Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Haematologica ; 2024 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-38841800

RESUMEN

Diffuse large B-cell lymphoma (DLBCL) is the most common malignancy that develops in patients with ataxia-telangiectasia, a cancer-predisposing inherited syndrome characterized by inactivating germline ATM mutations. ATM is also frequently mutated in sporadic DLBCL. To investigate lymphomagenic mechanisms and lymphoma-specific dependencies underlying defective ATM, we applied ribonucleic acid (RNA)-seq and genome-scale loss-offunction clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 screens to systematically interrogate B-cell lymphomas arising in a novel murine model (Atm-/-nu-/-) with constitutional Atm loss, thymic aplasia but residual T-cell populations. Atm-/-nu-/-lymphomas, which phenotypically resemble either activated B-cell-like or germinal center Bcell-like DLBCL, harbor a complex karyotype, and are characterized by MYC pathway activation. In Atm-/-nu-/-lymphomas, we discovered nucleotide biosynthesis as a MYCdependent cellular vulnerability that can be targeted through the synergistic nucleotidedepleting actions of mycophenolate mofetil (MMF) and the WEE1 inhibitor, adavosertib (AZD1775). The latter is mediated through a synthetically lethal interaction between RRM2 suppression and MYC dysregulation that results in replication stress overload in Atm-/-nu-/-lymphoma cells. Validation in cell line models of human DLBCL confirmed the broad applicability of nucleotide depletion as a therapeutic strategy for MYC-driven DLBCL independent of ATM mutation status. Our findings extend current understanding of lymphomagenic mechanisms underpinning ATM loss and highlight nucleotide metabolism as a targetable therapeutic vulnerability in MYC-driven DLBCL.

2.
Blood ; 134(7): 591-596, 2019 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-31186275

RESUMEN

Although a pathogenic role for the Epstein-Barr virus (EBV) is largely undisputed for tumors that are consistently EBV genome positive (eg, nasopharyngeal carcinoma, endemic Burkitt lymphoma), this is not the case for classical Hodgkin lymphoma (cHL), a tumor with only a variable EBV association. In light of recent developments in immunotherapeutics and small molecules targeting EBV, we believe it is now timely to reevaluate the role of EBV in cHL pathogenesis.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Herpesvirus Humano 4/fisiología , Enfermedad de Hodgkin/etiología , Enfermedad de Hodgkin/virología , Animales , Infecciones por Virus de Epstein-Barr/virología , Regulación Viral de la Expresión Génica , Genes Virales , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/aislamiento & purificación , Enfermedad de Hodgkin/patología , Enfermedad de Hodgkin/terapia , Humanos , Factores de Riesgo
3.
PLoS Biol ; 16(9): e2005046, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30180168

RESUMEN

The microenvironment of lymphoid organs can aid healthy immune function through provision of both structural and molecular support. In mice, fibroblastic reticular cells (FRCs) create an essential T-cell support structure within lymph nodes, while human FRCs are largely unstudied. Here, we show that FRCs create a regulatory checkpoint in human peripheral T-cell activation through 4 mechanisms simultaneously utilised. Human tonsil and lymph node-derived FRCs constrained the proliferation of both naïve and pre-activated T cells, skewing their differentiation away from a central memory T-cell phenotype. FRCs acted unilaterally without requiring T-cell feedback, imposing suppression via indoleamine-2,3-dioxygenase, adenosine 2A Receptor, prostaglandin E2, and transforming growth factor beta receptor (TGFßR). Each mechanistic pathway was druggable, and a cocktail of inhibitors, targeting all 4 mechanisms, entirely reversed the suppressive effect of FRCs. T cells were not permanently anergised by FRCs, and studies using chimeric antigen receptor (CAR) T cells showed that immunotherapeutic T cells retained effector functions in the presence of FRCs. Since mice were not suitable as a proof-of-concept model, we instead developed a novel human tissue-based in situ assay. Human T cells stimulated using standard methods within fresh tonsil slices did not proliferate except in the presence of inhibitors described above. Collectively, we define a 4-part molecular mechanism by which FRCs regulate the T-cell response to strongly activating events in secondary lymphoid organs while permitting activated and CAR T cells to utilise effector functions. Our results define 4 feasible strategies, used alone or in combinations, to boost primary T-cell responses to infection or cancer by pharmacologically targeting FRCs.


Asunto(s)
Diferenciación Celular/inmunología , Microambiente Celular , Ganglios Linfáticos/inmunología , Activación de Linfocitos/inmunología , Linfocitos T/citología , Adulto , Proliferación Celular , Niño , Fibroblastos/citología , Humanos , Memoria Inmunológica , Fenotipo
4.
J Pathol ; 248(2): 142-154, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30666658

RESUMEN

The Epstein-Barr virus (EBV) is found almost exclusively in the activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL), yet its contribution to this tumour remains poorly understood. We have focused on the EBV-encoded latent membrane protein-1 (LMP1), a constitutively activated CD40 homologue expressed in almost all EBV-positive DLBCLs and which can disrupt germinal centre (GC) formation and drive lymphomagenesis in mice. Comparison of the transcriptional changes that follow LMP1 expression with those that follow transient CD40 signalling in human GC B cells enabled us to define pathogenic targets of LMP1 aberrantly expressed in ABC-DLBCL. These included the down-regulation of S1PR2, a sphingosine-1-phosphate (S1P) receptor that is transcriptionally down-regulated in ABC-DLBCL, and when genetically ablated leads to DLBCL in mice. Consistent with this, we found that LMP1-expressing primary ABC-DLBCLs were significantly more likely to lack S1PR2 expression than were LMP1-negative tumours. Furthermore, we showed that the down-regulation of S1PR2 by LMP1 drives a signalling loop leading to constitutive activation of the phosphatidylinositol-3-kinase (PI3-K) pathway. Finally, core LMP1-PI3-K targets were enriched for lymphoma-related transcription factors and genes associated with shorter overall survival in patients with ABC-DLBCL. Our data identify a novel function for LMP1 in aggressive DLBCL. Copyright © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/metabolismo , Linfoma de Células B Grandes Difuso/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Proteínas de la Matriz Viral/metabolismo , Antígenos CD40/genética , Antígenos CD40/metabolismo , Línea Celular Tumoral , Transformación Celular Viral , Bases de Datos Genéticas , Infecciones por Virus de Epstein-Barr/mortalidad , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 4/genética , Interacciones Huésped-Patógeno , Humanos , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/mortalidad , Linfoma de Células B Grandes Difuso/virología , Fosfatidilinositol 3-Quinasa/metabolismo , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Receptores de Esfingosina-1-Fosfato/genética , Proteínas de la Matriz Viral/genética
5.
J Pathol ; 242(1): 62-72, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28240350

RESUMEN

Undifferentiated nasopharyngeal carcinoma (NPC) is a cancer with high metastatic potential that is consistently associated with Epstein-Barr virus (EBV) infection. In this study, we have investigated the functional contribution of sphingosine-1-phosphate (S1P) signalling to the pathogenesis of NPC. We show that EBV infection or ectopic expression of the EBV-encoded latent genes (EBNA1, LMP1, and LMP2A) can up-regulate sphingosine kinase 1 (SPHK1), the key enzyme that produces S1P, in NPC cell lines. Exogenous addition of S1P promotes the migration of NPC cells through the activation of AKT; shRNA knockdown of SPHK1 resulted in a reduction in the levels of activated AKT and inhibition of cell migration. We also show that S1P receptor 3 (S1PR3) mRNA is overexpressed in EBV-positive NPC patient-derived xenografts and a subset of primary NPC tissues, and that knockdown of S1PR3 suppressed the activation of AKT and the S1P-induced migration of NPC cells. Taken together, our data point to a central role for EBV in mediating the oncogenic effects of S1P in NPC and identify S1P signalling as a potential therapeutic target in this disease. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Asunto(s)
Infecciones por Virus de Epstein-Barr/complicaciones , Lisofosfolípidos/fisiología , Neoplasias Nasofaríngeas/virología , Proteína Oncogénica v-akt/metabolismo , Receptores de Lisoesfingolípidos/metabolismo , Esfingosina/análogos & derivados , Adulto , Anciano , Animales , Carcinoma , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/metabolismo , Infecciones por Virus de Epstein-Barr/patología , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Técnicas de Silenciamiento del Gen , Xenoinjertos , Humanos , Lisofosfolípidos/farmacología , Masculino , Persona de Mediana Edad , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/patología , ARN Mensajero/genética , Receptores de Lisoesfingolípidos/genética , Receptores de Lisoesfingolípidos/fisiología , Transducción de Señal/fisiología , Esfingosina/farmacología , Esfingosina/fisiología , Receptores de Esfingosina-1-Fosfato , Regulación hacia Arriba
6.
J Virol ; 89(10): 5222-37, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25787276

RESUMEN

UNLABELLED: Epstein-Barr virus (EBV) infects most of the world's population and is causally associated with several human cancers, but little is known about how EBV genetic variation might influence infection or EBV-associated disease. There are currently no published wild-type EBV genome sequences from a healthy individual and very few genomes from EBV-associated diseases. We have sequenced 71 geographically distinct EBV strains from cell lines, multiple types of primary tumor, and blood samples and the first EBV genome from the saliva of a healthy carrier. We show that the established genome map of EBV accurately represents all strains sequenced, but novel deletions are present in a few isolates. We have increased the number of type 2 EBV genomes sequenced from one to 12 and establish that the type 1/type 2 classification is a major feature of EBV genome variation, defined almost exclusively by variation of EBNA2 and EBNA3 genes, but geographic variation is also present. Single nucleotide polymorphism (SNP) density varies substantially across all known open reading frames and is highest in latency-associated genes. Some T-cell epitope sequences in EBNA3 genes show extensive variation across strains, and we identify codons under positive selection, both important considerations for the development of vaccines and T-cell therapy. We also provide new evidence for recombination between strains, which provides a further mechanism for the generation of diversity. Our results provide the first global view of EBV sequence variation and demonstrate an effective method for sequencing large numbers of genomes to further understand the genetics of EBV infection. IMPORTANCE: Most people in the world are infected by Epstein-Barr virus (EBV), and it causes several human diseases, which occur at very different rates in different parts of the world and are linked to host immune system variation. Natural variation in EBV DNA sequence may be important for normal infection and for causing disease. Here we used rapid, cost-effective sequencing to determine 71 new EBV sequences from different sample types and locations worldwide. We showed geographic variation in EBV genomes and identified the most variable parts of the genome. We identified protein sequences that seem to have been selected by the host immune system and detected variability in known immune epitopes. This gives the first overview of EBV genome variation, important for designing vaccines and immune therapy for EBV, and provides techniques to investigate relationships between viral sequence variation and EBV-associated diseases.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Variación Genética , Genoma Viral , Herpesvirus Humano 4/genética , Secuencia de Aminoácidos , Antígenos Virales/genética , Portador Sano/virología , Línea Celular Tumoral , ADN Viral/genética , Epítopos de Linfocito T/genética , Antígenos Nucleares del Virus de Epstein-Barr/genética , Herpesvirus Humano 4/clasificación , Herpesvirus Humano 4/aislamiento & purificación , Humanos , Filogenia , Polimorfismo de Nucleótido Simple , Recombinación Genética , Proteínas de la Matriz Viral/genética
7.
J Pathol ; 235(2): 312-22, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25294567

RESUMEN

Since the discovery in 1964 of the Epstein-Barr virus (EBV) in African Burkitt lymphoma, this virus has been associated with a remarkably diverse range of cancer types. Because EBV persists in the B cells of the asymptomatic host, it can easily be envisaged how it contributes to the development of B-cell lymphomas. However, EBV is also found in other cancers, including T-cell/natural killer cell lymphomas and several epithelial malignancies. Explaining the aetiological role of EBV is challenging, partly because the virus probably contributes differently to each tumour and partly because the available disease models cannot adequately recapitulate the subtle variations in the virus-host balance that exist between the different EBV-associated cancers. A further challenge is to identify the co-factors involved; because most persistently infected individuals will never develop an EBV-associated cancer, the virus cannot be working alone. This article will review what is known about the contribution of EBV to lymphoma development.


Asunto(s)
Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/patogenicidad , Linfoma/virología , Animales , Biopsia , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/inmunología , Infecciones por Virus de Epstein-Barr/patología , Herpesvirus Humano 4/inmunología , Interacciones Huésped-Patógeno , Humanos , Linfoma/inmunología , Linfoma/patología , Patología Molecular/métodos , Valor Predictivo de las Pruebas , Pronóstico , Factores de Riesgo , Virología/métodos , Virulencia
8.
J Pathol ; 235(3): 456-65, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25294670

RESUMEN

Undifferentiated nasopharyngeal carcinoma (NPC) is a highly metastatic disease that is consistently associated with Epstein-Barr virus (EBV) infection. In this study, we have investigated the contribution of lysophosphatidic acid (LPA) signalling to the pathogenesis of NPC. Here we demonstrate two distinct functional roles for LPA in NPC. First, we show that LPA enhances the migration of NPC cells and second, that it can inhibit the activity of EBV-specific cytotoxic T cells. Focusing on the first of these phenotypes, we show that one of the LPA receptors, LPA receptor 5 (LPAR5), is down-regulated in primary NPC tissues and that this down-regulation promotes the LPA-induced migration of NPC cell lines. Furthermore, we found that EBV infection or ectopic expression of the EBV-encoded LMP2A was sufficient to down-regulate LPAR5 in NPC cell lines. Our data point to a central role for EBV in mediating the oncogenic effects of LPA in NPC and identify LPA signalling as a potential therapeutic target in this disease.


Asunto(s)
Regulación hacia Abajo/fisiología , Infecciones por Virus de Epstein-Barr/fisiopatología , Regulación Neoplásica de la Expresión Génica/fisiología , Lisofosfolípidos/fisiología , Neoplasias Nasofaríngeas/fisiopatología , Receptores del Ácido Lisofosfatídico/fisiología , Transducción de Señal/fisiología , Adenocarcinoma/patología , Adenocarcinoma/fisiopatología , Carcinoma , Línea Celular Tumoral , Movimiento Celular/fisiología , Herpesvirus Humano 4/fisiología , Humanos , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/patología , Hidrolasas Diéster Fosfóricas/fisiología , Receptores del Ácido Lisofosfatídico/genética , Linfocitos T Citotóxicos/patología , Proteínas de la Matriz Viral/fisiología
9.
J Cell Mol Med ; 19(10): 2329-40, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26171944

RESUMEN

Almost all drugs approved for use in humans possess potentially beneficial 'off-target' effects in addition to their principal activity. In some cases this has allowed for the relatively rapid repurposing of drugs for other indications. In this review we focus on the potential for re-purposing FTY720 (also known as fingolimod, Gilenya(™)), an immunomodulatory drug recently approved for the treatment of multiple sclerosis (MS). The therapeutic benefit of FTY720 in MS is largely attributed to the immunosuppressive effects that result from its modulation of sphingosine 1-phosphate receptor signalling. However, this drug has also been shown to inhibit other cancer-associated signal transduction pathways in part because of its structural similarity to sphingosine, and consequently shows efficacy as an anti-cancer agent both in vitro and in vivo. Here, we review the effects of FTY720 on signal transduction pathways and cancer-related cellular processes, and discuss its potential use as an anti-cancer drug.


Asunto(s)
Antineoplásicos/farmacología , Reposicionamiento de Medicamentos , Clorhidrato de Fingolimod/farmacología , Sistemas de Liberación de Medicamentos , Humanos , Fenotipo , Transducción de Señal/efectos de los fármacos
10.
Blood ; 122(26): 4237-45, 2013 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-24136166

RESUMEN

The malignant Hodgkin and Reed-Sternberg (HRS) cells of Hodgkin lymphoma are surrounded by a tumor microenvironment that is composed of a variety of cell types, as well as noncellular components such as collagen. Although HRS cells harbor oncogenic Epstein-Barr virus (EBV) in approximately 50% of cases, it is not known if the tumor microenvironment contributes to EBV-driven lymphomagenesis. We show that expression of the EBV-encoded latent membrane protein-1 (LMP1) in primary human germinal center B cells, the presumed progenitors of HRS cells, upregulates discoidin domain receptor 1 (DDR1), a receptor tyrosine kinase activated by collagen. We also show that HRS cells intimately associated with collagen frequently overexpress DDR1 and that short-term exposure to collagen is sufficient to activate DDR1 in Hodgkin lymphoma-derived cell lines. The ectopic expression of DDR1 significantly increased the survival of collagen-treated DG75 Burkitt lymphoma cells, following etoposide treatment. Conversely, knockdown of DDR1 significantly decreased the survival of collagen-treated L428 Hodgkin lymphoma cells in the absence of specific apoptotic stimulus, suggesting that DDR1 also influences baseline survival. Our results identify a hitherto unknown function for collagen in protecting Hodgkin lymphoma cells from apoptosis and suggest an important contribution of the tumor microenvironment in promoting the oncogenic effects of EBV.


Asunto(s)
Linfocitos B/citología , Infecciones por Virus de Epstein-Barr/genética , Herpesvirus Humano 4/genética , Enfermedad de Hodgkin/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas de la Matriz Viral/genética , Linfocitos B/fisiología , Linfoma de Burkitt/genética , Linfoma de Burkitt/patología , Muerte Celular/fisiología , Células Cultivadas , Colágeno/metabolismo , Receptor con Dominio Discoidina 1 , Infecciones por Virus de Epstein-Barr/patología , Regulación Viral de la Expresión Génica , Técnicas de Silenciamiento del Gen , Centro Germinal/citología , Enfermedad de Hodgkin/patología , Humanos , Fosforilación/fisiología , Proteínas Tirosina Quinasas Receptoras/genética , Células de Reed-Sternberg/citología , Células de Reed-Sternberg/fisiología , Microambiente Tumoral/fisiología
11.
J Pathol ; 230(4): 399-409, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23592216

RESUMEN

Hodgkin's lymphoma is unusual among B cell lymphomas, in so far as the malignant Hodgkin/Reed-Sternberg (HRS) cells lack a functional B cell receptor (BCR), as well as many of the required downstream signalling components. In Epstein-Barr virus (EBV)-positive cases of Hodgkin's lymphoma, HRS cells express the viral latent membrane proteins (LMP)-1 and -2A. LMP2A is thought to contribute to the pathogenesis of Hodgkin's lymphoma by providing a surrogate BCR-like survival signal. However, LMP2A has also been shown to induce the virus-replicative cycle in B cells, an event presumably incompatible with lymphomagenesis. In an attempt to resolve this apparent paradox, we compared the transcriptional changes observed in primary HRS cells with those induced by LMP2A and by BCR activation in primary human germinal centre (GC) B cells, the presumed progenitors of HRS cells. We found a subset of genes that were up-regulated by both LMP2A expression and BCR activation but which were down-regulated in primary HRS cells. These genes included EGR1, an immediate-early gene that is required for BCR-induced entry to the virus-replicative cycle. We present data supporting a model for the pathogenesis of EBV-positive Hodgkin's lymphoma in which LMP2A-expressing HRS cells lacking BCR signalling functions cannot induce EGR1 and are consequently protected from entry to the virus lytic cycle. The primary microarray data are available from GEO (http://www.ncbi.nlm.nih.gov/geo/) under series Accession No 46143.


Asunto(s)
Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Herpesvirus Humano 4/metabolismo , Enfermedad de Hodgkin/metabolismo , Células de Reed-Sternberg/metabolismo , Proteínas de la Matriz Viral/metabolismo , Linfocitos B/metabolismo , Linfoma de Burkitt/metabolismo , Linfoma de Burkitt/patología , Linfoma de Burkitt/virología , Línea Celular Tumoral , Efecto Citopatogénico Viral , Proteína 1 de la Respuesta de Crecimiento Precoz/genética , Activación Enzimática , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Herpesvirus Humano 4/genética , Herpesvirus Humano 4/patogenicidad , Enfermedad de Hodgkin/genética , Enfermedad de Hodgkin/patología , Enfermedad de Hodgkin/virología , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de Antígenos de Linfocitos B/metabolismo , Células de Reed-Sternberg/patología , Células de Reed-Sternberg/virología , Transactivadores/metabolismo , Transfección , Regulación hacia Arriba , Proteínas de la Matriz Viral/genética
12.
Cancers (Basel) ; 16(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38339325

RESUMEN

BACKGROUND: A total of 30-40% of diffuse large B cell lymphoma (DLBCL) patients will either not respond to the standard therapy or their disease will recur. The first-line treatment for DLBCL is rituximab and combination chemotherapy. This treatment involves the chemotherapy-induced recruitment of tumor-associated macrophages that recognize and kill rituximab-opsonized DLBCL cells. However, we lack insights into the factors responsible for the recruitment and functionality of macrophages in DLBCL tumors. METHODS: We have studied the effects of the immunomodulatory lipid sphingosine-1-phosphate (S1P) on macrophage activity in DLBCL, both in vitro and in animal models. RESULTS: We show that tumor-derived S1P mediates the chemoattraction of both monocytes and macrophages in vitro and in animal models, an effect that is dependent upon the S1P receptor S1PR1. However, S1P inhibited M1 macrophage-mediated phagocytosis of DLBCL tumor cells opsonized with the CD20 monoclonal antibodies rituximab and ofatumumab, an effect that could be reversed by an S1PR1 inhibitor. CONCLUSIONS: Our data show that S1P signaling can modulate macrophage recruitment and tumor cell killing by anti-CD20 monoclonal antibodies in DLBCL. The administration of S1PR1 inhibitors could enhance the phagocytosis of tumor cells and improve outcomes for patients.

13.
Blood Adv ; 2024 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-38861355

RESUMEN

Burkitt lymphoma (BL) is characterized by tumor microenvironment (TME) in which macrophages represent the main component, determining a distinct histological appearance known as "starry sky" pattern. However, in some instances, BL may exhibit a granulomatous reaction that has been previously linked to a favorable prognosis and spontaneous regression. The aim of our study was to deeply characterize the immune landscape of 7 cases of EBV + BL with granulomatous reaction compared to 8 cases of EBV + BL and 8 EBV- BL, both with typical "starry sky" pattern, by Gene expression profiling performed on the NanoString nCounter platform. Subsequently, the data were validated by multiplex and combined immunostaining. Based on unsupervised clustering of differentially expressed genes, BL samples formed 3 distinct clusters differentially enriched in BL with a diffuse granulomatous reaction (cluster 1), EBV+ BL with typical starry sky pattern (Cluster 2), EBV - BL with typical "starry sky" (cluster 3). We observed variations in the immune response signature among BL with granulomatous reaction and BL with typical "starry sky", both EBV + and EBV -. The TME signature in BL with diffuse granulomatous reaction showed a proinflammatory response, while BLs with "starry sky" were characterized by up-regulation of M2- polarization and pro-tumor response. Moreover, the analysis of additional signatures revealed an up-regulation of Dark zone-signature and epigenetic-signature in BL with typical "starry sky". Tumor associated macrophages (TAM) and epigenetic regulators may be promising targets for additional therapies in BL lymphoma opening novel immunotherapeutic strategies.

14.
Int J Cancer ; 133(2): 446-54, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23292999

RESUMEN

Smoking is a strong risk factor of bladder cancer (BC), but it is currently unclear whether smoking is also associated with severity of BC at diagnosis. We performed a large hospital-based case-comparison study, examining the relation between smoking and clinical characteristics of BC at diagnosis. A total of 1,544 cases from participating hospitals in the West Midlands were recruited between 19 December 2005 and 21 April 2011. Eligible cases were adult BC patients without a previous history of this disease. At time of diagnosis, semi-structured face-to-face interviews were conducted by trained research nurses to collect smoking information. Clinical characteristics were obtained from medical records. Linear mixed models were performed to calculate predicted means in clinical outcomes for a variety of smoking behaviors. A p < 0.05 was considered statistically significant. After adjustment for age and gender, current smokers were on average 4.0 years younger at diagnosis (95% CI: -5.9 to -2.0), had larger tumors (mean difference: 0.48 cm, 95% CI: 0.04-0.91), a higher T stage (mean difference: 0.25, 95% CI: 0.08-0.41), and a borderline significantly higher grade than never smokers (mean difference: 0.15, 95% CI: 0.00-0.30). Our results suggest that smoking could be associated with a more malignant phenotype of BC at diagnosis. More research is needed on the relation between smoking and prognosis, but our results could strengthen the message about the potential risks of smoking to these patients.


Asunto(s)
Fumar/efectos adversos , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/patología , Edad de Inicio , Anciano , Estudios de Casos y Controles , Femenino , Humanos , Modelos Lineales , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Estadificación de Neoplasias , Fenotipo , Pronóstico , Factores de Riesgo , Reino Unido/epidemiología , Neoplasias de la Vejiga Urinaria/epidemiología
15.
Blood ; 117(22): 5907-17, 2011 Jun 02.
Artículo en Inglés | MEDLINE | ID: mdl-21411757

RESUMEN

An important pathogenic event in Epstein-Barr virus (EBV)-associated lymphomas is the suppression of virus replication, which would otherwise lead to cell death. Because virus replication in B cells is intimately linked to their differentiation toward plasma cells, we asked whether the physiologic signals that drive normal B-cell differentiation are absent in EBV-transformed cells. We focused on BLIMP1α, a transcription factor that is required for plasma cell differentiation and that is inactivated in diffuse large B-cell lymphomas. We show that BLIMP1α expression is down-regulated after EBV infection of primary germinal center B cells and that the EBV oncogene, latent membrane protein-1 (LMP-1), is alone capable of inducing this down-regulation in these cells. Furthermore, the down-regulation of BLIMP1α by LMP-1 was accompanied by a partial disruption of the BLIMP1α transcriptional program, including the aberrant induction of MYC, the repression of which is required for terminal differentiation. Finally, we show that the ectopic expression of BLIMP1α in EBV-transformed cells can induce the viral lytic cycle. Our results suggest that LMP-1 expression in progenitor germinal center B cells could contribute to the pathogenesis of EBV-associated lymphomas by down-regulating BLIMP1α, in turn preventing plasma cell differentiation and induction of the viral lytic cycle.


Asunto(s)
Linfocitos B/virología , Diferenciación Celular , Herpesvirus Humano 4/fisiología , Linfoma de Células B/etiología , Células Plasmáticas/patología , Proteínas Represoras/metabolismo , Proteínas de la Matriz Viral/metabolismo , Linfocitos B/metabolismo , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Western Blotting , Transformación Celular Viral , Células Cultivadas , Niño , Infecciones por Virus de Epstein-Barr/complicaciones , Infecciones por Virus de Epstein-Barr/genética , Infecciones por Virus de Epstein-Barr/virología , Perfilación de la Expresión Génica , Centro Germinal , Humanos , Técnicas para Inmunoenzimas , Linfoma de Células B/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Tonsila Palatina/citología , Tonsila Palatina/metabolismo , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , ARN Mensajero/genética , Proteínas Represoras/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas de la Matriz Viral/genética , Latencia del Virus , Replicación Viral
16.
Life (Basel) ; 13(2)2023 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-36836878

RESUMEN

Epstein-Barr virus (EBV), defined as a group I carcinogen by the World Health Organization (WHO), is present in the tumour cells of patients with different forms of B-cell lymphoma, including Burkitt lymphoma, Hodgkin lymphoma, post-transplant lymphoproliferative disorders, and, most recently, diffuse large B-cell lymphoma (DLBCL). Understanding how EBV contributes to the development of these different types of B-cell lymphoma has not only provided fundamental insights into the underlying mechanisms of viral oncogenesis, but has also highlighted potential new therapeutic opportunities. In this review, we describe the effects of EBV infection in normal B-cells and we address the germinal centre model of infection and how this can lead to lymphoma in some instances. We then explore the recent reclassification of EBV+ DLBCL as an established entity in the WHO fifth edition and ICC 2022 classifications, emphasising the unique nature of this entity. To that end, we also explore the unique genetic background of this entity and briefly discuss the potential role of the tumour microenvironment in lymphomagenesis and disease progression. Despite the recent progress in elucidating the mechanisms of this malignancy, much work remains to be done to improve patient stratification, treatment strategies, and outcomes.

17.
Int J Cancer ; 131(4): E348-61, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21913186

RESUMEN

Gene expression profiling has recently enabled the reclassification of aggressive non-Hodgkin lymphomas (aNHL) into distinct subgroups. In Burkitt lymphoma (BL) aberrant c-Myc activity results from IG-MYC translocations. However, MYC aberrations are not limited to BLs and then have a negative prognostic impact. In this study, we investigated to which extent aberrant c-Myc activity plays a functional role in other aNHL and whether it is independent from MYC translocations. Based on a combined microarray analysis of human germinal center (GC) B cells transfected with c-Myc and 220 aNHLs cases, we developed a "c-Myc index." This index measures the extent to which lymphomas express c-Myc responsive genes. It comprises genes that are affected in a variety of tumors compared to normal tissue. This supports the view that aberrant c-Myc expression in GC B cells triggers a tumor-like expression pattern. As expected, the "c-Myc index" is very high in molecular Burkitt lymphoma (mBL), but more importantly also high within other aNHL. It constitutes a negative prognostic marker independent of established risk factors and of the presence of a MYC translocation. Our data provide new insights into the role of c-Myc activity in different lymphomas and raises the question of treatment changes for those patients under risk.


Asunto(s)
Linfoma de Células B Grandes Difuso/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Secuencia de Bases , Western Blotting , Línea Celular Tumoral , Cartilla de ADN , Técnica del Anticuerpo Fluorescente , Humanos , Linfoma de Células B Grandes Difuso/patología , Pronóstico , Proteínas Proto-Oncogénicas c-myc/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
18.
J Virol ; 85(18): 9568-77, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21752916

RESUMEN

Although Epstein-Barr virus (EBV) usually establishes an asymptomatic lifelong infection, it is also implicated in the development of germinal center (GC) B-cell-derived malignancies, including Hodgkin's lymphoma (HL). Following primary infection, EBV remains latent in the memory B-cell population, where host-driven methylation of viral DNA contributes to the repression of viral gene expression. However, it is still unclear how EBV harnesses the cell's methylation machinery in B cells, how this contributes to viral persistence, and what impact this has on the methylation of cellular genes. We show that EBV infection of GC B cells is followed by upregulation of the DNA methyltransferase DNMT3A and downregulation of DNMT3B and DNMT1. We show that the EBV latent membrane protein 1 (LMP1) oncogene downregulates DNMT1 and that DNMT3A binds to the viral promoter Wp. Genome-wide promoter arrays performed with these cells showed that EBV-associated methylation changes in cellular genes were not randomly distributed across the genome but clustered at chromosomal locations, consistent with an instructive pattern of methylation, and were in part determined by promoter CpG content. Both DNMT3B and DNMT1 were downregulated and DNMT3A was upregulated in HL cell lines, recapitulating the pattern of expression observed following EBV infection of GC B cells. We also found, by using gene expression profiling, that genes differentially expressed following EBV infection of GC B cells were significantly enriched for those reported to be differentially expressed in HL. These observations suggest that EBV-infected GC B cells are a useful model for studying virus-associated changes contributing to the pathogenesis of HL.


Asunto(s)
Linfocitos B/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Centro Germinal/inmunología , Herpesvirus Humano 4/patogenicidad , Enfermedad de Hodgkin/virología , Transcripción Genética , Linfocitos B/virología , Células Cultivadas , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/biosíntesis , Metilación de ADN , ADN Metiltransferasa 3A , Perfilación de la Expresión Génica , Centro Germinal/virología , Enfermedad de Hodgkin/inmunología , Enfermedad de Hodgkin/patología , Humanos , Proteínas de la Matriz Viral/metabolismo , ADN Metiltransferasa 3B
19.
J Transl Med ; 10: 209, 2012 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-23050586

RESUMEN

BACKGROUND: Inactivaion of tumor suppressor genes (TSGs) by promoter CpG methylation frequently occurs in tumorigenesis, even in the early stages, contributing to the initiation and progression of human cancers. Deleted in lung and esophageal cancer 1 (DLEC1), located at the 3p22-21.3 TSG cluster, has been identified frequently silenced by promoter CpG methylation in multiple carcinomas, however, no study has been performed for lymphomas yet. METHODS: We examined the expression of DLEC1 by semi-quantitative reverse transcription (RT)-PCR, and evaluated the promoter methylation of DLEC1 by methylation-specific PCR (MSP) and bisulfite genomic sequencing (BGS) in common lymphoma cell lines and tumors. RESULTS: Here we report that DLEC1 is readily expressed in normal lymphoid tissues including lymph nodes and PBMCs, but reduced or silenced in 70% (16/23) of non-Hodgkin and Hodgkin lymphoma cell lines, including 2/6 diffuse large B-cell (DLBCL), 1/2 peripheral T cell lymphomas, 5/5 Burkitt, 6/7 Hodgkin and 2/3 nasal killer (NK)/T-cell lymphoma cell lines. Promoter CpG methylation was frequently detected in 80% (20/25) of lymphoma cell lines and correlated with DLEC1 downregulation/silencing. Pharmacologic demethylation reversed DLEC1 expression in lymphoma cell lines along with concomitant promoter demethylation. DLEC1 methylation was also frequently detected in 32 out of 58 (55%) different types of lymphoma tissues, but not in normal lymph nodes. Furthermore, DLEC1 was specifically methylated in the sera of 3/13 (23%) Hodgkin lymphoma patients. CONCLUSIONS: Thus, methylation-mediated silencing of DLEC1 plays an important role in multiple lymphomagenesis, and may serve as a non-invasive tumor marker for lymphoma diagnosis.


Asunto(s)
Cromosomas Humanos Par 3 , Epigénesis Genética , Silenciador del Gen , Enfermedad de Hodgkin/genética , Linfoma no Hodgkin/genética , Regiones Promotoras Genéticas , Proteínas Supresoras de Tumor/genética , Islas de CpG , Metilación de ADN , Humanos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Cancer Cell ; 1(5): 459-68, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12124175

RESUMEN

Mutations in the von Hippel-Lindau (VHL) gene are associated with hereditary and sporadic clear cell renal carcinoma. VHL acts in a ubiquitin ligase complex regulating hypoxia-inducible factor-1 (HIF-1), but the link between this function and cancer development is unclear. Here we show that in the kidneys of patients with VHL disease, HIF activation is an early event occurring in morphologically normal single cells within the renal tubules. In comparison, dysplastic lesions, cystic lesions, and tumors showed evidence of additional mechanisms that amplify HIF activation. Detection of cells with constitutive HIF activation identified a large number of previously unrecognized foci of VHL inactivation. In proximal tubules these were almost entirely unicellular, whereas multicellular foci were almost exclusively seen in the distal nephron.


Asunto(s)
Adenocarcinoma de Células Claras/metabolismo , Antígenos de Neoplasias , Carcinoma de Células Renales/metabolismo , Proteínas de Unión al ADN/metabolismo , Genes Supresores de Tumor/fisiología , Neoplasias Renales/metabolismo , Nefronas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción , Enfermedad de von Hippel-Lindau/metabolismo , Adenocarcinoma de Células Claras/genética , Adenocarcinoma de Células Claras/patología , Adulto , Antígenos CD/metabolismo , Apoptosis/fisiología , Anhidrasa Carbónica IX , Anhidrasas Carbónicas/metabolismo , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Transportador de Glucosa de Tipo 1 , Humanos , Factor 1 Inducible por Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia , Técnicas para Inmunoenzimas , Hibridación in Situ , Etiquetado Corte-Fin in Situ , Neoplasias Renales/genética , Neoplasias Renales/patología , Persona de Mediana Edad , Proteínas de Transporte de Monosacáridos/metabolismo , Proteínas de Neoplasias/metabolismo , Nefrectomía , Sondas ARN , Enfermedad de von Hippel-Lindau/genética , Enfermedad de von Hippel-Lindau/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA