Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 12129, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802399

ABSTRACT

Many targeted cancer therapies rely on biomarkers assessed by scoring of immunohistochemically (IHC)-stained tissue, which is subjective, semiquantitative, and does not account for expression heterogeneity. We describe an image analysis-based method for quantitative continuous scoring (QCS) of digital whole-slide images acquired from baseline human epidermal growth factor receptor 2 (HER2) IHC-stained breast cancer tissue. Candidate signatures for patient stratification using QCS of HER2 expression on subcellular compartments were identified, addressing the spatial distribution of tumor cells and tumor-infiltrating lymphocytes. Using data from trastuzumab deruxtecan-treated patients with HER2-positive and HER2-negative breast cancer from a phase 1 study (NCT02564900; DS8201-A-J101; N = 151), QCS-based patient stratification showed longer progression-free survival (14.8 vs 8.6 months) with higher prevalence of patient selection (76.4 vs 56.9%) and a better cross-validated log-rank p value (0.026 vs 0.26) than manual scoring based on the American Society of Clinical Oncology / College of American Pathologists guidelines. QCS-based features enriched the HER2-negative subgroup by correctly predicting 20 of 26 responders.


Subject(s)
Breast Neoplasms , Patient Selection , Receptor, ErbB-2 , Trastuzumab , Humans , Female , Receptor, ErbB-2/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Trastuzumab/therapeutic use , Middle Aged , Biomarkers, Tumor/metabolism , Adult , Immunoconjugates/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Aged , Immunohistochemistry , Camptothecin/analogs & derivatives
2.
Proc Natl Acad Sci U S A ; 118(43)2021 10 26.
Article in English | MEDLINE | ID: mdl-34686609

ABSTRACT

The Epstein-Barr virus (EBV) transforms resting B cells and is involved in the development of B cell lymphomas. We report here that the viral noncoding RNA EBER2 accelerates B cell growth by potentiating expression of the UCHL1 deubiquitinase that itself increased expression of the Aurora kinases and of cyclin B1. Importantly, this effect was also visible in Burkitt's lymphoma cells that express none of the virus's known oncogenes. Mechanistically, EBER2 bound the UCHL1 messenger RNA (mRNA), thereby bringing a protein complex that includes PU.1, a UCHL1 transactivator, to the vicinity of its promoter. Although the EBV oncogene LMP1 has been suggested to induce UCHL1, we show here that EBER2 plays a much more important role to reach significant levels of the deubiquitinase in infected cells. However, some viruses that carried a polymorphic LMP1 had an increased ability to achieve full UCHL1 expression. This work identifies a direct cellular target of a viral noncoding RNA that is likely to be central to EBV's oncogenic properties.


Subject(s)
Cell Proliferation/physiology , Deubiquitinating Enzymes/genetics , Herpesvirus 4, Human/physiology , RNA, Viral/physiology , Transcriptional Activation/physiology , B-Lymphocytes/cytology , Humans
3.
Nat Microbiol ; 4(12): 2475-2486, 2019 12.
Article in English | MEDLINE | ID: mdl-31501540

ABSTRACT

The Epstein-Barr virus M81 strain, isolated from a nasopharyngeal carcinoma, induces potent spontaneous virus production in infected B cells. We found that the M81 non-coding Epstein-Barr-encoded RNA EBER2, which carries polymorphisms that are mainly restricted to viruses found in endemic nasopharyngeal carcinomas, markedly stimulated this process. M81 EBER2 increased CXCL8 expression, and this chemokine enhanced spontaneous lytic replication levels in M81-infected B cells. Both events resulted from the endocytosis of extracellular vesicles containing EBER2 that were generated by neighbouring M81-infected B cells, thereby generating a paracrine loop. These effects were strictly dependent on a functional Toll-like receptor 7 (TLR7), a sensor of single-stranded RNA located in the endosome of these cells. These unique properties of M81 EBER2 could be ascribed to its unusually high expression level and to the ability of its single-stranded region to activate TLR7; both of these properties were dependent on M81-specific polymorphisms. Thus, M81 induced chronic inflammation in its target cells and this resulted in increased virus production. These observations provide a mechanistic molecular link between M81 virus replication-a central viral function and a cancer risk factor-and the production of a chemokine involved in inflammation and carcinogenesis.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/genetics , Nasopharyngeal Carcinoma/metabolism , Nasopharyngeal Carcinoma/virology , Nasopharyngeal Neoplasms/virology , RNA, Untranslated/genetics , Virus Replication , B-Lymphocytes/virology , Chemokines/metabolism , Epstein-Barr Virus Infections/immunology , HEK293 Cells , Host-Pathogen Interactions , Humans , Interleukin-8/metabolism , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Oncogenic Viruses , RNA, Viral , Toll-Like Receptor 7/metabolism , Virus Cultivation
4.
J Virol ; 93(5)2019 03 01.
Article in English | MEDLINE | ID: mdl-30541846

ABSTRACT

Epstein-Barr virus (EBV) infects the oropharynx but, surprisingly, frequently induces B cell proliferation in the gut of immunosuppressed individuals. We found that EBV infection in vitro induces the expression of the LPAM-1 integrin on tonsillar B cells and increases it on peripheral blood cells. Similarly, LPAM-1 was induced in the tonsils of patients undergoing primary infectious mononucleosis. EBV-induced LPAM-1 bound to the MAdCAM-1 addressin, which allows B cell homing to the gastrointestinal mucosa-associated lymphoid tissue (GALT). Thus, we hypothesized that EBV-induced LPAM-1 could induce relocation of infected B cells from the tonsil to the GALT. In situ hybridization with an EBER-specific probe revealed the frequent presence of EBV-infected cells in the pericolic lymph nodes of healthy individuals. Relocation of infected B cells into the GALT would expand the EBV reservoir, possibly protecting it from T cells primed in the oropharynx, and explain why EBV induces lymphoid tumors in the gut.IMPORTANCE EBV causes tumors in multiple organs, particularly in the oro- and nasopharyngeal area but also in the digestive system. This virus enters the body in the oropharynx and establishes a chronic infection in this area. The observation that the virus causes tumors in the digestive system implies that the infected cells can move to this organ. We found that EBV infection induces the expression of integrin beta 7 (ITGB7), an integrin that associates with integrin alpha 4 to form the LPAM-1 dimer. LPAM-1 is key for homing of B cells to the gastrointestinal tract, suggesting that induction of this molecule is the mechanism through which EBV-infected cells enter this organ. In favor of this hypothesis, we could also detect EBV-infected cells in the lymph nodes adjacent to the colon and in the appendix.


Subject(s)
B-Lymphocytes/metabolism , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/metabolism , Integrins/biosynthesis , Palatine Tonsil/metabolism , Animals , CHO Cells , Cell Movement/physiology , Cell Proliferation/physiology , Cells, Cultured , Cricetulus , Gastrointestinal Tract/cytology , Humans , Palatine Tonsil/cytology
5.
PLoS Pathog ; 14(12): e1007464, 2018 12.
Article in English | MEDLINE | ID: mdl-30521644

ABSTRACT

The ubiquitous Epstein-Barr virus (EBV) is the primary cause of infectious mononucleosis and is etiologically linked to the development of several malignancies and autoimmune diseases. EBV has a multifaceted life cycle that comprises virus lytic replication and latency programs. Considering EBV infection holistically, we rationalized that prophylactic EBV vaccines should ideally prime the immune system against lytic and latent proteins. To this end, we generated highly immunogenic particles that contain antigens from both these cycles. In addition to stimulating EBV-specific T cells that recognize lytic or latent proteins, we show that the immunogenic particles enable the ex vivo expansion of cytolytic EBV-specific T cells that efficiently control EBV-infected B cells, preventing their outgrowth. Lastly, we show that immunogenic particles containing the latent protein EBNA1 afford significant protection against wild-type EBV in a humanized mouse model. Vaccines that include antigens which predominate throughout the EBV life cycle are likely to enhance their ability to protect against EBV infection.


Subject(s)
Antigens, Viral/immunology , Epstein-Barr Virus Infections/immunology , Herpesvirus 4, Human/immunology , Herpesvirus Vaccines/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Mice , Virus Latency
7.
Nat Commun ; 8: 14257, 2017 02 10.
Article in English | MEDLINE | ID: mdl-28186092

ABSTRACT

Infections with Epstein-Barr virus (EBV) are associated with cancer development, and EBV lytic replication (the process that generates virus progeny) is a strong risk factor for some cancer types. Here we report that EBV infection of B-lymphocytes (in vitro and in a mouse model) leads to an increased rate of centrosome amplification, associated with chromosomal instability. This effect can be reproduced with virus-like particles devoid of EBV DNA, but not with defective virus-like particles that cannot infect host cells. Viral protein BNRF1 induces centrosome amplification, and BNRF1-deficient viruses largely lose this property. These findings identify a new mechanism by which EBV particles can induce chromosomal instability without establishing a chronic infection, thereby conferring a risk for development of tumours that do not necessarily carry the viral genome.


Subject(s)
Centrosome/virology , Chromosomal Instability , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line , Cell Line, Tumor , Cell Transformation, Neoplastic , Centrosome/metabolism , Epstein-Barr Virus Infections/genetics , HEK293 Cells , HeLa Cells , Herpesvirus 4, Human/genetics , Humans , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism , Virion/genetics , Virion/physiology
8.
Oncotarget ; 8(6): 10238-10254, 2017 Feb 07.
Article in English | MEDLINE | ID: mdl-28052012

ABSTRACT

The Epstein-Barr virus (EBV) is etiologically associated with the development of multiple types of tumors, but it is unclear whether this diversity is due to infection with different EBV strains. We report a comparative characterization of SNU719, GP202, and YCCEL1, three EBV strains that were isolated from gastric carcinomas, M81, a virus isolated in a nasopharyngeal carcinoma and several well-characterized laboratory type A strains. We found that B95-8, Akata and GP202 induced cell growth more efficiently than YCCEL1, SNU719 and M81 and this correlated positively with the expression levels of the viral BHRF1 miRNAs. In infected B cells, all strains except Akata and B95-8 induced lytic replication, a risk factor for carcinoma development, although less efficiently than M81. The panel of viruses induced tumors in immunocompromised mice with variable speed and efficacy that did not strictly mirror their in vitro characteristics, suggesting that additional parameters play an important role. We found that YCCEL1 and M81 infected primary epithelial cells, gastric carcinoma cells and gastric spheroids more efficiently than Akata or B95-8. Reciprocally, Akata and B95-8 had a stronger tropism for B cells than YCCEL1 or M81. These data suggest that different EBV strains will induce the development of lymphoid tumors with variable efficacy in immunocompromised patients and that there is a parallel between the cell tropism of the viral strains and the lineage of the tumors they induce. Thus, EBV strains can be endowed with properties that will influence their transforming abilities and the type of tumor they induce.


Subject(s)
Carcinoma/virology , Cell Transformation, Viral , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/pathogenicity , Nasopharyngeal Neoplasms/virology , Stomach Neoplasms/virology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , B-Lymphocytes/virology , Caco-2 Cells , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Cell Proliferation , Coculture Techniques , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/pathology , HEK293 Cells , Herpesvirus 4, Human/classification , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/isolation & purification , Host-Pathogen Interactions , Humans , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/metabolism , Nasopharyngeal Neoplasms/pathology , RNA, Viral/genetics , RNA, Viral/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Time Factors , Viral Proteins/genetics , Viral Proteins/metabolism , Viral Tropism , Virus Internalization , Virus Replication
9.
Cell Cycle ; 15(24): 3419-3431, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27754753

ABSTRACT

Topoisomerase IIα is an essential enzyme that resolves topological constraints in genomic DNA. It functions in disentangling intertwined chromosomes during anaphase leading to chromosome segregation thus preserving genomic stability. Here we describe a previously unrecognized mechanism regulating topoisomerase IIα activity that is dependent on the F-box protein Fbxo28. We find that Fbxo28, an evolutionarily conserved protein, is required for proper mitotic progression. Interfering with Fbxo28 function leads to a delay in metaphase-to-anaphase progression resulting in mitotic defects as lagging chromosomes, multipolar spindles and multinucleation. Furthermore, we find that Fbxo28 interacts and colocalizes with topoisomerase IIα throughout the cell cycle. Depletion of Fbxo28 results in an increase in topoisomerase IIα-dependent DNA decatenation activity. Interestingly, blocking the interaction between Fbxo28 and topoisomerase IIα also results in multinucleated cells. Our findings suggest that Fbxo28 regulates topoisomerase IIα decatenation activity and plays an important role in maintaining genomic stability.


Subject(s)
Antigens, Neoplasm/metabolism , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/metabolism , DNA/metabolism , Mitosis , SKP Cullin F-Box Protein Ligases/metabolism , Amino Acid Sequence , Chromosomes, Human/metabolism , Conserved Sequence , Down-Regulation , Evolution, Molecular , Gene Deletion , HEK293 Cells , HeLa Cells , Humans , Protein Binding , SKP Cullin F-Box Protein Ligases/chemistry
10.
PLoS Pathog ; 11(12): e1005344, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26694854

ABSTRACT

The Epstein-Barr virus (EBV) is a B lymphotropic virus that infects the majority of the human population. All EBV strains transform B lymphocytes, but some strains, such as M81, also induce spontaneous virus replication. EBV encodes 22 microRNAs (miRNAs) that form a cluster within the BART region of the virus and have been previously been found to stimulate tumor cell growth. Here we describe their functions in B cells infected by M81. We found that the BART miRNAs are downregulated in replicating cells, and that exposure of B cells in vitro or in vivo in humanized mice to a BART miRNA knockout virus resulted in an increased proportion of spontaneously replicating cells, relative to wild type virus. The BART miRNAs subcluster 1, and to a lesser extent subcluster 2, prevented expression of BZLF1, the key protein for initiation of lytic replication. Thus, multiple BART miRNAs cooperate to repress lytic replication. The BART miRNAs also downregulated pro- and anti-apoptotic mediators such as caspase 3 and LMP1, and their deletion did not sensitize B-cells to apoptosis. To the contrary, the majority of humanized mice infected with the BART miRNA knockout mutant developed tumors more rapidly, probably due to enhanced LMP1 expression, although deletion of the BART miRNAs did not modify the virus transforming abilities in vitro. This ability to slow cell growth could be confirmed in non-humanized immunocompromized mice. Injection of resting B cells exposed to a virus that lacks the BART miRNAs resulted in accelerated tumor growth, relative to wild type controls. Therefore, we found that the M81 BART miRNAs do not enhance B-cell tumorigenesis but rather repress it. The repressive effects of the BART miRNAs on potentially pathogenic viral functions in infected B cells are likely to facilitate long-term persistence of the virus in the infected host.


Subject(s)
B-Lymphocytes/virology , Epstein-Barr Virus Infections/genetics , Gene Expression Regulation, Viral/genetics , Herpesvirus 4, Human/genetics , MicroRNAs/genetics , Virus Replication/genetics , Animals , Blotting, Western , Cell Transformation, Viral/genetics , Genes, Viral , Humans , Immunohistochemistry , Immunoprecipitation , Mice , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Transfection
11.
PLoS Pathog ; 10(8): e1004333, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25165855

ABSTRACT

Epstein Barr virus (EBV) infection expands CD8+ T cells specific for lytic antigens to high frequencies during symptomatic primary infection, and maintains these at significant numbers during persistence. Despite this, the protective function of these lytic EBV antigen-specific cytotoxic CD8+ T cells remains unclear. Here we demonstrate that lytic EBV replication does not significantly contribute to virus-induced B cell proliferation in vitro and in vivo in a mouse model with reconstituted human immune system components (huNSG mice). However, we report a trend to reduction of EBV-induced lymphoproliferation outside of lymphoid organs upon diminished lytic replication. Moreover, we could demonstrate that CD8+ T cells against the lytic EBV antigen BMLF1 can eliminate lytically replicating EBV-transformed B cells from lymphoblastoid cell lines (LCLs) and in vivo, thereby transiently controlling high viremia after adoptive transfer into EBV infected huNSG mice. These findings suggest a protective function for lytic EBV antigen-specific CD8+ T cells against EBV infection and against virus-associated tumors in extra-lymphoid organs. These specificities should be explored for EBV-specific vaccine development.


Subject(s)
B-Lymphocytes/virology , Cell Transformation, Viral/physiology , Epstein-Barr Virus Infections/immunology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer , Animals , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Real-Time Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...