Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Altern Complement Med ; 26(10): 884-901, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32749874

ABSTRACT

Objective: To develop an evidence-based clinical practice guideline (CPG) through a broad-based consensus process on best practices for chiropractic management of patients with chronic musculoskeletal (MSK) pain. Design: CPG based on evidence-based recommendations of a panel of experts in chronic MSK pain management. Methods: Using systematic reviews identified in an initial literature search, a steering committee of experts in research and management of patients with chronic MSK pain drafted a set of recommendations. Additional supportive literature was identified to supplement gaps in the evidence base. A multidisciplinary panel of experienced practitioners and educators rated the recommendations through a formal Delphi consensus process using the RAND Corporation/University of California, Los Angeles, methodology. Results: The Delphi process was conducted January-February 2020. The 62-member Delphi panel reached consensus on chiropractic management of five common chronic MSK pain conditions: low-back pain (LBP), neck pain, tension headache, osteoarthritis (knee and hip), and fibromyalgia. Recommendations were made for nonpharmacological treatments, including acupuncture, spinal manipulation/mobilization, and other manual therapy; modalities such as low-level laser and interferential current; exercise, including yoga; mind-body interventions, including mindfulness meditation and cognitive behavior therapy; and lifestyle modifications such as diet and tobacco cessation. Recommendations covered many aspects of the clinical encounter, from informed consent through diagnosis, assessment, treatment planning and implementation, and concurrent management and referral. Appropriate referral and comanagement were emphasized. Conclusions: These evidence-based recommendations for a variety of conservative treatment approaches to the management of common chronic MSK pain conditions may advance consistency of care, foster collaboration between provider groups, and thereby improve patient outcomes.


Subject(s)
Evidence-Based Practice/standards , Manipulation, Chiropractic/standards , Musculoskeletal Pain/therapy , Practice Guidelines as Topic , Chiropractic/standards , Consensus , Delphi Technique , Humans , Low Back Pain/therapy , Musculoskeletal Diseases/therapy , Neck Pain/therapy
2.
PLoS Pathog ; 16(2): e1008365, 2020 02.
Article in English | MEDLINE | ID: mdl-32059024

ABSTRACT

Humans are infected with two distinct strains (Type 1 (T1) and Type 2 (T2)) of Epstein-Barr virus (EBV) that differ substantially in their EBNA2 and EBNA 3A/B/C latency genes and the ability to transform B cells in vitro. While most T1 EBV strains contain the "prototype" form of the BZLF1 immediate-early promoter ("Zp-P"), all T2 strains contain the "Zp-V3" variant, which contains an NFAT binding motif and is activated much more strongly by B-cell receptor signalling. Whether B cells infected with T2 EBV are more lytic than cells infected with T1 EBV is unknown. Here we show that B cells infected with T2 EBV strains (AG876 and BL5) have much more lytic protein expression compared to B cells infected with T1 EBV strains (M81, Akata, and Mutu) in both a cord blood-humanized (CBH) mouse model and EBV-transformed lymphoblastoid cell lines (LCLs). Although T2 LCLs grow more slowly than T1 LCLs, both EBV types induce B-cell lymphomas in CBH mice. T1 EBV strains (M81 and Akata) containing Zp-V3 are less lytic than T2 EBV strains, suggesting that Zp-V3 is not sufficient to confer a lytic phenotype. Instead, we find that T2 LCLs express much higher levels of activated NFATc1 and NFATc2, and that cyclosporine (an NFAT inhibitor) and knockdown of NFATc2 attenuate constitutive lytic infection in T2 LCLs. Both NFATc1 and NFATc2 induce lytic EBV gene expression when combined with activated CAMKIV (which is activated by calcium signaling and activates MEF2D) in Burkitt Akata cells. Together, these results suggest that B cells infected with T2 EBV are more lytic due to increased activity of the cellular NFATc1/c2 transcription factors in addition to the universal presence of the Zp-V3 form of BZLF1 promoter.


Subject(s)
B-Lymphocytes/metabolism , NFATC Transcription Factors/genetics , Animals , B-Lymphocytes/virology , Cell Line , DNA-Binding Proteins/metabolism , Epstein-Barr Virus Infections/genetics , Epstein-Barr Virus Nuclear Antigens , Gene Expression/genetics , Gene Expression Regulation, Viral/genetics , Herpesvirus 4, Human/genetics , Herpesvirus 4, Human/metabolism , Herpesvirus 4, Human/pathogenicity , Humans , Mice , Promoter Regions, Genetic/genetics , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/metabolism , Viral Proteins/metabolism , Virus Activation , Virus Latency
3.
PLoS Pathog ; 14(7): e1007179, 2018 07.
Article in English | MEDLINE | ID: mdl-30052684

ABSTRACT

Latent Epstein-Barr virus (EBV) infection contributes to both B-cell and epithelial-cell malignancies. However, whether lytic EBV infection also contributes to tumors is unclear, although the association between malaria infection and Burkitt lymphomas (BLs) may involve excessive lytic EBV replication. A particular variant of the viral promoter (Zp) that controls lytic EBV reactivation is over-represented, relative to its frequency in non-malignant tissue, in EBV-positive nasopharyngeal carcinomas and AIDS-related lymphomas. To date, no functional differences between the prototype Zp (Zp-P) and the cancer-associated variant (Zp-V3) have been identified. Here we show that a single nucleotide difference between the Zp-V3 and Zp-P promoters creates a binding site for the cellular transcription factor, NFATc1, in the Zp-V3 (but not Zp-P) variant, and greatly enhances Zp activity and lytic viral reactivation in response to NFATc1-inducing stimuli such as B-cell receptor activation and ionomycin. Furthermore, we demonstrate that restoring this NFATc1-motif to the Zp-P variant in the context of the intact EBV B95.8 strain genome greatly enhances lytic viral reactivation in response to the NFATc1-activating agent, ionomycin, and this effect is blocked by the NFAT inhibitory agent, cyclosporine, as well as NFATc1 siRNA. We also show that the Zp-V3 variant is over-represented in EBV-positive BLs and gastric cancers, and in EBV-transformed B-cell lines derived from EBV-infected breast milk of Kenyan mothers that had malaria during pregnancy. These results demonstrate that the Zp-V3 enhances EBV lytic reactivation to physiologically-relevant stimuli, and suggest that increased lytic infection may contribute to the increased prevalence of this variant in EBV-associated malignancies.


Subject(s)
Epstein-Barr Virus Infections/genetics , Trans-Activators/genetics , Virus Activation/genetics , Genetic Variation/genetics , Herpesvirus 4, Human/genetics , Humans , Promoter Regions, Genetic/genetics
4.
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
5.
J Virol ; 91(8)2017 04 15.
Article in English | MEDLINE | ID: mdl-28179525

ABSTRACT

Epstein-Barr virus (EBV)-associated diseases of epithelial cells, including tumors that have latent infection, such as nasopharyngeal carcinoma (NPC), and oral hairy leukoplakia (OHL) lesions that have lytic infection, frequently express the viral latent membrane protein 1 (LMP1). In lytically infected cells, LMP1 expression is activated by the BRLF1 (R) immediate early (IE) protein. However, the mechanisms by which LMP1 expression is normally regulated in epithelial cells remain poorly understood, and its potential roles in regulating lytic reactivation in epithelial cells are as yet unexplored. We previously showed that the differentiation-dependent cellular transcription factors KLF4 and BLIMP1 induce lytic EBV reactivation in epithelial cells by synergistically activating the two EBV immediate early promoters (Zp and Rp). Here we show that epithelial cell differentiation also induces LMP1 expression. We demonstrate that KLF4 and BLIMP1 cooperatively induce the expression of LMP1, even in the absence of the EBV IE proteins BZLF1 (Z) and R, via activation of the two LMP1 promoters. Furthermore, we found that differentiation of NOKs-Akata cells by either methylcellulose suspension or organotypic culture induces LMP1 expression prior to Z and R expression. We show that LMP1 enhances the lytic infection-inducing effects of epithelial cell differentiation, as well as 12-O-tetradecanoylphorbol-13-acetate (TPA) and sodium butyrate treatment, in EBV-infected epithelial cells by increasing expression of the Z and R proteins. Our results suggest that differentiation of epithelial cells activates a feed-forward loop in which KLF4 and BLIMP1 first activate LMP1 expression and then cooperate with LMP1 to activate Z and R expression.IMPORTANCE The EBV protein LMP1 is expressed in EBV-associated epithelial cell diseases, regardless of whether these diseases are due to lytic infection (such as oral hairy leukoplakia) or latent infection (such as nasopharyngeal carcinoma). However, surprisingly little is known about how LMP1 expression is regulated in epithelial cells, and there are conflicting reports about whether it plays any role in regulating viral lytic reactivation. In this study, we show that epithelial cell differentiation induces LMP1 expression by increasing expression of two cellular transcription factors (KLF4 and BLIMP1) which cooperatively activate the two LMP1 promoters. We also demonstrate that LMP1 promotes efficient lytic reactivation in EBV-infected epithelial cells by enhancing expression of the Z and R proteins. Thus, in EBV-infected epithelial cells, LMP1 expression is promoted by differentiation and positively regulates lytic viral reactivation.


Subject(s)
Cell Differentiation , Epithelial Cells/physiology , Epithelial Cells/virology , Herpesvirus 4, Human/physiology , Host-Pathogen Interactions , Viral Matrix Proteins/metabolism , Virus Activation , Gene Expression Regulation, Viral , Herpesvirus 4, Human/genetics , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/metabolism , Positive Regulatory Domain I-Binding Factor 1 , Repressor Proteins/metabolism
6.
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
7.
J Virol ; 87(18): 10126-38, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23843639

ABSTRACT

All eight human herpesviruses have a conserved herpesvirus protein kinase (CHPK) that is important for the lytic phase of the viral life cycle. In this study, we show that heat shock protein 90 (Hsp90) interacts directly with each of the eight CHPKs, and we demonstrate that an Hsp90 inhibitor drug, 17-dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG), decreases expression of all eight CHPKs in transfected HeLa cells. 17-DMAG also decreases expression the of the endogenous Epstein-Barr virus protein kinase (EBV PK, encoded by the BGLF4 gene) in lytically infected EBV-positive cells and inhibits phosphorylation of several different known EBV PK target proteins. Furthermore, 17-DMAG treatment abrogates expression of the human cytomegalovirus (HCMV) kinase UL97 in HCMV-infected human fibroblasts. Importantly, 17-DMAG treatment decreased the EBV titer approximately 100-fold in lytically infected AGS-Akata cells without causing significant cellular toxicity during the same time frame. Increased EBV PK expression in 17-DMAG-treated AGS-Akata cells did not restore EBV titers, suggesting that 17-DMAG simultaneously targets multiple viral and/or cellular proteins required for efficient viral replication. These results suggest that Hsp90 inhibitors, including 17-DMAG, may be a promising group of drugs that could have profound antiviral effects on herpesviruses.


Subject(s)
Antiviral Agents/metabolism , Benzoquinones/metabolism , Enzyme Inhibitors/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesvirus 4, Human/physiology , Lactams, Macrocyclic/metabolism , Protein Kinases/metabolism , Virus Replication/drug effects , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/virology , Fibroblasts/drug effects , Fibroblasts/virology , Herpesvirus 4, Human/drug effects , Humans , Protein Interaction Mapping , Viral Load , Virus Cultivation
8.
J Virol ; 86(24): 13360-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23015717

ABSTRACT

The Epstein-Barr virus (EBV) latent-to-lytic switch is mediated by the viral proteins BZLF1 (Z), BRLF1 (R), and BRRF1 (Na). Since we previously showed that DNA-damaging agents (including chemotherapy and irradiation) can induce EBV lytic reactivation and recently demonstrated that wild-type p53 contributes to lytic reactivation, we investigated the role of the ATM kinase during EBV reactivation. ATM phosphorylates and activates p53, as well as numerous other substrates involved in the cellular DNA damage response. Using an ATM inhibitor (KU55933), we found that ATM activity is required for efficient induction of EBV lytic gene expression by a variety of different stimuli, including a histone deacetylase (HDAC) inhibitor, the transforming growth factor ß (TGF-ß) cytokine, a demethylating agent (5-azacytidine), B cell receptor engagement with anti-IgG antibody, hydrogen peroxide, and the proteosome inhibitor bortezomib. In EBV-infected AGS (gastric) cells, knockdown of ATM, or p53, expression inhibits EBV reactivation. Conversely, treatment of these cells with nutlin-3 (which activates p53 and ATM) robustly induces lytic reactivation in a p53- and ATM-dependent manner. The ability of the EBV R and Na proteins to induce lytic reactivation in EBV-infected AGS cells is ATM dependent. However, overexpression of Z induces lytic gene expression in the presence or absence of ATM activity. Our results suggest that ATM enhances Z promoter activity in the context of the intact EBV genome and that p53 contributes to the ATM effect. Nevertheless, since we found that ATM inhibitors also reduce lytic reactivation in Burkitt lymphoma cells that have no p53, additional ATM substrates must also contribute to the ATM effect.


Subject(s)
Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Herpesvirus 4, Human/physiology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Proteins/metabolism , Virus Activation , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line , DNA Primers , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/genetics , Humans , Imidazoles/metabolism , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Phosphoinositide-3 Kinase Inhibitors , Piperazines/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Pyrones/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/antagonists & inhibitors , Tumor Suppressor Proteins/genetics
9.
J Virol ; 85(9): 4318-29, 2011 May.
Article in English | MEDLINE | ID: mdl-21325409

ABSTRACT

The Epstein-Barr virus (EBV) BRRF1 lytic gene product (Na) is encoded within the same immediate-early region as the BZLF1 (Z) and BRLF1(R) gene products, but its role during EBV infection has not been well defined. We previously showed that Na cooperates with the R protein to induce lytic gene expression in latently infected EBV-positive 293 cells, and in some EBV-negative cell lines it can activate the Z promoter in reporter gene assays. Here we show that overexpression of Na alone is sufficient to induce lytic gene expression in several different latently infected epithelial cell lines (Hone-Akata, CNE2-Akata, and AGS-Akata), while knockdown of endogenous Na expression reduces lytic gene expression. Consistent with its ability to interact with tumor necrosis factor receptor-associated factor 2 (TRAF2) in a yeast two-hybrid assay, we demonstrate that Na interacts with TRAF2 in cells. Furthermore, we show that TRAF2 is required for Na induction of lytic gene expression, that Na induces Jun N-terminal protein kinase (JNK) activation in a TRAF2-dependent manner, and that a JNK inhibitor abolishes the ability of Na to disrupt viral latency. Additionally, we show that Na and the tumor suppressor protein p53 cooperate to induce lytic gene expression in epithelial cells (including the C666-1 nasopharyngeal carcinoma cell line), although Na does not appear to affect p53 function. Together these data suggest that Na plays an important role in regulating the switch between latent and lytic infection in epithelial cells and that this effect requires both the TRAF2 and p53 cellular proteins.


Subject(s)
Herpesvirus 4, Human/growth & development , Herpesvirus 4, Human/pathogenicity , Protein Interaction Mapping , TNF Receptor-Associated Factor 2/metabolism , Tumor Suppressor Protein p53/metabolism , Cell Line , Cell Survival , Humans , Protein Binding
10.
J Virol ; 84(23): 12362-74, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20861254

ABSTRACT

The Epstein-Barr virus immediate-early protein, BZLF1 (Z), initiates the switch between latent and lytic infection and plays an essential role in mediating viral replication. Z also inhibits expression of the major receptor for tumor necrosis factor (TNF), TNFR1, thus repressing TNF cytokine signaling, but the mechanism for this effect is unknown. Here, we demonstrate that Z prevents both C/EBPα- and C/EBPß-mediated activation of the TNFR1 promoter (TNFR1p) by interacting directly with both C/EBP family members. We show that Z interacts directly with C/EBPα and C/EBPß in vivo and that a Z mutant altered at alanine residue 204 in the bZIP domain is impaired for the ability to interact with both C/EBP proteins. Furthermore, we find that the Z(A204D) mutant is attenuated in the ability to inhibit the TNFR1p but mediates lytic viral reactivation and replication in vitro in 293 cells as well as wild-type Z. Although Z does not bind directly to the TNFR1p in EMSA studies, chromatin immunoprecipitation studies indicate that Z is complexed with this promoter in vivo. The Z(A204D) mutant has reduced interaction with the TNFR1p in vivo but is similar to wild-type Z in its ability to complex with the IL-8 promoter. Finally, we show that the effect of Z on C/EBPα- and C/EBPß-mediated activation is promoter dependent. These results indicate that Z modulates the effects of C/EBPα and C/EBPß in a promoter-specific manner and that in some cases (including that of the TNFR1p), Z inhibits C/EBPα- and C/EBPß-mediated activation.


Subject(s)
CCAAT-Enhancer-Binding Protein-alpha/metabolism , CCAAT-Enhancer-Binding Protein-beta/metabolism , Promoter Regions, Genetic/genetics , Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors , Trans-Activators/metabolism , Virus Activation/physiology , Virus Replication/physiology , Chromatin Immunoprecipitation , Electrophoretic Mobility Shift Assay , HeLa Cells , Herpesvirus 4, Human , Humans , Immunoblotting , Immunoprecipitation , Mutagenesis , Receptors, Tumor Necrosis Factor, Type I/genetics , Trans-Activators/genetics , Trans-Activators/physiology
11.
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
12.
Int J Cancer ; 121(6): 1274-81, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17520680

ABSTRACT

Lytically infected EBV-positive lymphoblastoid cells enhance the growth of early-passage, but not late-passage, EBV-immortalized lymphoblastoid cell lines (LCLs) in SCID mice and have enhanced IL-6 secretion. Here, we have examined the importance of IL-6 for the growth of early-passage LCLs (EPL) in SCID mice, identified lytic EBV proteins that activate IL-6 production and compared viral and cellular differences between early versus late passage LCLs (LPL). IL-6 was required for efficient growth of EPL in SCID mice. The EBV immediate-early (IE) proteins, BRLF1 and BZLF1, each induced IL-6 secretion when transfected into 293 and BJAB cells. Interestingly, the combination of BZLF1 and the latent EBV protein, LMP-1, induced much more IL-6 expression in both 293 and BJAB cells than either protein alone. Both BZLF1 and BRLF1 also enhanced IL-10 production in 293 cells. In comparison to the EPL, LPL had much reduced expression of early lytic viral proteins and cellular IL-6. In contrast, expression of cellular IL-10 was similar in EPL versus LPL, while VEGF secretion was increased in late-passage LCLs. These results suggest that both BRLF1 and BZLF1 contribute to IL-6 secretion in lytically infected cells and that lytically infected cells may promote early lymphoproliferative disease in patients through enhanced IL-6 production.


Subject(s)
Epstein-Barr Virus Infections/metabolism , Interleukin-6/metabolism , Lymphoproliferative Disorders/virology , Tumor Virus Infections/virology , Animals , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immediate-Early Proteins/metabolism , Immunoblotting , Interleukin-10/metabolism , Lymphoproliferative Disorders/metabolism , Mice , Mice, SCID , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trans-Activators/metabolism , Tumor Virus Infections/metabolism , Viral Matrix Proteins/metabolism , Viral Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...