Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
PLoS Pathog ; 20(2): e1011954, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38300891

RESUMO

Epstein-Barr virus (EBV) is a ubiquitous human lymphotropic herpesvirus that is causally associated with several malignancies. In addition to latent factors, lytic replication contributes to cancer development. In this study, we examined whether the lytic gene BNRF1, which is conserved among gamma-herpesviruses, has an important role in lymphomagenesis. We found that lymphoblastoid cell lines (LCLs) established by BNRF1-knockout EBV exhibited remarkably lower pathogenicity in a mice xenograft model than LCLs produced by wild-type EBV (LCLs-WT). RNA-seq analyses revealed that BNRF1 elicited the expression of interferon-inducible protein 27 (IFI27), which promotes cell proliferation. IFI27 knockdown in LCLs-WT resulted in excessive production of reactive oxygen species, leading to cell death and significantly decreased their pathogenicity in vivo. We also confirmed that IFI27 was upregulated during primary infection in B-cells. Our findings revealed that BNRF1 promoted robust proliferation of the B-cells that were transformed by EBV latent infection via IFI27 upregulation both in vitro and in vivo.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesviridae , Humanos , Animais , Camundongos , Herpesvirus Humano 4 , Interferons/metabolismo , Regulação para Cima , Herpesviridae/metabolismo , Latência Viral , Proteínas de Membrana/metabolismo
2.
Nature ; 565(7739): 312-317, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30602793

RESUMO

Clonal expansion in aged normal tissues has been implicated in the development of cancer. However, the chronology and risk dependence of the expansion are poorly understood. Here we intensively sequence 682 micro-scale oesophageal samples and show, in physiologically normal oesophageal epithelia, the progressive age-related expansion of clones that carry mutations in driver genes (predominantly NOTCH1), which is substantially accelerated by alcohol consumption and by smoking. Driver-mutated clones emerge multifocally from early childhood and increase their number and size with ageing, and ultimately replace almost the entire oesophageal epithelium in the extremely elderly. Compared with mutations in oesophageal cancer, there is a marked overrepresentation of NOTCH1 and PPM1D mutations in physiologically normal oesophageal epithelia; these mutations can be acquired before late adolescence (as early as early infancy) and significantly increase in number with heavy smoking and drinking. The remodelling of the oesophageal epithelium by driver-mutated clones is an inevitable consequence of normal ageing, which-depending on lifestyle risks-may affect cancer development.


Assuntos
Envelhecimento/genética , Envelhecimento/patologia , Epitélio , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patologia , Mutação , Lesões Pré-Cancerosas/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Consumo de Bebidas Alcoólicas/genética , Biópsia , Contagem de Células , Transformação Celular Neoplásica/genética , Criança , Pré-Escolar , Células Clonais/metabolismo , Células Clonais/patologia , Variações do Número de Cópias de DNA , Epitélio/metabolismo , Epitélio/patologia , Evolução Molecular , Feminino , Interação Gene-Ambiente , Genoma Humano/genética , Humanos , Lactente , Estilo de Vida , Masculino , Pessoa de Meia-Idade , Acúmulo de Mutações , Proteína Fosfatase 2C/genética , Receptor Notch1/genética , Fatores de Risco , Análise de Sequência de DNA , Análise de Célula Única , Fumar/genética , Adulto Jovem
3.
J Clin Immunol ; 44(4): 103, 2024 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-38642164

RESUMO

Epstein-Barr virus (EBV) infection can lead to infectious mononucleosis (EBV-IM) and, more rarely, EBV-associated hemophagocytic lymphohistiocytosis (EBV-HLH), which is characterized by a life-threatening hyperinflammatory cytokine storm with immune dysregulation. Interferon-gamma (IFNγ) has been identified as a critical mediator for primary HLH; however, the detailed role of IFNγ and other cytokines in EBV-HLH is not fully understood. In this study, we used single-cell RNA sequencing to characterize the immune landscape of EBV-HLH and compared it with EBV-IM. Three pediatric patients with EBV-HLH with different backgrounds, one with X-linked lymphoproliferative syndrome type 1 (XLP1), two with chronic active EBV disease (CAEBV), and two patients with EBV-IM were enrolled. The TUBA1B + STMN1 + CD8 + T cell cluster, a responsive proliferating cluster with rich mRNA detection, was explicitly observed in EBV-IM, and the upregulation of SH2D1A-the gene responsible for XLP1-was localized in this cluster. This proliferative cluster was scarcely observed in EBV-HLH cases. In EBV-HLH cases with CAEBV, upregulation of LAG3 was observed in EBV-infected cells, which may be associated with an impaired response by CD8 + T cells. Additionally, genes involved in type I interferon (IFN) signaling were commonly upregulated in each cell fraction of EBV-HLH, and activation of type II IFN signaling was observed in CD4 + T cells, natural killer cells, and monocytes but not in CD8 + T cells in EBV-HLH. In conclusion, impaired responsive proliferation of CD8 + T cells and upregulation of type I IFN signaling were commonly observed in EBV-HLH cases, regardless of the patients' background, indicating the key features of EBV-HLH.


Assuntos
Infecções por Vírus Epstein-Barr , Linfo-Histiocitose Hemofagocítica , Transtornos Linfoproliferativos , Humanos , Criança , Herpesvirus Humano 4 , Linfo-Histiocitose Hemofagocítica/diagnóstico , Linfo-Histiocitose Hemofagocítica/genética , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/genética , Linfócitos T CD8-Positivos , Interferon gama/genética , Transtornos Linfoproliferativos/diagnóstico , Transtornos Linfoproliferativos/genética , Transtornos Linfoproliferativos/complicações , Perfilação da Expressão Gênica
4.
J Virol ; 97(6): e0043723, 2023 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-37195206

RESUMO

Enveloped viruses undergo a complex multistep process of assembly, maturation, and release into the extracellular space utilizing host secretory machinery. Several studies of the herpesvirus subfamily have shown that secretory vesicles derived from the trans-Golgi network (TGN) or endosomes transport virions into the extracellular space. However, the regulatory mechanism underlying the release of Epstein-Barr virus, a human oncovirus, remains unclear. We demonstrate that disruption of BBLF1, a tegument component, suppressed viral release and resulted in the accumulation of viral particles on the inner side of the vesicular membrane. Organelle separation revealed the accumulation of infectious viruses in fractions containing vesicles derived from the TGN and late endosomes. Deficiency of an acidic amino acid cluster in BBLF1 reduced viral secretion. Moreover, truncational deletion of the C-terminal region of BBLF1 increased infectious virus production. These findings suggest that BBLF1 regulates the viral release pathway and reveal a new aspect of tegument protein function. IMPORTANCE Several viruses have been linked to the development of cancer in humans. Epstein-Barr virus (EBV), the first identified human oncovirus, causes a wide range of cancers. Accumulating literature has demonstrated the role of viral reactivation in tumorigenesis. Elucidating the functions of viral lytic genes induced by reactivation, and the mechanisms of lytic infection, is essential to understanding pathogenesis. Progeny viral particles synthesized during lytic infection are released outside the cell after the assembly, maturation, and release steps, leading to further infection. Through functional analysis using BBLF1-knockout viruses, we demonstrated that BBLF1 promotes viral release. The acidic amino acid cluster in BBLF1 was also important for viral release. Conversely, mutants lacking the C terminus exhibited more efficient virus production, suggesting that BBLF1 is involved in the fine-tuning of progeny release during the EBV life cycle.


Assuntos
Herpesvirus Humano 4 , Vesículas Secretórias , Proteínas Virais , Liberação de Vírus , Replicação Viral , Humanos , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Vesículas Secretórias/metabolismo , Vesículas Secretórias/virologia , Vírion/fisiologia , Replicação Viral/fisiologia , Células HEK293 , Proteínas Virais/metabolismo , Liberação de Vírus/genética
5.
J Virol ; 96(18): e0073922, 2022 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-36094314

RESUMO

Epstein-Barr virus (EBV) persists in human cells as episomes. EBV episomes are chromatinized and their 3D conformation varies greatly in cells expressing different latency genes. We used HiChIP, an assay which combines genome-wide chromatin conformation capture followed by deep sequencing (Hi-C) and chromatin immunoprecipitation (ChIP), to interrogate the EBV episome 3D conformation in different cancer cell lines. In an EBV-transformed lymphoblastoid cell line (LCL) GM12878 expressing type III EBV latency genes, abundant genomic interactions were identified by H3K27ac HiChIP. A strong enhancer was located near the BILF2 gene and looped to multiple genes around BALFs loci. Perturbation of the BILF2 enhancer by CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) altered the expression of BILF2 enhancer-linked genes, including BARF0 and BALF2, suggesting that this enhancer regulates the expression of linked genes. H3K27ac ChIP followed by deep sequencing (ChIP-seq) identified several strong EBV enhancers in T/NK (natural killer) lymphoma cells that express type II EBV latency genes. Extensive intragenomic interactions were also found which linked enhancers to target genes. A strong enhancer at BILF2 also looped to the BALF loci. CRISPRi also validated the functional connection between BILF2 enhancer and BARF1 gene. In contrast, H3K27ac HiChIP found significantly fewer intragenomic interactions in type I EBV latency gene-expressing primary effusion lymphoma (PEL) cell lines. These data provided new insight into the regulation of EBV latency gene expression in different EBV-associated tumors. IMPORTANCE EBV is the first human DNA tumor virus identified, discovered over 50 years ago. EBV causes ~200,000 cases of various cancers each year. EBV-encoded oncogenes, noncoding RNAs, and microRNAs (miRNAs) can promote cell growth and survival and suppress senescence. Regulation of EBV gene expression is very complex. The viral C promoter regulates the expression of all EBV nuclear antigens (EBNAs), some of which are very far away from the C promoter. Another way by which the virus activates remote gene expression is through DNA looping. In this study, we describe the viral genome looping patterns in various EBV-associated cancer cell lines and identify important EBV enhancers in these cells. This study also identified novel opportunities to perturb and eventually control EBV gene expression in these cancer cells.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Plasmídeos , Latência Viral , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos/genética , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Antígenos Nucleares do Vírus Epstein-Barr/genética , Herpesvirus Humano 4/genética , Humanos , MicroRNAs/metabolismo , Neoplasias/virologia , Plasmídeos/química , Plasmídeos/genética , Plasmídeos/metabolismo , Proteínas Virais/genética , Latência Viral/genética
6.
J Virol ; 96(14): e0051822, 2022 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-35862711

RESUMO

Protein-protein interactions (PPIs) are crucial for various biological processes. Epstein-Barr virus (EBV) proteins typically form complexes, regulating the replication and persistence of the viral genome in human cells. However, the role of EBV protein complexes under physiological conditions remains unclear. In this study, we performed comprehensive analyses of EBV PPIs in living cells using the NanoBiT system. We identified 195 PPIs, many of which have not previously been reported. Computational analyses of these PPIs revealed that BLRF2, which is only found in gammaherpesviruses, is a central protein in the structural network of EBV tegument proteins. To characterize the role of BLRF2, we generated two BLRF2 knockout EBV clones using CRISPR/Cas9. BLRF2 knockout significantly decreased the production of infectious virus particles, which was partially restored by exogenous BLRF2 expression. In addition, self-association of BLRF2 protein was found, and mutation of the residues crucial for the self-association affected stability of the protein. Our data imply that BLRF2 is a tegument network hub that plays important roles in progeny virion maturation. IMPORTANCE EBV remains a significant public health challenge, causing infectious mononucleosis and several cancer types. Therefore, the better understanding of the molecular mechanisms underlying EBV replication is of high clinical importance. As protein-protein interactions (PPIs) are major regulators of virus-associated pathogenesis, comprehensive analyses of PPIs are essential. Previous studies on PPIs in EBV or other herpesviruses have predominantly employed the yeast two-hybrid (Y2H) system, immunoprecipitation, and pulldown assays. Herein, using a novel luminescence-based method, we identified 195 PPIs, most of which have not previously been reported. Computational and functional analyses using knockout viruses revealed that BLRF2 plays a central role in the EBV life cycle, which makes it a valuable target for drug development.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Mapas de Interação de Proteínas , Proteínas Virais , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Humanos , Proteínas Virais/genética , Replicação Viral
7.
Cell Commun Signal ; 20(1): 95, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35729616

RESUMO

BACKGROUND: Viruses must adapt to the environment of their host cells to establish infection and persist. Diverse mammalian cells, including virus-infected cells, release extracellular vesicles such as exosomes containing proteins and miRNAs, and use these vesicles to mediate intercellular communication. However, the roles of exosomes in viral infection remain unclear. RESULTS: We screened viral proteins to identify those responsible for the exosome-mediated enhancement of Epstein-Barr virus (EBV) infection. We identified BGLF2 protein encapsulated in exosomes, which were released by EBV-infected cells. BGLF2 protein is a tegument protein that exists in the space between the envelope and nucleocapsid, and it is released into the cytoplasm shortly after infection. BGLF2 protein-containing exosomes enhanced viral gene expression and repressed innate immunity, thereby supporting the EBV infection. CONCLUSIONS: The EBV tegument protein BGLF2 is encapsulated in exosomes and released by infected cells to facilitate the establishment of EBV infection. These findings suggest that tegument proteins support viral infection not only between the envelope and nucleocapsid, as well as in extraviral particles such as exosomes. Video abstract.


Assuntos
Infecções por Vírus Epstein-Barr , Exossomos , Animais , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Exossomos/metabolismo , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/metabolismo , Mamíferos/metabolismo , Proteínas Virais de Fusão , Proteínas Virais
8.
Cancer Sci ; 112(12): 5088-5099, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34609775

RESUMO

Epstein-Barr virus-associated lymphoproliferative disease (EBV-LPD) is frequently fatal. Innate immunity plays a key role in protecting against pathogens and cancers. The stimulator of interferon genes (STING) is regarded as a key adaptor protein allowing DNA sensors recognizing exogenous cytosolic DNA to activate the type I interferon signaling cascade. In terms of EBV tumorigenicity, the role of STING remains elusive. Here we showed that treatment with the STING inhibitor, C-176, suppressed EBV-induced transformation in peripheral blood mononuclear cells. In an EBV-LPD mouse model, C-176 treatment also inhibited tumor formation and prolonged survival. Treatment with B cells alone did not affect EBV transformation, but suppression of EBV-induced transformation was observed in the presence of T cells. Even without direct B cell-T cell contact in a transwell system, the inhibitor reduced the transformation activity, indicating that intercellular communication by humoral factors was critical to prevent EBV-induced transformation. These findings suggest that inhibition of STING signaling pathway with C-176 could be a new therapeutic target of EBV-LPD.


Assuntos
Antineoplásicos/administração & dosagem , Transformação Celular Viral/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Linfoma de Células B/prevenção & controle , Proteínas de Membrana/antagonistas & inibidores , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Infecções por Vírus Epstein-Barr/imunologia , Células HEK293 , Herpesvirus Humano 4 , Humanos , Células Jurkat , Linfoma de Células B/imunologia , Linfoma de Células B/virologia , Camundongos , Análise de Sobrevida , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Rev Med Virol ; 30(2): e2095, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31845495

RESUMO

Epstein-Barr virus (EBV) is a causative agent of infectious mononucleosis and several malignancies involving lymphocytes and epithelial cells. We recently reported genomic analyses of chronic active EBV infection (CAEBV), a proliferative disorder of T and/or NK cells, as well as other lymphoid malignancies. We found that T and/or NK cells undergoing clonal expansion in CAEBV patients gain somatic driver mutations as the disorder progresses. Investigation of the viral genome revealed viral genomes harboring intragenic deletions in the BamHI-rightward transcripts (BART) region and in essential lytic genes. Interestingly, we observed that these deletions resulted in leaky expression of viral lytic genes. This increased expression of viral lytic genes is reminiscent of the "pre-latent abortive lytic" state, in which a substantial number of lytic genes are produced for weeks in the absence of progeny production, which contributes to cell survival upon de novo infection. It has been known that EBV can choose either latent or lytic state, but this dualistic concept may need to be reconsidered, as our data suggest the presence of the third, intermediate state; leaky expression of lytic genes that does not lead to completion of the full lytic amplification cycle. Leaky expression of lytic genes likely contributes to the formation and maintenance of several types of EBV-associated tumors. We also presented significant circumstantial evidence suggesting that EBV infects lymphoid progenitor cells in CAEBV before differentiation into T and NK cells. Taken together, our new data shed light on oncogenesis of CAEBV and other EBV-associated malignancies.


Assuntos
Transformação Celular Viral , Infecções por Vírus Epstein-Barr/complicações , Infecções por Vírus Epstein-Barr/virologia , Herpesvirus Humano 4/fisiologia , Suscetibilidade a Doenças , Infecções por Vírus Epstein-Barr/imunologia , Regulação Viral da Expressão Gênica , Genoma Viral , Interações Hospedeiro-Patógeno/genética , Humanos , Deleção de Sequência , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Subpopulações de Linfócitos T/virologia
10.
Cancer Sci ; 111(1): 279-287, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31743514

RESUMO

Epstein-Barr virus (EBV) is a well-established tumor virus that has been implicated in a wide range of immunodeficiency-associated lymphoproliferative disorders (LPDs). Although rituximab, a CD20 mAb, has proven effective against EBV-associated LPDs, prolonged use of this drug could lead to resistance due to the selective expansion of CD20- cells. We have previously shown that cyclin-dependent kinase (CDK) inhibitors are able to specifically suppress the expression of viral late genes, particularly those encoding structural proteins; however, the therapeutic effect of CDK inhibitors against EBV-associated LPDs is not clear. In this study, we examined whether CDK inhibitors confer a therapeutic effect against LPDs in vivo. Treatment with alsterpaullone, an inhibitor of the CDK2 complex, resulted in a survival benefit and suppressed tumor invasion in a mouse model of LPDs. Inhibition of CDK efficiently induced G1 cell cycle arrest and apoptosis in EBV-positive B cells. These results suggest that alsterpaullone suppresses cell cycle progression, resulting in the antitumor effect observed in vivo.


Assuntos
Antineoplásicos/farmacologia , Benzazepinas/farmacologia , Herpesvirus Humano 4/patogenicidade , Indóis/farmacologia , Transtornos Linfoproliferativos/tratamento farmacológico , Transtornos Linfoproliferativos/virologia , Inibidores de Proteínas Quinases/farmacologia , Animais , Apoptose/efeitos dos fármacos , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Infecções por Vírus Epstein-Barr/tratamento farmacológico , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Fase G1/efeitos dos fármacos , Células HEK293 , Humanos , Transtornos Linfoproliferativos/genética , Camundongos , Camundongos Endogâmicos NOD
11.
J Virol ; 93(8)2019 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-30700607

RESUMO

Temporally controlled gene expression is necessary for the propagation of herpesviruses. To achieve this, herpesviruses encode several transcriptional regulators. In Epstein-Barr virus, BcRF1 associates with five viral proteins (BDLF4, BGLF3, BFRF2, BVLF1, and BDLF3.5) to form the viral late (L) gene regulatory complex, which is called the viral preinitiation complex (vPIC), on TATT-containing promoters. However, regulation of the vPIC has been largely unexplored. In this study, we performed two screens using a kinase inhibitor library and identified a series of cyclin-dependent kinase (CDK) inhibitors that downregulated the expression of L genes without any impact on viral DNA replication through destabilization of the BDLF4 protein. Knockdown of CDK2 by short hairpin RNA (shRNA) and proteasome inhibitor treatment showed that phosphorylation of the BDLF4 protein prevented ubiquitin-mediated degradation. Moreover, we demonstrated that cyclin A- and E-associated CDK2 complexes phosphorylated BDLF4 in vitro, and we identified several serine/threonine phosphorylation sites in BDLF4. Phosphoinactive and phosphomimic mutants revealed that phosphorylation at threonine 91 plays a role in stabilizing BDLF4. Therefore, our findings indicate that S-like-phase CDKs mediate the regulation of L gene expression through stabilization of the BDLF4 protein, which makes the temporal L gene expression system more robust.IMPORTANCE Late (L) genes represent more than one-third of the herpesvirus genome, suggesting that many of these genes are indispensable for the life cycle of the virus. With the exception of BCRF1, BDLF2, and BDLF3, Epstein-Barr virus L genes are transcribed by viral regulators, which are known as the viral preinitiation complex (vPIC) and the host RNA polymerase II complex. Because the vPIC is conserved in beta- and gammaherpesviruses, studying the control of viral L gene expression by the vPIC contributes to the development of drugs that specifically inhibit these processes in beta- and gammaherpesvirus infections/diseases. In this study, we demonstrated that CDK inhibitors induced destabilization of the vPIC component BDLF4, leading to a reduction in L gene expression and subsequent progeny production. Our findings suggest that CDK inhibitors may be a therapeutic option against beta- and gammaherpesviruses in combination with existing inhibitors of herpesvirus lytic replication, such as ganciclovir.


Assuntos
Quinase 2 Dependente de Ciclina/metabolismo , Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/metabolismo , Proteólise , Transcrição Gênica , Proteínas Virais/metabolismo , Quinase 2 Dependente de Ciclina/antagonistas & inibidores , Quinase 2 Dependente de Ciclina/genética , Técnicas de Silenciamento de Genes , Células HEK293 , Herpesvirus Humano 4/genética , Humanos , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Inibidores de Proteassoma/farmacologia , Estabilidade Proteica , Ubiquitina/genética , Ubiquitina/metabolismo , Proteínas Virais/genética
12.
J Appl Clin Med Phys ; 21(10): 109-121, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32888359

RESUMO

PURPOSE: To evaluate the optimal period of replanning to spare the rectal dose by investigating daily rectal movements during computed tomography (CT) image-guided proton therapy for prostate cancer. MATERIALS AND METHODS: To evaluate the optimum reference period for replanning, we analyzed 1483 sets of daily CT (dCT) images acquired from 40 prostate cancer patients and measured the daily rectal movement along the anterior-posterior direction based on the simulator CT (sCT) images and dCT images. We calculated daily dose distributions based on initial plans on the sCT images and replans on the dCT images for 13 representative patients, and evaluated daily dose volume histograms (DVHs) for the prostate, seminal vesicles, and rectum. RESULTS: The rectal anterior side on the dCT images around the seminal vesicles largely deviated toward the anterior side relative to the position on the reference sCT images, but the deviation decreased by referring to the dCT images and became nearly zero when we referred to the dCT images after 10-day treatment. The daily DVH values for the prostate showed good dose coverage. For six patients showing rectal movement toward the anterior side, the daily rectal DVH (V77% ) showed a 3.0 ± 1.7 cc excess from the initial plan and this excess was correlated with 9.9 ± 6.8 mm rectal movement. To identify the patients (37.5% in total) for whom the replanning on the 10th-day and 20th-day CT images reduced the V77% excess to 0.4 ± 1.5 cc and -0.2 ± 1.3 cc, respectively, we evaluated the accumulated mean doses with a 1.2 cc criterion. CONCLUSION: Our data demonstrate that the daily movement of the rectal anterior side tends to move toward the anterior side, which results in a rectal overdose, and the mean of the movement gradually decreases with the passage of days. In such cases, replanning with the reference CT after 10 days is effective to spare the rectal dose.


Assuntos
Neoplasias da Próstata , Prótons , Humanos , Masculino , Movimento , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/radioterapia , Dosagem Radioterapêutica , Planejamento da Radioterapia Assistida por Computador , Reto/diagnóstico por imagem , Tomografia Computadorizada por Raios X
13.
Uirusu ; 70(1): 83-90, 2020.
Artigo em Japonês | MEDLINE | ID: mdl-33967117

RESUMO

Productive (lytic) replication of DNA viruses elicits host cell DNA damage responses, which cause both beneficial and detrimental effects on viral replication. Viruses utilize them and selectively cancel the 'noisy' downstream signaling pathways, leading to maintain high S-phase CDK activities required for viral replication. To achieve this fine tuning of cellular environment, herpesviruses encode many (>70) genes in their genome, which are expressed in a strictly regulated temporal cascade (immediate-early, early, and late). Here, I introduce and discuss how Epstein-Barr virus, an oncogenic herpesvirus, hijacks the cellular environment and adapt it for the progeny production.


Assuntos
Infecções por Vírus Epstein-Barr , Herpesvirus Humano 4 , Replicação do DNA , Herpesvirus Humano 4/genética , Humanos , Fosforilação , Transdução de Sinais , Replicação Viral/genética
14.
Gan To Kagaku Ryoho ; 46(13): 1975-1977, 2019 Dec.
Artigo em Japonês | MEDLINE | ID: mdl-32157031

RESUMO

A 67-year-old man visited our hospital due to progressing appetite loss and fever. He presented with a fist-sized palpable mass in his right hypochondrium. Abdominal CT showed a 10 cm diameter tumor that originated from the gall bladder infiltrating the abdominal wall, liver, duodenum, and colon. Blood tests revealed leukocytosis, elevated C-reactive protein level, and severe malnutrition. FDG-PET showed markedly high uptake in the tumor and diffuse uptake in the spine. Owing to the inability of oral intake, he underwent laparoscopic gastrojejunostomy and intraoperative tumor biopsy, which demonstrated pathologically G-CSF-producing carcinoma in the gall bladder. For the rapidly progressive tumor, he underwent proton beam chemoradiotherapy as preoperative treatment. The tumor markedly shrunk with dramatic improvement of his inflammatory and nutritional status. Consequently, R0 resection could be performed by combination surgeries of right hemi-colectomy, pancreatoduodenectomy, and partial liver resection. He received adjuvant chemotherapy and was alive without recurrence 12 months after tumor resection. To our knowledge, this is the first report of the use of neoadjuvant proton beam chemoradiotherapy in biliary cancer.


Assuntos
Quimiorradioterapia , Neoplasias da Vesícula Biliar , Terapia Neoadjuvante , Idoso , Neoplasias da Vesícula Biliar/terapia , Humanos , Masculino , Recidiva Local de Neoplasia , Prótons
15.
J Virol ; 91(23)2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-28904200

RESUMO

Epstein-Barr virus (EBV), a member of human gammaherpesvirus, infects mainly B cells. EBV has two alternative life cycles, latent and lytic, and is reactivated occasionally from the latent stage to the lytic cycle. To combat EBV-associated disorders, understanding the molecular mechanisms of the EBV lytic replication cycle is also important. Here, we focused on an EBV lytic gene, BKRF4. Using our anti-BKRF4 antibody, we revealed that the BKRF4 gene product is expressed during the lytic cycle with late kinetics. To characterize the role of BKRF4, we constructed BKRF4-knockout mutants using the bacterial artificial chromosome (BAC) and CRISPR/Cas9 systems. Although disruption of the BKRF4 gene had almost no effect on viral protein expression and DNA synthesis, it significantly decreased progeny virion levels in HEK293 and Akata cells. Furthermore, we show that BKRF4 is involved not only in production of progeny virions but also in increasing the infectivity of the virus particles. Immunoprecipitation assays revealed that BKRF4 interacted with a virion protein, BGLF2. We showed that the C-terminal region of BKRF4 was critical for this interaction and for efficient progeny production. Immunofluorescence analysis revealed that BKRF4 partially colocalized with BGLF2 in the nucleus and perinuclear region. Finally, we showed that BKRF4 is a phosphorylated, possible tegument protein and that the EBV protein kinase BGLF4 may be important for this phosphorylation. Taken together, our data suggest that BKRF4 is involved in the production of infectious virions.IMPORTANCE Although the latent genes of EBV have been studied extensively, the lytic genes are less well characterized. This study focused on one such lytic gene, BKRF4, which is conserved only among gammaherpesviruses (ORF45 of Kaposi's sarcoma-associated herpesvirus or murine herpesvirus 68). After preparing the BKRF4 knockout virus using B95-8 EBV-BAC, we demonstrated that the BKRF4 gene was involved in infectious progeny particle production. Importantly, we successfully generated a BKRF4 knockout virus of Akata using CRISPR/Cas9 technology, confirming the phenotype in this separate strain. We further showed that BKRF4 interacted with another virion protein, BGLF2, and demonstrated the importance of this interaction in infectious virion production. These results shed light on the elusive process of EBV progeny maturation in the lytic cycle. Notably, this study describes a successful example of the generation and characterization of an EBV construct with a disrupted lytic gene using CRISPR/Cas9 technology.


Assuntos
Replicação do DNA , Herpesvirus Humano 4/fisiologia , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Virais/genética , Proteínas Virais/metabolismo , Replicação Viral , Proteínas Associadas a CRISPR/genética , Cromossomos Artificiais Bacterianos , Técnicas de Inativação de Genes , Células HEK293 , Herpesvirus Humano 4/genética , Humanos , Cinética , Mutação , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Virais de Fusão/metabolismo , Proteínas Virais/química , Montagem de Vírus
16.
Nature ; 490(7421): 547-51, 2012 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-23023132

RESUMO

Mitochondrial respiratory function is frequently impaired in human cancers. However, the mechanisms by which mitochondrial dysfunction contributes to tumour progression remain elusive. Here we show in Drosophila imaginal epithelium that defects in mitochondrial function potently induce tumour progression of surrounding tissue in conjunction with oncogenic Ras. Our data show that Ras activation and mitochondrial dysfunction cooperatively stimulate production of reactive oxygen species, which causes activation of c-Jun amino (N)-terminal kinase (JNK) signalling. JNK cooperates with oncogenic Ras to inactivate the Hippo pathway, leading to upregulation of its targets Unpaired (an interleukin-6 homologue) and Wingless (a Wnt homologue). Mitochondrial dysfunction in Ras-activated cells further cooperates with Ras signalling in neighbouring cells with normal mitochondrial function, causing benign tumours to exhibit metastatic behaviour. Our findings provide a mechanistic basis for interclonal tumour progression driven by mitochondrial dysfunction and oncogenic Ras.


Assuntos
Progressão da Doença , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Mitocôndrias/patologia , Neoplasias/patologia , Proteínas Serina-Treonina Quinases/metabolismo , Transdução de Sinais , Animais , Transformação Celular Neoplásica , Células Clonais/metabolismo , Células Clonais/patologia , Olho Composto de Artrópodes/crescimento & desenvolvimento , Olho Composto de Artrópodes/patologia , Olho Composto de Artrópodes/ultraestrutura , Modelos Animais de Doenças , Drosophila melanogaster/enzimologia , Drosophila melanogaster/genética , Discos Imaginais/metabolismo , Discos Imaginais/patologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mitocôndrias/metabolismo , Neoplasias/metabolismo , Proteína Oncogênica p21(ras)/genética , Proteína Oncogênica p21(ras)/metabolismo , Estresse Oxidativo , Espécies Reativas de Oxigênio/metabolismo , Fatores de Transcrição/metabolismo , Regulação para Cima , Proteína Wnt1/metabolismo
17.
J Virol ; 90(8): 3873-3889, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26819314

RESUMO

UNLABELLED: Latent membrane protein 1 (LMP1) is a major oncogene essential for primary B cell transformation by Epstein-Barr virus (EBV). Previous studies suggested that some transcription factors, such as PU.1, RBP-Jκ, NF-κB, and STAT, are involved in this expression, but the underlying mechanism is unclear. Here, we identified binding sites for PAX5, AP-2, and EBF in the proximal LMP1 promoter (ED-L1p). We first confirmed the significance of PU.1 and POU domain transcription factor binding for activation of the promoter in latency III. We then focused on the transcription factors AP-2 and early B cell factor (EBF). Interestingly, among the three AP-2-binding sites in the LMP1 promoter, two motifs were also bound by EBF. Overexpression, knockdown, and mutagenesis in the context of the viral genome indicated that AP-2 plays an important role in LMP1 expression in latency II in epithelial cells. In latency III B cells, on the other hand, the B cell-specific transcription factor EBF binds to the ED-L1p and activates LMP1 transcription from the promoter. IMPORTANCE: Epstein-Barr virus (EBV) latent membrane protein 1 (LMP1) is crucial for B cell transformation and oncogenesis of other EBV-related malignancies, such as nasopharyngeal carcinoma and T/NK lymphoma. Its expression is largely dependent on the cell type or condition, and some transcription factors have been implicated in its regulation. However, these previous reports evaluated the significance of specific factors mostly by reporter assay. In this study, we prepared point-mutated EBV at the binding sites of such transcription factors and confirmed the importance of AP-2, EBF, PU.1, and POU domain factors. Our results will provide insight into the transcriptional regulation of the major oncogene LMP1.


Assuntos
Regulação Viral da Expressão Gênica , Herpesvirus Humano 4/genética , Transativadores/metabolismo , Fator de Transcrição AP-2/metabolismo , Proteínas da Matriz Viral/genética , Sítios de Ligação , Transformação Celular Viral , Células HEK293 , Células HeLa , Humanos , Mutação , Oncogenes , Fatores do Domínio POU/metabolismo , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas/metabolismo
19.
J Appl Clin Med Phys ; 18(4): 155-160, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28594079

RESUMO

PURPOSE/OBJECTIVE(S): Accurate and reproducible positioning of the breast is difficult due to its deformability and softness; thus, targeting a breast tumor or tumor bed with fractionated radiotherapy using external beam radiation is difficult. The aim of this study was to develop a novel bra to aid in breast immobilization in the prone position. MATERIALS & METHODS: To assess the accuracy of prone position fixation of breast tumors, 33 breast cancer patients with 34 lesions were recruited. The bra used in this verification was customized from a commercially available bra. Duplicate MRI were acquired in the prone position, alternating with and without the bra, and for each series, patients were asked to step off the MRI table and re-set up in the prone position. Patients were also asked to remove and re-fit the bra for the second MRI. Each pair of images were superimposed to match the shape of the skin surface, and the maximum difference in tumor geometric center in three axes was measured. The required set up margin was calculated as: required margin = mean difference in geometric center + 2.5 standard deviation. The volumetric overlap of the tumor, as well as contouring uncertainties, was evaluated using contour analysis software. RESULTS: The median breast size was 498 cc. The required margins for the lateral, vertical, and longitudinal directions were estimated to be 4.1, 4.1, and 5.0 mm, respectively, with the bra, and 5.1, 6.9, and 6.7 mm, respectively, without the bra. These margins covered the dislocation of more than 33 lesions in total. With the bra, 33 lesions had achieved an objective overlap of 95% and 99% with 2 and 4 mm margins, respectively, whereas 4 and 8 mm, respectively, were needed without the bra. CONCLUSION: The use of an immobilizing bra reduced the setup margin for prone position fixation of breast tumors.


Assuntos
Neoplasias da Mama/radioterapia , Mama , Vestuário , Imobilização/métodos , Posicionamento do Paciente/métodos , Decúbito Ventral , Erros de Configuração em Radioterapia/prevenção & controle , Mama/anatomia & histologia , Neoplasias da Mama/diagnóstico por imagem , Feminino , Humanos , Tamanho do Órgão , Planejamento da Radioterapia Assistida por Computador , Reprodutibilidade dos Testes
20.
J Virol ; 89(19): 10120-4, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26202235

RESUMO

Epstein-Barr virus (EBV) is a gammaherpesvirus, associated with infectious mononucleosis and various types of malignancy. We focused here on the BDLF4 gene of EBV and identified it as a lytic gene, expressed with early kinetics. Viral late gene expression of the BDLF4 knockout strain was severely restricted; this could be restored by an exogenous supply of BDLF4. These results indicate that BDLF4 is important for the EBV lytic replication cycle, especially in late gene expression.


Assuntos
Genes Virais , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/fisiologia , Regulação Viral da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Fases de Leitura Aberta , Replicação Viral/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA