Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Oncol ; 2023: 9870174, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37854773

RESUMEN

[This corrects the article DOI: 10.1155/2021/6461552.].

2.
Virol Sin ; 37(6): 913-921, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36075565

RESUMEN

The AKT/mTOR and NF-κB signalings are crucial pathways activated in cancers including nasopharyngeal carcinoma (NPC), which is prevalent in southern China and closely related to Epstein-Barr virus (EBV) infection. How these master pathways are persistently activated in EBV-associated NPC remains to be investigated. Here we demonstrated that EBV-encoded latent membrane protein 1 (LMP1) promoted cyclophilin A (CYPA) expression through the activation of NF-κB. The depletion of CYPA suppressed cell proliferation and facilitated apoptosis. CYPA was able to bind to AKT1, thus activating AKT/mTOR/NF-κB signaling cascade. Moreover, the use of mTOR inhibitor, rapamycin, subverted the activation of the positive feedback loop, NF-κB/CYPA/AKT/mTOR. It is reasonable that LMP1 expression derived from initial viral infection is enough to assure the constant potentiation of AKT/mTOR and NF-κB signalings. This may partly explain the fact that EBV serves as a tumor-promoting factor with minimal expression of the viral oncoprotein LMP1 in malignancies. Our findings provide new insight into the understanding of causative role of EBV in tumorigenicity during latent infection.


Asunto(s)
Ciclofilina A , Infecciones por Virus de Epstein-Barr , Neoplasias Nasofaríngeas , Humanos , Ciclofilina A/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/fisiología , Carcinoma Nasofaríngeo/etiología , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , FN-kappa B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Transducción de Señal/fisiología
3.
Mol Neurobiol ; 59(3): 1925-1937, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35032318

RESUMEN

N6-methyladenosine (m6A) is a dynamic reversible methylation modification of the adenosine N6 position and is the most common chemical epigenetic modification among mRNA post-transcriptional modifications, including methylation, demethylation, and recognition. Post-transcriptional modification involves multiple protein molecules, including METTL3, METTL14, WTAP, KIAA1429, ALKBH5, YTHDF1/2/3, and YTHDC1/2. m6A-related proteins are expressed in almost all cells. However, the abnormal expression of m6A-related proteins may occur in the nervous system, thereby affecting neuritogenesis, brain volume, learning and memory, memory formation and consolidation, etc., and is implicated in the development of diseases, such as Parkinson's disease, Alzheimer's disease, multiple sclerosis, depression, epilepsy, and brain tumors. This review focuses on the functions of m6A in the development of central nervous system diseases, thus contributing to a deeper understanding of disease pathogenesis and providing potential clinical therapeutic targets for neurological diseases.


Asunto(s)
Adenosina , Metiltransferasas , Adenosina/análogos & derivados , Adenosina/metabolismo , Epigénesis Genética , Metilación , Metiltransferasas/metabolismo
4.
J Oncol ; 2021: 6461552, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34858499

RESUMEN

N6-Methyladenosine (m6A) modification is a dynamic and reversible methylation modification at the N6-position of adenosine. As one of the most prevalent posttranscriptional methylation modifications of RNA, m6A modification participates in several mRNA processes, including nuclear export, splicing, translation, and degradation. Some proteins, such as METTL3, METTL14, WTAP, ALKBH5, FTO, and YTHDF1/2/3, are involved in methylation. These proteins are subdivided into writers (METTL3, METTL14, WTAP), erasers (ALKBH5, FTO), and readers (YTHDF1/2/3) according to their functions in m6A modification. Several studies have shown that abnormal m6A modification occurs in tumors, including colorectal cancer, liver cancer, breast cancer, nasopharyngeal carcinoma, and gastric cancer. The proteins for m6A modification are involved in tumor proliferation, angiogenesis, metastasis, immunity, and other processes. Herein, the roles of m6A modification in cancer are discussed, which will improve the understanding of tumorigenesis, as well as the diagnosis, treatment, and prognosis of tumors.

5.
Front Cell Neurosci ; 15: 743353, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34588957

RESUMEN

Exosomes are a subset of extracellular vesicles that act as messengers to facilitate communication between cells. Non-coding RNAs, proteins, lipids, and microRNAs are delivered by the exosomes to target molecules (such as proteins, mRNAs, or DNA) of host cells, thereby playing a key role in the maintenance of normal brain function. However, exosomes are also involved in the occurrence, prognosis, and clinical treatment of brain diseases, such as Alzheimer's disease, Parkinson's disease, stroke, and traumatic brain injury. In this review, we have summarized novel findings that elucidate the role of exosomes in the occurrence, prognosis, and treatment of brain diseases.

6.
Front Mol Biosci ; 8: 816098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35083281

RESUMEN

Epstein-Barr virus (EBV), also known as human herpesvirus 4, is a double-stranded DNA virus that is ubiquitous in 90-95% of the population as a gamma herpesvirus. It exists in two main states, latent infection and lytic replication, each encoding viral proteins with different functions. Human B-lymphocytes and epithelial cells are EBV-susceptible host cells. EBV latently infects B cells and nasopharyngeal epithelial cells throughout life in most immunologically active individuals. EBV-infected cells, free viruses, their gene products, and abnormally elevated EBV titers are observed in the cerebrospinal fluid. Studies have shown that EBV can infect neurons directly or indirectly via infected B-lymphocytes, induce neuroinflammation and demyelination, promote the proliferation, degeneration, and necrosis of glial cells, promote proliferative disorders of B- and T-lymphocytes, and contribute to the occurrence and development of nervous system diseases, such as Alzheimer's disease, Parkinson's disease, multiple sclerosis, acute cerebellar ataxia, meningitis, acute disseminated encephalomyelitis, and brain tumors. However, the specific underlying molecular mechanisms are unclear. In this paper, we review the mechanisms underlying the role of EBV in the development of central nervous system diseases, which could bebeneficial in providing new research ideas and potential clinical therapeutic targets for neurological diseases.

7.
Mol Ther Nucleic Acids ; 17: 175-184, 2019 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-31265948

RESUMEN

Nasopharyngeal carcinoma (NPC) is an invasive head-and-neck tumor with Epstein-Barr virus (EBV) as an important etiological cause. The EBV oncoprotein Latent membrane protein 1 (LMP1) can be trafficked into exosomes with unclear roles, and this trafficking is a potential problem in NPC control. MicroRNA-203 (miR-203) was found by us to be downregulated by LMP1, and it functions as a tumor suppressor in NPC. In this study, aspirin reversed the epithelial-mesenchymal transition (EMT) by promoting miR-203 expression in cells, and, remarkably, it repressed exosomal LMP1 (exo-LMP1) secretion from EBV-positive cells. Nuclear factor κB (NF-κB) activation was required for the exo-LMP1 production. The exo-LMP1 uptake influenced the EMT potential of EBV-negative recipient NPC cells. The exo-LMP1 level was upregulated in clinical NPC plasma samples. Aspirin treatment observably inhibited NPC lung metastasis in nude mice. The study revealed that aspirin is a promising drug for NPC therapy via its targeting of exo-LMP1 transfer and the regulatory effect of LMP1 on miR-203 expression. EBV can regulate its own tumorigenesis via the LMP1/NF-κB/exo-LMP1 axis, opening a new avenue for understanding the pathogenesis of this tumor virus. Our study also provides a rationale for the use of exo-LMP1 or exosomal miR-203 (exo-miR203) in EBV-targeted therapy by aspirin in invasive NPC.

8.
J Med Virol ; 91(10): 1845-1855, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31199512

RESUMEN

Epstein-Barr virus (EBV) is a prevalent γ-herpesvirus associated with a variety of cancers, including epithelial nasopharyngeal carcinoma (NPC) and gastric carcinoma (GC). Long noncoding RNAs (lncRNAs) are emerging as powerful regulators that have demonstrated to play crucial roles in cancer development. In this approach, based on genome-wide RNA sequencing, we examined the lncRNA expression profiles in four EBV genome-infected and EBV-negative 293 cells. A series of lncRNAs were found to be dysregulated in a comparative analysis. Then, eight typical lncRNAs were selected for validation of expression levels by real-time quantitative polymerase chain reaction and were detected in EBV-positive or EBV-negative GC and NPC cells. The differential expression patterns of the eight lncRNAs for validation were fundamentally identical to those revealed in RNA sequencing. Particularly, the differential expression of these lncRNAs in GC and NPC cells indicated their possible roles in EBV infection and tumorigenesis. In addition, a predicted lncRNA-microRNA-messenger RNA network suggested their potential interactions. This study reveals the first lncRNAome related to EBV infection in the epithelial cells and provides novel clues for the study of viral role in epithelial cancers through the interaction between EBV and host lncRNAs.


Asunto(s)
Infecciones por Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4 , ARN Largo no Codificante/metabolismo , Transcriptoma , Línea Celular , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , ARN Largo no Codificante/genética , Análisis de Secuencia de ARN
9.
Cancer Med ; 8(6): 3142-3151, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31063269

RESUMEN

Exosomes have emerged as novel vehicles for proteins and other contents in cancer progression. Cyclophilin A (CYPA) is a pivotal member of immunophilin family. Whether CYPA can be detected in sera of nasopharyngeal carcinoma (NPC) patients remains to be explored. Epstein-Barr virus (EBV) is the first identified human tumor virus and is a causative agent of NPC. The antibody of EBV capsid antigen immunoglobulin A (EBV-VCA-IgA) is a known biomarker of NPC, with a proportion of no more than 70% being detected positively. Hence, novel biomarkers need to be discovered for early diagnosis, prognosis, and monitoring of EBV-associated NPC. A total of 110 NPC and 36 normal control serum samples were collected. Exosomes from these samples were extracted. The mRNA and protein expression levels of the above samples were validated by reverse transcription -quantitative polymerase chain reaction, Western blotting, or enzyme-linked immunosorbent assay (ELISA). Finally, the results demonstrated that both the serum and exosomal CYPA levels of NPC patients were significantly higher than that of normal cases. In addition, exosomal CYPA had a much higher level than that in the whole sera. The positive rate of EBV-VCA-IgA antibody was 68.2% in NPC sera, and noticeably, among the cases with EBV-VCA-IgA negative, 80% of them presented high levels of CYPA above the standard (cutoff value). In particular, CYPA in exosomes was uniformly with higher significance than that in whole sera. Combined analysis of CYPA protein and EBV-VCA-IgA antibody showed a greatly higher discriminatory ability in diagnosis of NPC. Moreover, exosomal CYPA level had a positive correlation with that of the EBV-encoded latent membrane protein 1 (LMP1) in exosomes. EBV-positive cancer cells secreted significantly higher levels of exosomal CYPA. This study established the utility of circulating exosomal CYPA as a potential noninvasive diagnostic biomarker for EBV-associated NPC.


Asunto(s)
Ciclofilina A/metabolismo , Infecciones por Virus de Epstein-Barr/complicaciones , Exosomas/metabolismo , Herpesvirus Humano 4 , Carcinoma Nasofaríngeo/diagnóstico , Carcinoma Nasofaríngeo/metabolismo , Biomarcadores , Línea Celular Tumoral , Ciclofilina A/genética , Infecciones por Virus de Epstein-Barr/virología , Exosomas/ultraestructura , Femenino , Expresión Génica , Humanos , Masculino , Carcinoma Nasofaríngeo/etiología , Neoplasias Nasofaríngeas/diagnóstico , Neoplasias Nasofaríngeas/etiología , Neoplasias Nasofaríngeas/metabolismo , Pronóstico , Curva ROC
10.
Virol Sin ; 34(3): 253-261, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-30911896

RESUMEN

Epstein-Barr virus (EBV) is an important human dsDNA virus, which has been shown to be associated with several malignancies including about 10% of gastric carcinomas. How EBV enters an epithelial cell has been an interesting project for investigation. "Cell-in-cell" infection was recently reported an efficient way for the entry of EBV into nasopharynx epithelial cells. The present approach was to explore the feasibility of this mode for EBV infection in gastric epithelial cells and the dynamic change of host inflammatory reaction. The EBV-positive lymphoblastic cells of Akata containing a GFP tag in the viral genome were co-cultured with the gastric epithelial cells (GES-1). The infection situation was observed under fluorescence and electron microscopies. Real-time quantitative PCR and Western-blotting assay were employed to detect the expression of a few specific cytokines and inflammatory factors. The results demonstrated that EBV could get into gastric epithelial cells by "cell-in-cell" infection but not fully successful due to the host fighting. IL-1ß, IL-6 and IL-8 played prominent roles in the cellular response to the infection. The activation of NF-κB and HSP70 was also required for the host antiviral response. The results imply that the gastric epithelial cells could powerfully resist the virus invader via cell-in-cell at the early stage through inflammatory and innate immune responses.


Asunto(s)
Formación de la Célula en Célula , Células Epiteliales/virología , Tracto Gastrointestinal/virología , Herpesvirus Humano 4/fisiología , Línea Celular , Citocinas/inmunología , Células Epiteliales/inmunología , Fluorescencia , Tracto Gastrointestinal/citología , Proteínas Fluorescentes Verdes , Proteínas del Choque Térmico HSP72/metabolismo , Herpesvirus Humano 4/genética , Humanos , Inmunidad Innata , Hibridación in Situ , Inflamación , Microscopía Electrónica de Transmisión , FN-kappa B/metabolismo
11.
Front Microbiol ; 10: 2879, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31921057

RESUMEN

Epstein-Barr virus (EBV) nuclear antigen 1 (EBNA1)-mediated DNA episomal genome replication and persistence are essential for the viral pathogenesis. Cyclophilin A (CYPA) is upregulated in EBV-associated nasopharyngeal carcinoma (NPC) with unknown roles. In the present approach, cytosolic CYPA was found to be bound with EBNA1 into the nucleus. The amino acid 376-459 of the EBNA1 domain was important for the binding. CYPA depletion attenuated and ectopic CYPA expression improved EBNA1 expression in EBV-positive cells. The loss of viral copy number was also accelerated by CYPA consumption in daughter cells during culture passages. Mechanistically, CYPA mediated the connection of EBNA1 with oriP (origin of EBV DNA replication) and subsequent oriP transcription, which is a key step for the initiation of EBV genome replication. Moreover, CYPA overexpression markedly antagonized the connection of EBNA1 to Ubiquitin-specific protease 7 (USP7), which is a strong host barrier with a role of inhibiting EBV genome replication. The PPIase activity of CYPA was required for the promotion of oriP transcription and antagonism with USP7. The results revealed a strategy that EBV recruited a host factor to counteract the host defense, thus facilitating its own latent genome replication. This study provides a new insight into EBV pathogenesis and potential virus-targeted therapeutics in EBV-associated NPC, in which CYPA is upregulated at all stages.

12.
Virol Sin ; 32(5): 349-356, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29116589

RESUMEN

Herpesviruses are remarkable pathogens that have evolved multiple mechanisms to evade host immunity, ensuring their proliferation and egress. Among these mechanisms, herpesviruses utilize elaborate extracellular vesicles, including exosomes, for the intricate interplay between infected host and recipient cells. Herpesviruses incorporate genome expression products and direct cellular products into exosomal cargoes. These components alter the content and function of exosomes released from donor cells, thus affecting the downstream signalings of recipient cells. In this way, herpesviruses hijack exosomal pathways to ensure their survival and persistence, and exosomes are emerging as critical mediators for virus infection-associated intercellular communication and microenvironment alteration. In this review, the function and effects of exosomes in herpesvirus infection will be discussed, so that we will have a better understanding about the pathogenesis of herpesviruses.


Asunto(s)
Exosomas/metabolismo , Vesículas Extracelulares/metabolismo , Infecciones por Herpesviridae/metabolismo , Animales , Exosomas/patología , Infecciones por Herpesviridae/patología , Humanos , Transducción de Señal/fisiología
13.
Virol Sin ; 32(3): 175-187, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28669004

RESUMEN

Epstein-Barr virus (EBV) is an oncogenic virus that ubiquitously establishes life-long persistence in humans. To ensure its survival and maintain its B cell transformation function, EBV has developed powerful strategies to evade host immune responses. Emerging evidence has shown that microRNAs (miRNAs) are powerful regulators of the maintenance of cellular homeostasis. In this review, we summarize current progress on how EBV utilizes miRNAs for immune evasion. EBV encodes miRNAs targeting both viral and host genes involved in the immune response. The miRNAs are found in two gene clusters, and recent studies have demonstrated that lack of these clusters increases the CD4+ and CD8+ T cell response of infected cells. These reports strongly indicate that EBV miRNAs are critical for immune evasion. In addition, EBV is able to dysregulate the expression of a variety of host miRNAs, which influence multiple immune-related molecules and signaling pathways. The transport via exosomes of EBV-regulated miRNAs and viral proteins contributes to the construction and modification of the inflammatory tumor microenvironment. During EBV immune evasion, viral proteins, immune cells, chemokines, pro-inflammatory cytokines, and pro-apoptosis molecules are involved. Our increasing knowledge of the role of miRNAs in immune evasion will improve the understanding of EBV persistence and help to develop new treatments for EBV-associated cancers and other diseases.


Asunto(s)
Regulación de la Expresión Génica , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Evasión Inmune , MicroARNs/metabolismo , Humanos , ARN Viral/metabolismo
14.
Oncotarget ; 6(38): 41033-44, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26517512

RESUMEN

A tumor model that Epstein-Barr virus (EBV) latent infection facilitated the tumorigenicity was previously established using the Maxi-EBV system. In the present approach, EBV-lost cell clones demonstrated significantly decreased tumorigenesis. On the other hand, the LMP1 gene in Maxi-EBV genome was replaced by that of nasopharyngeal carcinoma origin. The resultant cell line, 293-1/NL showed much lower malignancy than the original 293-EBV. The result was opposite to our expectation. The change of 293 sublineage cells for EBV harboring also got similar result. To seek the underlying reason, the copy number of EBV genome in all the cell lines was detected. The result indicated that 293-EBV contained about 4.5-fold higher EBV copies than 293-1/NL did. Parallel EBV genomes led to relatively stable copies in different 293 sublineages, suggesting the viral genome structure is a factor for the sustainability of EBV's copy number. Moreover, the LMP1 transcription in high copy-containing cells showed abnormally high level. Furthermore, the main LMP1-driven pathway, transcription factor NF-κB, was highly activated in high-copy cells. Here we first manifest by experimental model that the copy number of EBV latent genome correlates with the viral pathogenesis, which depends on the activation level of LMP1 and NF-κB. Overall, both the presence and amount of EBV genome are crucial for the viral oncogenicity.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Herpesvirus Humano 4/metabolismo , FN-kappa B/metabolismo , Proteínas de la Matriz Viral/metabolismo , Animales , Western Blotting , Dosificación de Gen , Regulación Viral de la Expresión Génica , Genoma Viral/genética , Células HEK293 , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiología , Interacciones Huésped-Patógeno , Humanos , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Trasplante Heterólogo , Carga Tumoral , Proteínas de la Matriz Viral/genética , Proteínas de la Matriz Viral/fisiología , Latencia del Virus/genética
15.
Bing Du Xue Bao ; 30(4): 476-82, 2014 Jul.
Artículo en Chino | MEDLINE | ID: mdl-25272606

RESUMEN

Epstein-Barr virus (EBV) is a human herpesvirus associated with important human diseases, including infectious mononucleosis syndrome, malignant lymphoma, and nasopharyngeal carcinoma. The mechanism of EBV entry into host cells remains a subject of intensive research. After decades of study, researchers have identified several key proteins and different patterns of EBV intrusion into host cells. The viral surface glycoproteins, gp350/220, gp42, gB, gH, and gL, are involved in interactions with the CR2 receptor on the surface of B lymphocytes during viral entry. However, the majority of epithelial cells lack CR2 receptor expression, which makes viral invasion much more complex than in B lymphocytes. Three different models have been proposed to explain how EBV enters epithelial cells: (1) "transfer of infection", mediated by B lymphocytes or Langerhans cells; (2) EBV utilizes its own proteins during the process of fusion with the cell membrane; and (3) progeny virions arising from EBV-infected epithelial cells cross lateral membranes into adjacent epithelial cells. This review will discuss the relevant mechanism of viral entry into B lymphocytes and epithelial cells during EBV infection.


Asunto(s)
Linfocitos B/virología , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Internalización del Virus , Animales , Herpesvirus Humano 4/genética , Humanos , Proteínas Virales/genética , Proteínas Virales/metabolismo
16.
J Virol ; 86(6): 3088-99, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22205737

RESUMEN

The Epstein-Barr virus (EBV) is highly associated with nasopharyngeal carcinoma (NPC), and it regulates some microRNAs (miRNAs) that are involved in the development of cancer. The role of EBV in the deregulation of cellular miRNAs and how this affects the progression of NPC remain to be investigated. An analysis of the miRNA profile in an EBV-infected cell line revealed that miRNA 203 (miR-203) was downregulated. miR-203 is expressed specifically in epithelial cells. This downregulation of miR-203 was further verified and functionally analyzed. miR-203 was downregulated substantially in epithelial cells and NPC tissues that were latently infected with EBV. Downregulation of miR-203 also occurred during the early stage of EBV infection. Furthermore, the viral oncoprotein, latent membrane protein 1 (LMP1), was responsible for downregulation of miR-203. Removal of the latent EBV genome or suppression of LMP1 resulted in restoration of miR-203 expression. EBV-LMP1 mediated the downregulation of miR-203 at the primary transcript level. E2F3 and CCNG1 were identified as target genes of miR-203. Ectopic expression of miR-203 inhibited EBV-induced S-phase entry and transformation in vivo. Overexpression of the targets overcame the effects of miR-203 mimics on the cell cycle, and the expression of target genes in tumor models was inhibited by miR-203. Inhibitors of Jun N-terminal protein kinase (JNK) and NF-κB blocked miR-203 downregulation. These results imply that EBV promotes malignancy by downregulating cellular miR-203, which contributes to the etiology of NPC.


Asunto(s)
Regulación hacia Abajo , Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/metabolismo , MicroARNs/genética , Proteínas de la Matriz Viral/metabolismo , Animales , Células Epiteliales/patología , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Infecciones por Virus de Epstein-Barr/virología , Femenino , Herpesvirus Humano 4/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , MicroARNs/metabolismo , Proteínas de la Matriz Viral/genética , Latencia del Virus
17.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 36(7): 624-30, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21873786

RESUMEN

OBJECTIVE: To determine the maintenance and loss of Epstein-Barr virus (EBV) genome during the clonal expansion of the EBV-infected epithelial cells. METHODS: The epithelial tumor cell line, 293-EBV, in which the EBV genome was observed with green fluorescent protein (GFP) readout. After a dozen of passages, it contained cells with strong or weak GFP expression, and some with complete loss of EBV genome. The cell growth was then continuously observed under a confocal microscope. The cell dividing and GFP expression were also observed during the clonal expansion by being made into very low density. RESULTS: The cells moved around due to adherence and mobility, while the GFP expression remained unchanged in the undivided cells. The cells could form compact or loosen clones. The EBV genome easily persisted in those clones when cells were growing compactly. As the cell number increased, the GFP expression became weak or even died away at the sites of low density in the loosen clones. CONCLUSION: EBV-positive epithelial cells are able to sustain the EBV genome during its clonal expansion. The cells maintain EBV genomes by passing them to the daughter cells after replication. When the cells unsuccessfully inherit the EBV genome, the daughter cells may lose them which is related to the low cell density as well as the epithelial environment.


Asunto(s)
Carcinoma/virología , Genoma Viral , Herpesvirus Humano 4/fisiología , Neoplasias/virología , Latencia del Virus/genética , Línea Celular Tumoral , Transformación Celular Viral , Células Clonales , Células Epiteliales/virología , Herpesvirus Humano 4/genética , Humanos
18.
J Virol Methods ; 176(1-2): 103-7, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21723326

RESUMEN

The Maxi-EBV is a bacterial artificial chromosome (BAC)-based plasmid that contains the complete Epstein-Barr virus (EBV) genome of 172 kb. This plasmid also carries an additional cassette of 11.5 kb in size for the expression of a mini F factor, selection markers and GFP. In the intracellular study of EBV infection based on the Maxi-EBV system, a parallel control that only contains this 11.5 kb vector is desirable but unavailable. In order to construct the vector in this approach, a clean deletion of the complete EBV genome from the Maxi-EBV was performed. This was achieved by homologous recombination using the bacteriophage λ Red system. Initially, an FRT-flanked kanamycin-resistance (kan) fragment of 1.4 kb with 61 bp homologies on the ends was introduced into the Maxi-EBV plasmid, replacing the 172-kb EBV genome. The kan gene was then removed by Flp/FRT excision. The results of identification demonstrated that the mutation was generated precisely. The results highlight the feasibility for a genome as large as 172 kb to be replaced by a greatly shorter fragment and for a much smaller vector backbone to be retrieved. Cell lines derived from the transfection of the vector will subsequently be appropriate controls in the related study.


Asunto(s)
Cromosomas Artificiales Bacterianos/genética , Genoma Viral/genética , Herpesvirus Humano 4/genética , Recombinación Homóloga , Plásmidos , Eliminación de Secuencia , Animales , Bacteriófago lambda/enzimología , Línea Celular , Ingeniería Genética/métodos , Vectores Genéticos , Humanos , Ratones , Ratones Desnudos , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...