea Celular Tumoral , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Células HeLa , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas Oncogénicas Virales/genética , Proteínas Oncogénicas Virales/metabolismo , Estabilidad Proteica , Ubiquitina-Proteína Ligasas/metabolismo
2.
J Biol Chem ; 293(47): 18387-18399, 2018 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-30257870

RESUMEN

Deregulation of the HECT ubiquitin ligase UBE3A/E6AP has been implicated in Angelman syndrome as well as autism spectrum disorders. We and others have previously identified the 26S proteasome as one of the major UBE3A-interacting protein complexes. Here, we characterize the interaction of UBE3A and the proteasomal subunit PSMD4 (Rpn10/S5a). We map the interaction to the highly conserved Zn2+-binding N-terminal (AZUL) domain of UBE3A, the integrity of which is crucial for binding to PSMD4. Interestingly, two Angelman syndrome point mutations that affect the AZUL domain show an impaired ability to bind PSMD4. Although not affecting the ubiquitin ligase or the estrogen receptor α-mediated transcriptional regulation activities, these AZUL domain mutations prevent UBE3A from stimulating the Wnt/ß-catenin signaling pathway. Taken together, our data indicate that impaired binding to the 26S proteasome and consequential deregulation of Wnt/ß-catenin signaling might contribute to the functional defect of these mutants in Angelman syndrome.


Asunto(s)
Síndrome de Angelman/enzimología , Mutación Puntual , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/genética , Zinc/metabolismo , Síndrome de Angelman/genética , Humanos , Complejo de la Endopetidasa Proteasomal/genética , Proteínas de Unión al ARN , Ubiquitina-Proteína Ligasas/metabolismo , Vía de Señalización Wnt
3.
J Virol ; 92(15)2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29848583

RESUMEN

The papillomavirus E2 protein executes numerous essential functions related to viral transcription, replication of viral DNA, and viral genome maintenance. Because E2 lacks enzymatic activity, many of these functions are mediated by interactions with host cellular proteins. Unbiased proteomics approaches have successfully identified a number of E2-host protein interactions. We have extended such studies and have identified and validated the cellular proteins structural maintenance of chromosome 5 (SMC5) and SMC6 as interactors of the viral E2 protein. These two proteins make up the core components of the SMC5/6 complex. The SMC5/6 complex is a member of the conserved structural maintenance of chromosomes (SMC) family of proteins, which are essential for genome maintenance. We have examined the role of SMC5/6 in various E2 functions. Our data suggest that SMC6 is not required for E2-mediated transcriptional activation, E1/E2-mediated transient replication, or differentiation-dependent amplification of viral DNA. Our data, however, suggest a role for SMC5/6 in viral genome maintenance.IMPORTANCE The high-risk human papillomaviruses (HPVs) are the etiological cause of cervical cancer and the most common sexually transmitted infection. While the majority of infections may be asymptomatic or cause only benign lesions, persistent infection with the oncogenic high-risk HPV types may lead to serious diseases, such as cervical cancer, anogenital carcinoma, or head and neck oropharyngeal squamous cell carcinoma. The identification of virus-host protein interactions provides insights into the mechanisms of viral DNA persistence, viral genome replication, and cellular transformation. Elucidating the mechanism of early events in the virus replication cycle as well as of integration of viral DNA into host chromatin may present novel antiviral strategies and targets for counteracting persistent infection. The E2 protein is an important viral regulatory protein whose functions are mediated through interactions with host cell proteins. Here we explore the interaction of E2 with SMC5/6 and the functional consequences.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/fisiología , Línea Celular Tumoral , Replicación del ADN , Células HEK293 , Humanos , Papillomaviridae/genética , Proteómica , Activación Transcripcional , Replicación Viral
4.
J Virol ; 92(6)2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29263274

RESUMEN

Retinoic acid-inducible gene I (RIG-I) is a key pattern recognition receptor that senses viral RNA and interacts with the mitochondrial adaptor MAVS, triggering a signaling cascade that results in the production of type I interferons (IFNs). This signaling axis is initiated by K63-linked ubiquitination of RIG-I mediated by the E3 ubiquitin ligase TRIM25, which promotes the interaction of RIG-I with MAVS. USP15 was recently identified as an upstream regulator of TRIM25, stabilizing the enzyme through removal of degradative K48-linked polyubiquitin, ultimately promoting RIG-I-dependent cytokine responses. Here, we show that the E6 oncoprotein of human papillomavirus type 16 (HPV16) as well as of other HPV types form a complex with TRIM25 and USP15 in human cells. In the presence of E6, the K48-linked ubiquitination of TRIM25 was markedly increased, and in line with this, TRIM25 degradation was enhanced. Our results further showed that E6 inhibited the TRIM25-mediated K63-linked ubiquitination of RIG-I and its CARD-dependent interaction with MAVS. HPV16 E6, but not E7, suppressed the RIG-I-mediated induction of IFN-ß, chemokines, and IFN-stimulated genes (ISGs). Finally, CRISPR-Cas9 gene targeting in human keratinocytes showed that the TRIM25-RIG-I-MAVS triad is important for eliciting an antiviral immune response to HPV16 infection. Our study thus identifies a novel immune escape mechanism that is conserved among different HPV strains and further indicates that the RIG-I signaling pathway plays an important role in the innate immune response to HPV infection.IMPORTANCE Persistent infection and tumorigenesis by HPVs are known to require viral manipulation of a variety of cellular processes, including those involved in innate immune responses. Here, we show that the HPV E6 oncoprotein antagonizes the activation of the cytoplasmic innate immune sensor RIG-I by targeting its upstream regulatory enzymes TRIM25 and USP15. We further show that the RIG-I signaling cascade is important for an antiviral innate immune response to HPV16 infection, providing evidence that RIG-I, whose role in sensing RNA virus infections has been well characterized, also plays a crucial role in the antiviral host response to small DNA viruses of the Papillomaviridae family.


Asunto(s)
Proteína 58 DEAD Box/inmunología , Papillomavirus Humano 6/inmunología , Inmunidad Innata , Queratinocitos/inmunología , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/inmunología , Transducción de Señal/inmunología , Factores de Transcripción/inmunología , Proteínas de Motivos Tripartitos/inmunología , Ubiquitina-Proteína Ligasas/inmunología , Proteasas Ubiquitina-Específicas/inmunología , Proteína 58 DEAD Box/genética , Células HEK293 , Papillomavirus Humano 6/genética , Humanos , Queratinocitos/patología , Queratinocitos/virología , Proteínas Oncogénicas Virales/genética , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Receptores Inmunológicos , Transducción de Señal/genética , Factores de Transcripción/genética , Proteínas de Motivos Tripartitos/genética , Ubiquitina-Proteína Ligasas/genética , Proteasas Ubiquitina-Específicas/genética
5.
PLoS Pathog ; 13(10): e1006668, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29028833

RESUMEN

Merkel cell carcinoma (MCC) frequently contains integrated copies of Merkel cell polyomavirus DNA that express a truncated form of Large T antigen (LT) and an intact Small T antigen (ST). While LT binds RB and inactivates its tumor suppressor function, it is less clear how ST contributes to MCC tumorigenesis. Here we show that ST binds specifically to the MYC homolog MYCL (L-MYC) and recruits it to the 15-component EP400 histone acetyltransferase and chromatin remodeling complex. We performed a large-scale immunoprecipitation for ST and identified co-precipitating proteins by mass spectrometry. In addition to protein phosphatase 2A (PP2A) subunits, we identified MYCL and its heterodimeric partner MAX plus the EP400 complex. Immunoprecipitation for MAX and EP400 complex components confirmed their association with ST. We determined that the ST-MYCL-EP400 complex binds together to specific gene promoters and activates their expression by integrating chromatin immunoprecipitation with sequencing (ChIP-seq) and RNA-seq. MYCL and EP400 were required for maintenance of cell viability and cooperated with ST to promote gene expression in MCC cell lines. A genome-wide CRISPR-Cas9 screen confirmed the requirement for MYCL and EP400 in MCPyV-positive MCC cell lines. We demonstrate that ST can activate gene expression in a EP400 and MYCL dependent manner and this activity contributes to cellular transformation and generation of induced pluripotent stem cells.


Asunto(s)
Antígenos Virales de Tumores/metabolismo , Carcinoma de Células de Merkel/virología , Transformación Celular Viral/fisiología , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Antígenos Transformadores de Poliomavirus/metabolismo , Carcinoma de Células de Merkel/genética , Carcinoma de Células de Merkel/metabolismo , Línea Celular Tumoral , Humanos , Immunoblotting , Inmunoprecipitación , Poliomavirus de Células de Merkel , Infecciones por Polyomavirus/complicaciones , Infecciones por Polyomavirus/genética , Infecciones por Polyomavirus/metabolismo , Infecciones Tumorales por Virus/complicaciones , Infecciones Tumorales por Virus/genética , Infecciones Tumorales por Virus/metabolismo
7.
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
8.
Mol Cell ; 32(5): 718-26, 2008 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-19061646

RESUMEN

The neuronal gene repressor REST/NRSF recruits corepressors, including CoREST, to modify histones and repress transcription. REST also functions as a tumor suppressor, but the mechanism remains unclear. We identified chromodomain on Y-like (CDYL) as a REST corepressor that physically bridges REST and the histone methylase G9a to repress transcription. Importantly, RNAi knockdown of REST, CDYL, and G9a, but not CoREST, induced oncogenic transformation of immortalized primary human cells and derepression of the proto-oncogene TrkC. Significantly, transgenic expression of TrkC also induced transformation. This implicates CDYL-G9a, but not CoREST, in REST suppression of transformation, possibly by oncogene repression. CDYL knockdown also augments transformation in a cell culture model of cervical cancer, where loss of heterozygosity of the CDYL locus occurs. These findings demonstrate molecular strategies by which REST carries out distinct biological functions via different corepressors and provide critical insights into the role of histone-modifying complexes in regulating cellular transformation.


Asunto(s)
Transformación Celular Neoplásica/genética , Regulación hacia Abajo/genética , Proteína Metiltransferasas/metabolismo , Proteínas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Co-Represoras , Proteínas de Unión al ADN/metabolismo , Células Epiteliales/metabolismo , Células HeLa , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina , Histonas/metabolismo , Humanos , Hidroliasas , Lisina/metabolismo , Metilación , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Proteínas E7 de Papillomavirus , Unión Proteica , Proto-Oncogenes Mas , Interferencia de ARN , Receptor trkC/metabolismo , Transcripción Genética
9.
J Proteome Res ; 14(2): 953-66, 2015 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-25476789

RESUMEN

HERC2 is a large E3 ubiquitin ligase with multiple structural domains that has been implicated in an array of cellular processes. Mutations in HERC2 are linked to developmental delays and impairment caused by nervous system dysfunction, such as Angelman Syndrome and autism-spectrum disorders. However, HERC2 cellular activity and regulation remain poorly understood. We used a broad proteomic approach to survey the landscape of cellular proteins that interact with HERC2. We identified nearly 300 potential interactors, a subset of which we validated binding to HERC2. The potential HERC2 interactors included the eukaryotic translation initiation factor 3 complex, the intracellular transport COPI coatomer complex, the glycogen regulator phosphorylase kinase, beta-catenin, PI3 kinase, and proteins involved in fatty acid transport and iron homeostasis. Through a complex bioinformatic analysis of potential interactors, we linked HERC2 to cellular processes including intracellular protein trafficking and transport, metabolism of cellular energy, and protein translation. Given its size, multidomain structure, and association with various cellular activities, HERC2 may function as a scaffold to integrate protein complexes and bridge critical cellular pathways. This work provides a significant resource with which to interrogate HERC2 function more deeply and evaluate its contributions to mechanisms governing cellular homeostasis and disease.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/metabolismo , Mapeo de Interacción de Proteínas/métodos , Proteoma/análisis , Proteoma/metabolismo , Factores de Intercambio de Guanina Nucleótido/análisis , Humanos , Proteínas/análisis , Proteínas/metabolismo , Proteínas/fisiología , Proteómica , Ubiquitina-Proteína Ligasas
10.
J Virol ; 88(15): 8201-12, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24850740

RESUMEN

UNLABELLED: The genus beta human papillomaviruses (beta HPVs) cause cutaneous lesions and are thought to be involved in the initiation of some nonmelanoma skin cancers (NMSCs), particularly in patients with the genetic disorder epidermodysplasia verruciformis (EV). We have previously reported that at least two of the genus beta HPV E6 proteins bind to and/or increase the steady-state levels of p53 in squamous epithelial cells. This is in contrast to a well-characterized ability of the E6 proteins of cancer-associated HPVs of genus alpha HPV, which inactivate p53 by targeting its ubiquitin-mediated proteolysis. In this study, we have investigated the ability of genus beta E6 proteins from eight different HPV types to block the transactivation of p53 target genes following DNA damage. We find that the E6 proteins from diverse beta HPV species and types vary in their capacity to block the induction of MDM2, p21, and proapoptotic genes after genotoxic stress. We conclude that some genus beta HPV E6 proteins inhibit at least some p53 target genes, although perhaps not by the same mechanism or to the same degree as the high-risk genus alpha HPV E6 proteins. IMPORTANCE: This study addresses the ability of various human papillomavirus E6 proteins to block the activation of p53-responsive cellular genes following DNA damage in human keratinocytes, the normal host cell for HPVs. The E6 proteins encoded by the high-risk, cancer-associated HPV types of genus alpha HPV have a well-established activity to target p53 degradation and thereby inhibit the response to DNA damage. In this study, we have investigated the ability of genus beta HPV E6 proteins from eight different HPV types to block the ability of p53 to transactivate downstream genes following DNA damage. We find that some, but not all, genus beta HPV E6 proteins can block the transactivation of some p53 target genes. This differential response to DNA damage furthers the understanding of cutaneous HPV biology and may help to explain the potential connection between some beta HPVs and cancer.


Asunto(s)
Betapapillomavirus/fisiología , Interacciones Huésped-Patógeno , Proteínas Oncogénicas Virales/metabolismo , Activación Transcripcional , Apoptosis , Línea Celular , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/antagonistas & inhibidores , Daño del ADN , Humanos , Proteínas Proto-Oncogénicas c-mdm2/antagonistas & inhibidores
11.
Trans Am Clin Climatol Assoc ; 126: 117-32, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26330666
12.
Proc Natl Acad Sci U S A ; 109(5): E260-7, 2012 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-22232672

RESUMEN

More than 120 human papillomaviruses (HPVs) have now been identified and have been associated with a variety of clinical lesions. To understand the molecular differences among these viruses that result in lesions with distinct pathologies, we have begun a MS-based proteomic analysis of HPV-host cellular protein interactions and have created the plasmid and cell line libraries required for these studies. To validate our system, we have characterized the host cellular proteins that bind to the E7 proteins expressed from 17 different HPV types. These studies reveal a number of interactions, some of which are conserved across HPV types and others that are unique to a single HPV species or HPV genus. Binding of E7 to UBR4/p600 is conserved across all virus types, whereas the cellular protein ENC1 binds specifically to the E7s from HPV18 and HPV45, both members of genus alpha, species 7. We identify a specific interaction of HPV16 E7 with ZER1, a substrate specificity factor for a cullin 2 (CUL2)-RING ubiquitin ligase, and show that ZER1 is required for the binding of HPV16 E7 to CUL2. We further show that ZER1 is required for the destabilization of the retinoblastoma tumor suppressor RB1 in HPV16 E7-expressing cells and propose that a CUL2-ZER1 complex functions to target RB1 for degradation in HPV16 E7-expressing cells. These studies refine the current understanding of HPV E7 functions and establish a platform for the rapid identification of virus-host interactions.


Asunto(s)
Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/metabolismo , Secuencia de Aminoácidos , Western Blotting , Línea Celular , Humanos , Datos de Secuencia Molecular , Proteínas Oncogénicas Virales/química , Papillomaviridae/clasificación , Unión Proteica , Homología de Secuencia de Aminoácido , Especificidad de la Especie , Espectrometría de Masas en Tándem
13.
Proc Natl Acad Sci U S A ; 109(23): E1473-80, 2012 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-22547818

RESUMEN

The Notch signaling pathway is a key determinant in keratinocyte differentiation and growth cycle arrest, and has been reported to have a tumor suppressor function in skin. The papillomavirus life cycle is intricately linked to the differentiation status of keratinocytes. Papillomaviruses are associated with benign proliferative epithelial lesions in their respective hosts. Although human papillomaviruses (HPVs) associated with genital tract lesions have been extensively studied, studies of the cutaneous HPVs are more limited. In particular, it is well established that the E6 proteins of high-risk HPVs of the α-genus such as HPV16 and HPV18 mediate the degradation of p53 by its association with the ubiquitin ligase E6AP. In contrast, less is known about the cellular activities of the cutaneous HPVs of the ß-genus. By using an unbiased proteomic approach, we identify MAML1 and other members of the Notch transcription complex as high-confidence cellular interacting proteins of E6 proteins of the ß-genus HPVs and of the bovine papillomavirus type 1 associated with cutaneous fibropapillomas. We show that bovine papillomavirus type 1 and ß-HPV E6 repress Notch transcriptional activation, and that this repression is dependent on an interaction with MAML1. Finally, we show that the expression levels of endogenous Notch target genes are repressed by ß-HPV E6 proteins. These findings elucidate a mechanism of viral antagonism of Notch signaling, and suggest that Notch signaling is an important epithelial cell pathway target for the ß-HPVs.


Asunto(s)
Betapapillomavirus/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Oncogénicas Virales/metabolismo , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Animales , Western Blotting , Línea Celular , Humanos , Luciferasas , Ratones , Plásmidos/genética , Proteómica , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética , Transfección
14.
J Virol ; 86(24): 13174-86, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23015706

RESUMEN

We have begun to define the human papillomavirus (HPV)-associated proteome for a subset of the more than 120 HPV types that have been identified to date. Our approach uses a mass spectrometry-based platform for the systematic identification of interactions between human papillomavirus and host cellular proteins, and here we report a proteomic analysis of the E6 proteins from 16 different HPV types. The viruses included represent high-risk, low-risk, and non-cancer-associated types from genus alpha as well as viruses from four different species in genus beta. The E6 interaction data set consists of 153 cellular proteins, including several previously reported HPV E6 interactors such as p53, E6AP, MAML1, and p300/CBP and proteins containing PDZ domains. We report the genus-specific binding of E6s to either E6AP or MAML1, define the specific HPV E6s that bind to p300, and demonstrate several new features of interactions involving beta HPV E6s. In particular, we report that several beta HPV E6s bind to proteins containing PDZ domains and that at least two beta HPV E6s bind to p53. Finally, we report the newly discovered interaction of proteins of E6 of beta genus, species 2, with the Ccr4-Not complex, the first report of a viral protein binding to this complex. This data set represents a comprehensive survey of E6 binding partners that provides a resource for the HPV field and will allow continued studies on the diverse biology of the human papillomaviruses.


Asunto(s)
Proteínas Oncogénicas Virales/metabolismo , Papillomaviridae/metabolismo , Secuencia de Aminoácidos , Línea Celular , Humanos , Espectrometría de Masas , Datos de Secuencia Molecular , Proteínas Oncogénicas Virales/química , Sistemas de Lectura Abierta , Papillomaviridae/clasificación , Unión Proteica , Homología de Secuencia de Aminoácido
15.
Proc Natl Acad Sci U S A ; 107(8): 3752-7, 2010 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-20133580

RESUMEN

An essential step in the pathogenesis of human papillomavirus (HPV)-associated cancers is the dysregulated expression of the viral oncogenes. The papillomavirus E2 protein can silence the long control region (LCR) promoter that controls viral E6 and E7 oncogene expression. The mechanisms by which E2 represses oncogene expression and the cellular factors through which E2 mediates this silencing are largely unknown. We conducted an unbiased, genome-wide siRNA screen and series of secondary screens that identified 96 cellular genes that contribute to the repression of the HPV LCR. In addition to confirming a role for the E2-binding bromodomain protein Brd4 in E2-mediated silencing, we identified a number of genes that have not previously been implicated in E2 repression, including the demethylase JARID1C/SMCX as well as EP400, a component of the NuA4/TIP60 histone acetyltransferase complex. Each of these genes contributes independently and additively to E2-mediated silencing, indicating that E2 functions through several distinct cellular complexes to repress E6 and E7 expression.


Asunto(s)
ADN Helicasas/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Regulación Viral de la Expresión Génica , Papillomavirus Humano 18/genética , Proteínas Nucleares/metabolismo , Proteínas Oncogénicas Virales/genética , Oxidorreductasas N-Desmetilantes/metabolismo , Factores de Transcripción/metabolismo , Proteínas de Ciclo Celular , Línea Celular Tumoral , ADN Helicasas/genética , Femenino , Silenciador del Gen , Estudio de Asociación del Genoma Completo , Células HeLa , Histona Demetilasas , Humanos , Proteínas Nucleares/genética , Proteínas Oncogénicas Virales/metabolismo , Oxidorreductasas N-Desmetilantes/genética , Infecciones por Papillomavirus/metabolismo , Infecciones por Papillomavirus/virología , ARN Interferente Pequeño/genética , Factores de Transcripción/genética , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/virología , Proteínas Virales/metabolismo , Replicación Viral/genética
16.
J Virol ; 84(9): 4451-60, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20181716

RESUMEN

The papillomavirus E2 open reading frame encodes the full-length E2 protein as well as an alternatively spliced product called E8;E2C. E8;E2C has been best studied for the high-risk human papillomaviruses, where it has been shown to regulate viral genome levels and, like the full-length E2 protein, to repress transcription from the viral promoter that directs the expression of the viral E6 and E7 oncogenes. The repression function of E8;E2C is dependent on the 12-amino-acid N-terminal sequence from the E8 open reading frame (ORF). In order to understand the mechanism by which E8;E2C mediates transcriptional repression, we performed an unbiased proteomic analysis from which we identified six high-confidence candidate interacting proteins (HCIPs) for E8;E2C; the top two are NCoR1 and TBLR1. We established an interaction of E8;E2C with an NCoR1/HDAC3 complex and demonstrated that this interaction requires the wild-type E8 open reading frame. Small interfering RNA (siRNA) knockdown studies demonstrated the involvement of NCoR1/HDAC3 in the E8;E2C-dependent repression of the viral long control region (LCR) promoter. Additional genetic work confirmed that the papillomavirus E2 and E8;E2C proteins repress transcription through distinct mechanisms.


Asunto(s)
Regulación Viral de la Expresión Génica , Silenciador del Gen , Co-Represor 1 de Receptor Nuclear/metabolismo , Papillomaviridae/fisiología , Proteínas Represoras/metabolismo , Proteínas Virales/metabolismo , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Unión Proteica , Mapeo de Interacción de Proteínas , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo
17.
Virology ; 560: 96-109, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34051479

RESUMEN

Approximately 5% of cancers are caused by high-risk human papillomaviruses. Although very effective preventive vaccines will reduce this cancer burden significantly over the next several decades, they have no therapeutic effect for those already infected and remaining at risk for malignant progression of hrHPV lesions. HPV-associated cancers are dependent upon the expression of the viral E6 and E7 oncogenes. The oncogenic function of hrHPV E6 relies partially on its ability to induce p53 degradation. Since p53 is generally wildtype in hrHPV-associated cancers, p53 stabilization arrests proliferation, induces apoptosis and/or results in senescence. Here we describe a live cell, image-based high-throughput screen to identify compounds that stabilize p53 and/or affect viability in HPV-positive cancer HeLa cells. We validate the robustness and potential of this screening assay by assessing the activities of approximately 6,500 known bioactive compounds, illustrating its capability to function as a platform to identify novel therapeutics for hrHPV.


Asunto(s)
Aurora Quinasas/antagonistas & inhibidores , Quinasas Ciclina-Dependientes/antagonistas & inhibidores , Ensayos Analíticos de Alto Rendimiento/métodos , Inhibidores de Histona Desacetilasas/farmacología , Inhibidores de Topoisomerasa/farmacología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proteínas de Unión al ADN/metabolismo , Femenino , Células HeLa , Papillomavirus Humano 18/genética , Humanos , Proteínas Oncogénicas Virales/metabolismo , Infecciones por Papillomavirus/diagnóstico , Infecciones por Papillomavirus/diagnóstico por imagen , Infecciones por Papillomavirus/patología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/virología
18.
J Virol ; 83(17): 8885-92, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553310

RESUMEN

Proteomic identification of human papillomavirus type 16 (HPV16) E6-interacting proteins revealed several proteins involved in ubiquitin-mediated proteolysis. In addition to the well-characterized E6AP ubiquitin-protein ligase, a second HECT domain protein (HERC2) and a deubiquitylating enzyme (USP15) were identified by tandem affinity purification of HPV16 E6-associated proteins. This study focuses on the functional consequences of the interaction of E6 with USP15. Overexpression of USP15 resulted in increased levels of the E6 protein, and the small interfering RNA-mediated knockdown of USP15 decreased E6 protein levels. These results implicate USP15 directly in the regulation of E6 protein stability and suggest that ubiquitylated E6 could be a substrate for USP15 ubiquitin peptidase activity. It remains possible that E6 could affect the activity of USP15 on specific cellular substrates, a hypothesis that can be tested as more is learned about the substrates and pathways controlled by USP15.


Asunto(s)
Endopeptidasas/fisiología , Interacciones Huésped-Patógeno , Papillomavirus Humano 16/inmunología , Proteínas Oncogénicas Virales/metabolismo , Proteínas Represoras/metabolismo , Línea Celular , Endopeptidasas/genética , Técnicas de Silenciamiento del Gen/métodos , Silenciador del Gen , Humanos , Estabilidad Proteica , Proteasas Ubiquitina-Específicas
19.
J Virol ; 83(17): 8683-92, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19553317

RESUMEN

The papillomavirus (PV) E2 protein is an important regulator of the viral life cycle. It has diverse roles in viral transcription, DNA replication, and genome maintenance. Our laboratory has previously identified the cellular bromodomain protein Brd4 as a key interacting partner of E2. Brd4 mediates the transcriptional activation function of E2 and plays an important role in viral genome maintenance in dividing cells. E2 interacts with the C-terminal domain (CTD) of Brd4, and the CTD functions in a dominant-negative manner through binding E2 and interfering with E2's interaction with the full-length Brd4 protein. Previous studies have shown that PV E2 proteins are short lived; however, the mechanisms regulating their stability and degradation have not yet been well established. In this study, we explored the role of Brd4 in the regulation of bovine PV 1 (BPV1) and human PV 16 (HPV16) E2 stability. Expression of the Brd4 CTD dramatically increases E2 levels. Both BPV1 E2 and HPV16 E2 are regulated by ubiquitylation, and Brd4 CTD expression blocks this ubiquitylation, thus stabilizing the E2 protein. Furthermore, we have identified the cullin-based E3 ligases and specifically cullin-3 as potential components of the ubiquitylation machinery that targets both BPV1 and HPV16 E2 for ubiquitylation. Expression of the Brd4 CTD blocks the interaction between E2 and the cullin-3 complex. In addition to Brd4's role in mediating E2 transcription and genome tethering activities, these data suggest a potential role for Brd4 in regulating E2 stability and protein levels within PV-infected cells.


Asunto(s)
Papillomavirus Bovino 1/fisiología , Proteínas de Unión al ADN/metabolismo , Papillomavirus Humano 16/fisiología , Proteínas Nucleares/fisiología , Proteínas Oncogénicas Virales/metabolismo , Factores de Transcripción/fisiología , Proteínas Virales/metabolismo , Animales , Bovinos , Proteínas de Ciclo Celular , Proteínas Cullin/metabolismo , Humanos , Estabilidad Proteica , Ubiquitinación
20.
BMC Biochem ; 9: 4, 2008 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-18234089

RESUMEN

BACKGROUND: The delivery of ubiquitinated proteins to the proteasome for degradation is a key step in the regulation of the ubiquitin-proteasome pathway, yet the mechanisms underlying this step are not understood in detail. The Rad23 family of proteins is known to bind ubiquitinated proteins through its two ubiquitin-associated (UBA) domains, and may participate in the delivery of ubiquitinated proteins to the proteasome through docking via the Rad23 ubiquitin-like (UBL) domain. RESULTS: In this study, we investigate how the interaction between the UBL and UBA domains may modulate ubiquitin recognition and the delivery of ubiquitinated proteins to the proteasome by autoinhibition. We have explored a competitive binding model using specific mutations in the UBL domain. Disrupting the intramolecular UBL-UBA domain interactions in HHR23A indeed potentiates ubiquitin-binding. Additionally, the analogous surface on the Rad23 UBL domain overlaps with that required for interaction with both proteasomes and the ubiquitin ligase Ufd2. We have found that mutation of residues on this surface affects the ability of Rad23 to deliver ubiquitinated proteins to the proteasome. CONCLUSION: We conclude that the competition of ubiquitin-proteasome pathway components for surfaces on Rad23 is important for the role of the Rad23 family proteins in proteasomal targeting.


Asunto(s)
Unión Competitiva , Enzimas Reparadoras del ADN/química , Enzimas Reparadoras del ADN/metabolismo , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Ubiquitina/metabolismo , Unión Competitiva/genética , Humanos , Modelos Biológicos , Mutación , Poliubiquitina/metabolismo , Estructura Terciaria de Proteína/genética , Ubiquitina/genética , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitinación/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA