Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
ACS Nano ; 17(9): 8622-8633, 2023 May 09.
Article in English | MEDLINE | ID: mdl-37129379

ABSTRACT

We have achieved the synthesis of dual-metal single atoms and atomic clusters that co-anchor on a highly graphitic carbon support. The catalyst comprises Ni4 (and Fe4) nanoclusters located adjacent to the corresponding NiN4 (and FeN4) single-atom sites, which is verified by systematic X-ray absorption characterization and density functional theory calculations. A distinct cooperation between Fe4 (Ni4) nanoclusters and the corresponding FeN4 (NiN4) atomic sites optimizes the adsorption energy of reaction intermediates and reduces the energy barrier of the potential-determining steps. This catalyst exhibits enhanced oxygen reduction and evolution activity and long-cycle stability compared to counterparts without nanoclusters and commercial Pt/C. The fabricated Zn-air batteries deliver a high power density and long-term cyclability, demonstrating their prospects in energy storage device applications.

2.
ACS Appl Mater Interfaces ; 15(5): 7392-7404, 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36693331

ABSTRACT

Conductive polymer composite-based strain sensors are essential components of flexible wearable devices. However, nonmonotonic responses with shoulder peaks limit their practical application. Herein, we innovatively optimized the shoulder-peak phenomenon in a strain-sensing composite nanofiber filament by regulating carbon nanomaterial dispersion. Further, the preparation methods, characteristics, and performances of the filament strain sensors were systematically introduced. On this basis, transmission electron microscopy, finite element analysis, and mathematic and structural evolution models were used to explore the origin of shoulder peaks and explain the sensing mechanism of conductive networks. Results confirmed that the beacon tower-shaped conductive network designed by constructing nanofiller agglomerates could cause strain concentration and resist the Poisson transverse contraction of nanofibers, considerably improving the monotonicity and sensitivity of the sensor. The strain-sensing performance was optimal when the nanofillers were dispersed using 2.5 wt % of an anionic dispersant. The sensor exhibited a maximum detective strain of 120%, an ultralow detection limit of 0.01%, and high sensitivity and linearity of 9.66 and 0.996 within 20% strain, respectively. Moreover, it showed the advantages of a fast response time (120 ms), excellent durability (3000 cycles), anti-interference, washability, and antibacterial capability. Finally, a smart Kinesio tape was developed for protecting/treating the human body and detecting joint/muscle movement via simple sewing.


Subject(s)
Nanofibers , Nanostructures , Wearable Electronic Devices , Humans , Nanofibers/chemistry , Carbon , Shoulder
3.
iScience ; 25(10): 105162, 2022 Oct 21.
Article in English | MEDLINE | ID: mdl-36212024

ABSTRACT

Recently, various strain-sensing yarns have been developed without ideal stitchability. Herein, we used spherical carbon black particles (CBs), linear carbon nanotubes (CNTs), and lamellar graphene flakes (GRs) as conductive nanofillers to construct multi-element conductive networks inside a thermoplastic polyurethane (TPU) matrix. First, a highly stretchable and conductive multidimensional carbon-based nanomaterial/TPU composite nanofiber yarn was fabricated using electrospinning, which could be used as a flexible strain sensor without post-processing. Accordingly, the effects of nanomaterials' dimensionality and synergy on yarns' conductivity, mechanical properties, and strain sensing performances were explored. The yarn containing multiple networks formed by CB/CNT/GR ternary hybrid networks, CNT and GR auxiliary networks exhibited the best performances. Subsequently, the structural evolution of the ternary conductive network under stretching was revealed to further analyze the sensing mechanism. Finally, the yarn endowed a medicated plaster with an intelligent function to detect motions in the rehabilitation of joint pain by simple sewing.

4.
Life Sci Alliance ; 4(7)2021 07.
Article in English | MEDLINE | ID: mdl-34112724

ABSTRACT

Invariant natural killer T (iNKT) cells are a conserved population of innate T lymphocytes that interact with key antigen-presenting cells to modulate adaptive T-cell responses in ways that can either promote protective immunity, or limit pathological immune activation. Understanding the immunological networks engaged by iNKT cells to mediate these opposing functions is a key pre-requisite to effectively using iNKT cells for therapeutic applications. Using a human umbilical cord blood xenotransplantation model, we show here that co-transplanted allogeneic CD4+ iNKT cells interact with monocytes and T cells in the graft to coordinate pro-hematopoietic and immunoregulatory pathways. The nexus of iNKT cells, monocytes, and cord blood T cells led to the release of cytokines (IL-3, GM-CSF) that enhance hematopoietic stem and progenitor cell activity, and concurrently induced PGE2-mediated suppression of T-cell inflammatory responses that limit hematopoietic stem and progenitor cell engraftment. This resulted in successful long-term hematopoietic engraftment without pretransplant conditioning, including multi-lineage human chimerism and colonization of the spleen by antibody-producing human B cells. These results highlight the potential for using iNKT cellular immunotherapy to improve rates of hematopoietic engraftment independently of pretransplant conditioning.


Subject(s)
Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Transplantation Immunology/immunology , Animals , Antigen-Presenting Cells/immunology , Cytokines/immunology , Female , Fetal Blood/immunology , Humans , Immunity, Innate/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Tissue Transplantation/methods
5.
J Virol ; 94(10)2020 05 04.
Article in English | MEDLINE | ID: mdl-32132242

ABSTRACT

Epstein-Barr virus (EBV) causes B cell lymphomas and transforms B cells in vitro The EBV protein EBNA3A collaborates with EBNA3C to repress p16 expression and is required for efficient transformation in vitro An EBNA3A deletion mutant EBV strain was recently reported to establish latency in humanized mice but not cause tumors. Here, we compare the phenotypes of an EBNA3A mutant EBV (Δ3A) and wild-type (WT) EBV in a cord blood-humanized (CBH) mouse model. The hypomorphic Δ3A mutant, in which a stop codon is inserted downstream from the first ATG and the open reading frame is disrupted by a 1-bp insertion, expresses very small amounts of EBNA3A using an alternative ATG at residue 15. Δ3A caused B cell lymphomas at rates similar to their induction by WT EBV but with delayed onset. Δ3A and WT tumors expressed equivalent levels of EBNA2 and p16, but Δ3A tumors in some cases had reduced LMP1. Like the WT EBV tumors, Δ3A lymphomas were oligoclonal/monoclonal, with typically one dominant IGHV gene being expressed. Transcriptome sequencing (RNA-seq) analysis revealed small but consistent gene expression differences involving multiple cellular genes in the WT EBV- versus Δ3A-infected tumors and increased expression of genes associated with T cells, suggesting increased T cell infiltration of tumors. Consistent with an impact of EBNA3A on immune function, we found that the expression of CLEC2D, a receptor that has previously been shown to influence responses of T and NK cells, was markedly diminished in cells infected with EBNA3A mutant virus. Together, these studies suggest that EBNA3A contributes to efficient EBV-induced lymphomagenesis in CBH mice.IMPORTANCE The EBV protein EBNA3A is expressed in latently infected B cells and is important for efficient EBV-induced transformation of B cells in vitro In this study, we used a cord blood-humanized mouse model to compare the phenotypes of an EBNA3A hypomorph mutant virus (Δ3A) and wild-type EBV. The Δ3A virus caused lymphomas with delayed onset compared to the onset of those caused by WT EBV, although the tumors occurred at a similar rate. The WT EBV and EBNA3A mutant tumors expressed similar levels of the EBV protein EBNA2 and cellular protein p16, but in some cases, Δ3A tumors had less LMP1. Our analysis suggested that Δ3A-infected tumors have elevated T cell infiltrates and decreased expression of the CLEC2D receptor, which may point to potential novel roles of EBNA3A in T cell and NK cell responses to EBV-infected tumors.


Subject(s)
Epstein-Barr Virus Infections/virology , Epstein-Barr Virus Nuclear Antigens/genetics , Epstein-Barr Virus Nuclear Antigens/metabolism , Fetal Blood/metabolism , Herpesvirus 4, Human/genetics , Lymphoma/virology , Animals , B-Lymphocytes/virology , Cell Transformation, Viral , Disease Models, Animal , Gene Expression Regulation, Neoplastic , HEK293 Cells , Herpesvirus 4, Human/physiology , Humans , Killer Cells, Natural/immunology , Lymphoma/genetics , Lymphoma/pathology , Lymphoma, B-Cell , Mice , Mutagenesis, Site-Directed , Sequence Analysis, RNA , Sequence Deletion , T-Lymphocytes/immunology , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Latency/genetics
6.
PLoS Pathog ; 14(8): e1007221, 2018 08.
Article in English | MEDLINE | ID: mdl-30125329

ABSTRACT

EBV causes human B-cell lymphomas and transforms B cells in vitro. EBNA3C, an EBV protein expressed in latently-infected cells, is required for EBV transformation of B cells in vitro. While EBNA3C undoubtedly plays a key role in allowing EBV to successfully infect B cells, many EBV+ lymphomas do not express this protein, suggesting that cellular mutations and/or signaling pathways may obviate the need for EBNA3C in vivo under certain conditions. EBNA3C collaborates with EBNA3A to repress expression of the CDKN2A-encoded tumor suppressors, p16 and p14, and EBNA3C-deleted EBV transforms B cells containing a p16 germline mutation in vitro. Here we have examined the phenotype of an EBNAC-deleted virus (Δ3C EBV) in a cord blood-humanized mouse model (CBH). We found that the Δ3C virus induced fewer lymphomas (occurring with a delayed onset) in comparison to the wild-type (WT) control virus, although a subset (10/26) of Δ3C-infected CBH mice eventually developed invasive diffuse large B cell lymphomas with type III latency. Both WT and Δ3C viruses induced B-cell lymphomas with restricted B-cell populations and heterogeneous T-cell infiltration. In comparison to WT-infected tumors, Δ3C-infected tumors had greatly increased p16 levels, and RNA-seq analysis revealed a decrease in E2F target gene expression. However, we found that Δ3C-infected tumors expressed c-Myc and cyclin E at similar levels compared to WT-infected tumors, allowing cells to at least partially bypass p16-mediated cell cycle inhibition. The anti-apoptotic proteins, BCL2 and IRF4, were expressed in Δ3C-infected tumors, likely helping cells avoid c-Myc-induced apoptosis. Unexpectedly, Δ3C-infected tumors had increased T-cell infiltration, increased expression of T-cell chemokines (CCL5, CCL20 and CCL22) and enhanced type I interferon response in comparison to WT tumors. Together, these results reveal that EBNA3C contributes to, but is not essential for, EBV-induced lymphomagenesis in CBH mice, and suggest potentially important immunologic roles of EBNA3C in vivo.


Subject(s)
Cell Transformation, Viral/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Nuclear Antigens/genetics , Herpesvirus 4, Human/physiology , Lymphoma, B-Cell/virology , Virus Latency/genetics , Animals , Cells, Cultured , Disease Models, Animal , Epstein-Barr Virus Infections/genetics , Fetal Blood/immunology , HEK293 Cells , Herpesvirus 4, Human/genetics , Humans , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/pathology , Mice , Mice, Inbred NOD , Mice, Transgenic
7.
JCI Insight ; 2(13)2017 Jul 06.
Article in English | MEDLINE | ID: mdl-28679955

ABSTRACT

A central issue for adoptive cellular immunotherapy is overcoming immunosuppressive signals to achieve tumor clearance. While γδ T cells are known to be potent cytolytic effectors that can kill a variety of cancers, it is not clear whether they are inhibited by suppressive ligands expressed in tumor microenvironments. Here, we have used a powerful preclinical model where EBV infection drives the de novo generation of human B cell lymphomas in vivo, and autologous T lymphocytes are held in check by PD-1/CTLA-4-mediated inhibition. We show that a single dose of adoptively transferred Vδ2+ T cells has potent antitumor effects, even in the absence of checkpoint blockade or activating compounds. Vδ2+ T cell immunotherapy given within the first 5 days of EBV infection almost completely prevented the outgrowth of tumors. Vδ2+ T cell immunotherapy given more than 3 weeks after infection (after neoplastic transformation is evident) resulted in a dramatic reduction in tumor burden. The immunotherapeutic Vδ2+ T cells maintained low cell surface expression of PD-1 in vivo, and their recruitment to tumors was followed by a decrease in B cells expressing PD-L1 and PD-L2 inhibitory ligands. These results suggest that adoptively transferred PD-1lo Vδ2+ T cells circumvent the tumor checkpoint environment in vivo.

8.
PLoS Pathog ; 13(6): e1006404, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28617871

ABSTRACT

When confronted with poor oxygenation, cells adapt by activating survival signaling pathways, including the oxygen-sensitive transcriptional regulators called hypoxia-inducible factor alphas (HIF-αs). We report here that HIF-1α also regulates the life cycle of Epstein-Barr virus (EBV). Incubation of EBV-positive gastric carcinoma AGS-Akata and SNU-719 and Burkitt lymphoma Sal and KemIII cell lines with a prolyl hydroxylase inhibitor, L-mimosine or deferoxamine, or the NEDDylation inhibitor MLN4924 promoted rapid and sustained accumulation of both HIF-1α and lytic EBV antigens. ShRNA knockdown of HIF-1α significantly reduced deferoxamine-mediated lytic reactivation. HIF-1α directly bound the promoter of the EBV primary latent-lytic switch BZLF1 gene, Zp, activating transcription via a consensus hypoxia-response element (HRE) located at nt -83 through -76 relative to the transcription initiation site. HIF-1α did not activate transcription from the other EBV immediate-early gene, BRLF1. Importantly, expression of HIF-1α induced EBV lytic-gene expression in cells harboring wild-type EBV, but not in cells infected with variants containing base-pair substitution mutations within this HRE. Human oral keratinocyte (NOK) and gingival epithelial (hGET) cells induced to differentiate by incubation with either methyl cellulose or growth in organotypic culture accumulated both HIF-1α and Blimp-1α, another cellular factor implicated in lytic reactivation. HIF-1α activity also accumulated along with Blimp-1α during B-cell differentiation into plasma cells. Furthermore, most BZLF1-expressing cells observed in lymphomas induced by EBV in NSG mice with a humanized immune system were located distal to blood vessels in hypoxic regions of the tumors. Thus, we conclude that HIF-1α plays central roles in both EBV's natural life cycle and EBV-associated tumorigenesis. We propose that drugs that induce HIF-1α protein accumulation are good candidates for development of a lytic-induction therapy for treating some EBV-associated malignancies.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Epstein-Barr Virus Infections/virology , Gene Expression Regulation, Viral , Herpesvirus 4, Human/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphoma/metabolism , Trans-Activators/genetics , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Carcinogenesis , Cell Line, Tumor , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/growth & development , Host-Pathogen Interactions , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Lymphoma/genetics , Lymphoma/virology , Mice , Promoter Regions, Genetic , Protein Binding , Trans-Activators/metabolism , Virus Activation
9.
Oncotarget ; 8(27): 44266-44280, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28574826

ABSTRACT

EBV infection causes mononucleosis and is associated with specific subsets of B cell lymphomas. Immunosuppressed patients such as organ transplant recipients are particularly susceptible to EBV-induced lymphoproliferative disease (LPD), which can be fatal. Leflunomide (a drug used to treat rheumatoid arthritis) and its active metabolite teriflunomide (used to treat multiple sclerosis) inhibit de novo pyrimidine synthesis by targeting the cellular dihydroorotate dehydrogenase, thereby decreasing T cell proliferation. Leflunomide also inhibits the replication of cytomegalovirus and BK virus via both "on target" and "off target" mechanisms and is increasingly used to treat these viruses in organ transplant recipients. However, whether leflunomide/teriflunomide block EBV replication or inhibit EBV-mediated B cell transformation is currently unknown. We show that teriflunomide inhibits cellular proliferation, and promotes apoptosis, in EBV-transformed B cells in vitro at a clinically relevant dose. In addition, teriflunomide prevents the development of EBV-induced lymphomas in both a humanized mouse model and a xenograft model. Furthermore, teriflunomide inhibits lytic EBV infection in vitro both by preventing the initial steps of lytic viral reactivation, and by blocking lytic viral DNA replication. Leflunomide/teriflunomide might therefore be clinically useful for preventing EBV-induced LPD in patients who have high EBV loads yet require continued immunosuppression.


Subject(s)
Crotonates/pharmacology , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/drug effects , Herpesvirus 4, Human/physiology , Isoxazoles/pharmacology , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/pathology , Toluidines/pharmacology , Virus Replication/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , B-Lymphocytes/virology , Cell Line, Transformed , Cell Proliferation/drug effects , Cyclin E/genetics , Disease Models, Animal , Epstein-Barr Virus Infections/drug therapy , Gene Expression Regulation/drug effects , Gene Expression Regulation, Viral/drug effects , Genes, myc , Humans , Hydroxybutyrates , Leflunomide , Lymphoproliferative Disorders/drug therapy , Mice , NF-kappa B/metabolism , Nitriles , Virus Activation/drug effects , Virus Latency/drug effects , Virus Latency/genetics , Xenograft Model Antitumor Assays
10.
J Virol ; 91(7)2017 04 01.
Article in English | MEDLINE | ID: mdl-28077657

ABSTRACT

Epstein-Barr virus (EBV) infection is associated with B cell lymphomas in humans. The ability of EBV to convert human B cells into long-lived lymphoblastoid cell lines (LCLs) in vitro requires the collaborative effects of EBNA2 (which hijacks Notch signaling), latent membrane protein 1 (LMP1) (which mimics CD40 signaling), and EBV-encoded nuclear antigen 3A (EBNA3A) and EBNA3C (which inhibit oncogene-induced senescence and apoptosis). However, we recently showed that an LMP1-deleted EBV mutant induces B cell lymphomas in a newly developed cord blood-humanized mouse model that allows EBV-infected B cells to interact with CD4 T cells (the major source of CD40 ligand). Here we examined whether the EBV LMP2A protein, which mimics constitutively active B cell receptor signaling, is required for EBV-induced lymphomas in this model. We find that the deletion of LMP2A delays the onset of EBV-induced lymphomas but does not affect the tumor phenotype or the number of tumors. The simultaneous deletion of both LMP1 and LMP2A results in fewer tumors and a further delay in tumor onset. Nevertheless, the LMP1/LMP2A double mutant induces lymphomas in approximately half of the infected animals. These results indicate that neither LMP1 nor LMP2A is absolutely essential for the ability of EBV to induce B cell lymphomas in the cord blood-humanized mouse model, although the simultaneous loss of both LMP1 and LMP2A decreases the proportion of animals developing tumors and increases the time to tumor onset. Thus, the expression of either LMP1 or LMP2A may be sufficient to promote early-onset EBV-induced tumors in this model.IMPORTANCE EBV causes human lymphomas, but few models are available for dissecting how EBV causes lymphomas in vivo in the context of a host immune response. We recently used a newly developed cord blood-humanized mouse model to show that EBV can cooperate with human CD4 T cells to cause B cell lymphomas even when a major viral transforming protein, LMP1, is deleted. Here we examined whether the EBV protein LMP2A, which mimics B cell receptor signaling, is required for EBV-induced lymphomas in this model. We find that the deletion of LMP2A alone has little effect on the ability of EBV to cause lymphomas but delays tumor onset. The deletion of both LMP1 and LMP2A results in a smaller number of lymphomas in infected animals, with an even more delayed time to tumor onset. These results suggest that LMP1 and LMP2A collaborate to promote early-onset lymphomas in this model, but neither protein is absolutely essential.


Subject(s)
Epstein-Barr Virus Infections/virology , Herpesvirus 4, Human/physiology , Lymphoma, Large B-Cell, Diffuse/virology , Viral Matrix Proteins/physiology , Animals , Cell Transformation, Neoplastic , Cells, Cultured , Epstein-Barr Virus Infections/immunology , Gene Knockout Techniques , Humans , Lymphocytes, Tumor-Infiltrating/physiology , Lymphoma, Large B-Cell, Diffuse/immunology , Mice, Inbred NOD , Mice, SCID
11.
Surgery ; 161(1): 195-201, 2017 01.
Article in English | MEDLINE | ID: mdl-27847111

ABSTRACT

BACKGROUND: Epstein-Barr virus is associated with lymphoid and epithelial malignancies and has been reported to infect thyroid cells. The Epstein-Barr virus protein, EBNA2, regulates viral and cellular promoters by binding to RBP-jκ. Similarly, NOTCH1, a tumor suppressor protein in thyroid epithelial cells, competes with EBNA2 for binding to overlapping sites on RBP-jκ. EBNA2 activates a subset of NOTCH-responsive genes in lymphocytes and myocytes; however, the effect of EBNA2 expression on NOTCH targets in epithelial cells is unknown. Here we have explored whether EBNA2 activates NOTCH1 targets in thyroid cancer lines and examined its effect on cellular proliferation. METHODS: Two human thyroid cancer lines, follicular FTC-236 and anaplastic HTh7, were transfected with EBNA2, NOTCH1, or control vectors. Notch targets were measured using quantitative reverse transcriptase polymerase chain reaction. Cellular proliferation was measured by MTT analysis. RESULTS: EBNA2 activated only a subset of NOTCH1 targets. Expression of HES1 and HEY1 were increased 10-fold in FTC-236 and HTh7 cells, respectively, but the majority of NOTCH1 targets examined were not affected. In contrast to NOTCH1, EBNA2 did not suppress proliferation. CONCLUSION: EBNA2 does not activate most Notch1-responsive genes or suppress proliferation in human thyroid cancer cells. Instead, EBNA2 may compete with NOTCH1 for limiting amounts of RBP-jκ in epithelial cells and inhibit certain aspects of NOTCH1 signaling.


Subject(s)
Carrier Proteins/genetics , Herpesvirus 4, Human/genetics , Receptor, Notch1/genetics , Thyroid Neoplasms/virology , Transcriptional Activation/genetics , Cell Line , Gene Expression Regulation, Viral , Humans , Membrane Proteins/genetics , RNA-Binding Proteins , Sampling Studies , Sensitivity and Specificity , Signal Transduction , Thyroid Neoplasms/genetics
12.
PLoS Pathog ; 12(5): e1005642, 2016 05.
Article in English | MEDLINE | ID: mdl-27186886

ABSTRACT

Epstein-Barr virus (EBV) infection causes B cell lymphomas in humanized mouse models and contributes to a variety of different types of human lymphomas. T cells directed against viral antigens play a critical role in controlling EBV infection, and EBV-positive lymphomas are particularly common in immunocompromised hosts. We previously showed that EBV induces B cell lymphomas with high frequency in a cord blood-humanized mouse model in which EBV-infected human cord blood is injected intraperitoneally into NOD/LtSz-scid/IL2Rγnull (NSG) mice. Since our former studies showed that it is possible for T cells to control the tumors in another NSG mouse model engrafted with both human fetal CD34+ cells and human thymus and liver, here we investigated whether monoclonal antibodies that block the T cell inhibitory receptors, PD-1 and CTLA-4, enhance the ability of cord blood T cells to control the outgrowth of EBV-induced lymphomas in the cord-blood humanized mouse model. We demonstrate that EBV-infected lymphoma cells in this model express both the PD-L1 and PD-L2 inhibitory ligands for the PD-1 receptor, and that T cells express the PD-1 and CTLA-4 receptors. Furthermore, we show that the combination of CTLA-4 and PD-1 blockade strikingly reduces the size of lymphomas induced by a lytic EBV strain (M81) in this model, and that this anti-tumor effect requires T cells. PD-1/CTLA-4 blockade markedly increases EBV-specific T cell responses, and is associated with enhanced tumor infiltration by CD4+ and CD8+ T cells. In addition, PD-1/CTLA-4 blockade decreases the number of both latently, and lytically, EBV-infected B cells. These results indicate that PD-1/CTLA-4 blockade enhances the ability of cord blood T cells to control outgrowth of EBV-induced lymphomas, and suggest that PD-1/CTLA-4 blockade might be useful for treating certain EBV-induced diseases in humans.


Subject(s)
Epstein-Barr Virus Infections/complications , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/virology , Programmed Cell Death 1 Receptor/metabolism , Animals , CTLA-4 Antigen/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Epstein-Barr Virus Infections/immunology , Fetal Blood , Flow Cytometry , Herpesvirus 4, Human , Humans , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred NOD , Mice, SCID
13.
Proc Natl Acad Sci U S A ; 112(52): E7257-65, 2015 Dec 29.
Article in English | MEDLINE | ID: mdl-26663912

ABSTRACT

Latent Epstein-Barr virus (EBV) infection and cellular hypermethylation are hallmarks of undifferentiated nasopharyngeal carcinoma (NPC). However, EBV infection of normal oral epithelial cells is confined to differentiated cells and is lytic. Here we demonstrate that the EBV genome can become 5-hydroxymethylated and that this DNA modification affects EBV lytic reactivation. We show that global 5-hydroxymethylcytosine (5hmC)-modified DNA accumulates during normal epithelial-cell differentiation, whereas EBV+ NPCs have little if any 5hmC-modified DNA. Furthermore, we find that increasing cellular ten-eleven translocation (TET) activity [which converts methylated cytosine (5mC) to 5hmC] decreases methylation, and increases 5hmC modification, of lytic EBV promoters in EBV-infected cell lines containing highly methylated viral genomes. Conversely, inhibition of endogenous TET activity increases lytic EBV promoter methylation in an EBV-infected telomerase-immortalized normal oral keratinocyte (NOKs) cell line where lytic viral promoters are largely unmethylated. We demonstrate that these cytosine modifications differentially affect the ability of the two EBV immediate-early proteins, BZLF1 (Z) and BRLF1 (R), to induce the lytic form of viral infection. Although methylation of lytic EBV promoters increases Z-mediated and inhibits R-mediated lytic reactivation, 5hmC modification of lytic EBV promoters has the opposite effect. We also identify a specific CpG-containing Z-binding site on the BRLF1 promoter that must be methylated for Z-mediated viral reactivation and show that TET-mediated 5hmC modification of this site in NOKs prevents Z-mediated viral reactivation. Decreased 5-hydroxymethylation of cellular and viral genes may contribute to NPC formation.


Subject(s)
DNA Methylation , Genome, Viral/genetics , Herpesvirus 4, Human/genetics , Virus Activation/genetics , Virus Latency/genetics , Base Sequence , Binding Sites/genetics , Carcinoma , Cell Differentiation/genetics , Cell Line , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Dioxygenases , HEK293 Cells , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Immunoblotting , Keratinocytes/metabolism , Keratinocytes/virology , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/genetics , Nasopharyngeal Neoplasms/pathology , Nasopharyngeal Neoplasms/virology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism
14.
PLoS Pathog ; 11(10): e1005195, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26431332

ABSTRACT

Epstein-Barr virus (EBV) is a human herpesvirus associated with B-cell and epithelial cell malignancies. EBV lytically infects normal differentiated oral epithelial cells, where it causes a tongue lesion known as oral hairy leukoplakia (OHL) in immunosuppressed patients. However, the cellular mechanism(s) that enable EBV to establish exclusively lytic infection in normal differentiated oral epithelial cells are not currently understood. Here we show that a cellular transcription factor known to promote epithelial cell differentiation, KLF4, induces differentiation-dependent lytic EBV infection by binding to and activating the two EBV immediate-early gene (BZLF1 and BRLF1) promoters. We demonstrate that latently EBV-infected, telomerase-immortalized normal oral keratinocyte (NOKs) cells undergo lytic viral reactivation confined to the more differentiated cell layers in organotypic raft culture. Furthermore, we show that endogenous KLF4 expression is required for efficient lytic viral reactivation in response to phorbol ester and sodium butyrate treatment in several different EBV-infected epithelial cell lines, and that the combination of KLF4 and another differentiation-dependent cellular transcription factor, BLIMP1, is highly synergistic for inducing lytic EBV infection. We confirm that both KLF4 and BLIMP1 are expressed in differentiated, but not undifferentiated, epithelial cells in normal tongue tissue, and show that KLF4 and BLIMP1 are both expressed in a patient-derived OHL lesion. In contrast, KLF4 protein is not detectably expressed in B cells, where EBV normally enters latent infection, although KLF4 over-expression is sufficient to induce lytic EBV reactivation in Burkitt lymphoma cells. Thus, KLF4, together with BLIMP1, plays a critical role in mediating lytic EBV reactivation in epithelial cells.


Subject(s)
Epithelial Cells/virology , Epstein-Barr Virus Infections/metabolism , Kruppel-Like Transcription Factors/metabolism , Repressor Proteins/metabolism , Virus Activation/physiology , Adult , Cell Differentiation/physiology , Cell Line , Chromatin Immunoprecipitation , Epithelial Cells/pathology , Fluorescent Antibody Technique , Host-Pathogen Interactions/physiology , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Kruppel-Like Factor 4 , Laser Capture Microdissection , Leukoplakia, Hairy/metabolism , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Positive Regulatory Domain I-Binding Factor 1 , Virus Latency/physiology
15.
J Clin Invest ; 125(1): 304-15, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25485679

ABSTRACT

Epstein-Barr virus (EBV) infection transforms B cells in vitro and is associated with human B cell lymphomas. The major EBV oncoprotein, latent membrane protein 1 (LMP1), mimics constitutively active CD40 and is essential for outgrowth of EBV-transformed B cells in vitro; however, EBV-positive diffuse large B cell lymphomas and Burkitt lymphomas often express little or no LMP1. Thus, EBV may contribute to the development and maintenance of human lymphomas even in the absence of LMP1. Here, we found that i.p. injection of human cord blood mononuclear cells infected with a LMP1-deficient EBV into immunodeficient mice induces B cell lymphomas. In this model, lymphoma development required the presence of CD4+ T cells in cord blood and was inhibited by CD40-blocking Abs. In contrast, LMP1-deficient EBV established persistent latency but did not induce lymphomas when directly injected into mice engrafted with human fetal CD34+ cells and human thymus. WT EBV induced lymphomas in both mouse models and did not require coinjected T cells in the cord blood model. Together, these results demonstrate that LMP1 is not essential for EBV-induced lymphomas in vivo and suggest that T cells supply signals that substitute for LMP1 in EBV-positive B cell lymphomagenesis.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/genetics , Lymphoma/virology , Viral Matrix Proteins/genetics , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Antigens/metabolism , Carcinogenesis , Cell Proliferation , Epstein-Barr Virus Infections/immunology , Gene Expression , Gene Knockout Techniques , Herpesvirus 4, Human/immunology , Humans , Lymphoma/immunology , Lymphoma/pathology , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Signal Transduction , Tumor Cells, Cultured , Viral Matrix Proteins/metabolism , Virus Latency
16.
Biol Blood Marrow Transplant ; 19(9): 1310-22, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23806772

ABSTRACT

Chronic graft-versus-host disease (cGVHD) is a significant roadblock to long-term hematopoietic stem cell (HSC) transplantation success. Effective treatments for cGVHD have been difficult to develop, in part because of a paucity of animal models that recapitulate the multiorgan pathologies observed in clinical cGVHD. Here we present an analysis of the pathology that occurs in immunodeficient mice engrafted with human fetal HSCs and implanted with fragments of human fetal thymus and liver. Starting at time points generally later than 100 days post-transplantation, the mice developed signs of illness, including multiorgan cellular infiltrates containing human T cells, B cells, and macrophages; fibrosis in sites such as lungs and liver; and thickened skin with alopecia. Experimental manipulations that delayed or reduced the efficiency of the HSC engraftment did not affect the timing or progression of disease manifestations, suggesting that pathology in this model is driven more by factors associated with the engrafted human thymic organoid. Disease progression was typically accompanied by extensive fibrosis and degradation of the thymic organoid, and there was an inverse correlation of disease severity with the frequency of FoxP3(+) thymocytes. Hence, the human thymic tissue may contribute T cells with pathogenic potential, but the generation of regulatory T cells in the thymic organoid may help to control these cells before pathology resembling cGVHD eventually develops. This model thus provides a new system to investigate disease pathophysiology relating to human thymic events and to evaluate treatment strategies to combat multiorgan fibrotic pathology produced by human immune cells.


Subject(s)
Fetal Tissue Transplantation/methods , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/immunology , Thymus Gland/transplantation , Animals , Disease Models, Animal , Disease Progression , Female , Flow Cytometry , Graft vs Host Disease/pathology , Hematopoietic Stem Cells/pathology , Humans , Male , Mice , Mice, Inbred NOD , Transplantation, Heterologous
17.
J Virol ; 86(15): 7976-87, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22623780

ABSTRACT

Immunosuppressed patients are at risk for developing Epstein-Barr Virus (EBV)-positive lymphomas that express the major EBV oncoprotein, LMP1. Although increasing evidence suggests that a small number of lytically infected cells may promote EBV-positive lymphomas, the impact of enhanced lytic gene expression on the ability of EBV to induce lymphomas is unclear. Here we have used immune-deficient mice, engrafted with human fetal hematopoietic stem cells and thymus and liver tissue, to compare lymphoma formation following infection with wild-type (WT) EBV versus infection with a "superlytic" (SL) mutant with enhanced BZLF1 (Z) expression. The same proportions (2/6) of the WT and SL virus-infected animals developed B-cell lymphomas by day 60 postinfection; the remainder of the animals had persistent tumor-free viral latency. In contrast, all WT and SL virus-infected animals treated with the OKT3 anti-CD3 antibody (which inhibits T-cell function) developed lymphomas by day 29. Lymphomas in OKT3-treated animals (in contrast to lymphomas in the untreated animals) contained many LMP1-expressing cells. The SL virus-infected lymphomas in both OKT3-treated and untreated animals contained many more Z-expressing cells (up to 30%) than the WT virus-infected lymphomas, but did not express late viral proteins and thus had an abortive lytic form of EBV infection. LMP1 and BMRF1 (an early lytic viral protein) were never coexpressed in the same cell, suggesting that LMP1 expression is incompatible with lytic viral reactivation. These results show that the SL mutant induces an "abortive" lytic infection in humanized mice that is compatible with continued cell growth and at least partially resistant to T-cell killing.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Gene Expression Regulation, Viral , Herpesvirus 4, Human/metabolism , Lymphoma/metabolism , Lymphoma/virology , Mutation , Trans-Activators/biosynthesis , Animals , Disease Models, Animal , Epstein-Barr Virus Infections/genetics , Herpesvirus 4, Human/genetics , Humans , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Mutant Strains , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , T-Lymphocytes/virology , Trans-Activators/genetics , Virus Latency/genetics
18.
J Autoimmun ; 37(1): 28-38, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21486688

ABSTRACT

NKT cells are innate lymphocytes that can recognize self or foreign lipids presented by CD1d molecules. NKT cells have been shown to inhibit the development of autoimmunity in murine model systems, however, the pathways by which they foster immune tolerance remain poorly understood. Here we show that autoreactive human NKT cells stimulate monocytes to differentiate into myeloid APCs that have a regulatory phenotype characterized by poor conjugate formation with T cells. The NKT cell instructed myeloid APCs show elevated expression of the inhibitory ligand PD-L2, and blocking PD-L1 and PD-L2 during interactions of the APCs with T cells results in improved cluster formation and significantly increased T cell proliferative responses. The elevated expression of PD-L molecules on NKT-instructed APCs appears to result from exposure to extracellular ATP that is produced during NKT-monocyte interactions, and blocking purinergic signaling during monocyte differentiation results in APCs that form clusters with T cells and stimulate their proliferation. Finally, we show that human monocytes and NKT cells that are injected into immunodeficient mice co-localize together in spleen and liver, and after 3 days in vivo in the presence of NKT cells a fraction of the myeloid cells have upregulated markers associated with differentiation into professional APCs. These results suggest that autoreactive human NKT cells may promote tolerance by inducing the differentiation of regulatory myeloid APCs that limit T cell proliferation through expression of PD-L molecules.


Subject(s)
Antigen-Presenting Cells/cytology , Antigens, CD/immunology , Cell Differentiation , Gene Expression Regulation , Myeloid Cells/cytology , Natural Killer T-Cells/immunology , T-Lymphocytes , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/pharmacology , Animals , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, CD/metabolism , B7-1 Antigen/metabolism , B7-H1 Antigen , Cell Differentiation/immunology , Humans , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Myeloid Cells/drug effects , Myeloid Cells/immunology , Phenotype , Programmed Cell Death 1 Ligand 2 Protein , T-Lymphocytes/immunology
19.
J Virol ; 85(1): 165-77, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20980506

ABSTRACT

Epstein-Barr virus (EBV) infects cells in latent or lytic forms, but the role of lytic infection in EBV-induced lymphomas is unclear. Here, we have used a new humanized mouse model, in which both human fetal CD34(+) hematopoietic stem cells and thymus/liver tissue are transplanted, to compare EBV pathogenesis and lymphoma formation following infection with a lytic replication-defective BZLF1-deleted (Z-KO) virus or a lytically active BZLF1(+) control. Both the control and Z-KO viruses established long-term viral latency in all infected animals. The infection appeared well controlled in some animals, but others eventually developed CD20(+) diffuse large B cell lymphomas (DLBCL). Animals infected with the control virus developed tumors more frequently than Z-KO virus-infected animals. Specific immune responses against EBV-infected B cells were generated in mice infected with either the control virus or the Z-KO virus. In both cases, forms of viral latency (type I and type IIB) were observed that are less immunogenic than the highly transforming form (type III) commonly found in tumors of immunocompromised hosts, suggesting that immune pressure contributed to the outcome of the infection. These results point to an important role for lytic EBV infection in the development of B cell lymphomas in the context of an active host immune response.


Subject(s)
Epstein-Barr Virus Infections/immunology , Epstein-Barr Virus Infections/pathology , Herpesvirus 4, Human/pathogenicity , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/virology , Viral Proteins/metabolism , Animals , Antigens, CD34/metabolism , Cell Line , Disease Models, Animal , Epstein-Barr Virus Infections/virology , Hematopoietic Stem Cell Transplantation , Herpesvirus 4, Human/genetics , Humans , Liver Transplantation , Lymphoma, B-Cell/immunology , Mice , Mice, Knockout , T-Lymphocytes/immunology , Thymus Gland/transplantation , Trans-Activators/genetics , Transplantation, Heterologous , Viral Proteins/genetics , Virus Latency
20.
Proc Natl Acad Sci U S A ; 107(7): 3146-51, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133771

ABSTRACT

EBV causes infectious mononucleosis and is associated with certain malignancies. EBV nuclear antigen 1 (EBNA1) mediates EBV genome replication, partition, and transcription, and is essential for persistence of the viral genome in host cells. Here we demonstrate that Hsp90 inhibitors decrease EBNA1 expression and translation, and that this effect requires the Gly-Ala repeat domain of EBNA1. Hsp90 inhibitors induce the death of established, EBV-transformed lymphoblastoid cell lines at doses nontoxic to normal cells, and this effect is substantially reversed when lymphoblastoid cell lines are stably infected with a retrovirus expressing a functional EBNA1 mutant lacking the Gly-Ala repeats. Hsp90 inhibitors prevent EBV transformation of primary B cells, and strongly inhibit the growth of EBV-induced lymphoproliferative disease in SCID mice. These results suggest that Hsp90 inhibitors may be particularly effective for treating EBV-induced diseases requiring the continued presence of the viral genome.


Subject(s)
Benzoquinones/pharmacology , Epstein-Barr Virus Nuclear Antigens/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 4, Human , Lactams, Macrocyclic/pharmacology , Lymphoproliferative Disorders/drug therapy , Animals , Apoptosis/drug effects , Benzoquinones/therapeutic use , Cell Line, Tumor , DNA Primers/genetics , Dipeptides/genetics , Gene Expression Regulation, Viral/drug effects , Humans , Immunoblotting , Immunoprecipitation , Lactams, Macrocyclic/therapeutic use , Lymphoproliferative Disorders/virology , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Virus Replication/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...