Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Sci Rep ; 11(1): 10002, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33976304

ABSTRACT

Cytotoxicity assays are essential for the testing and development of novel immunotherapies for the treatment of cancer. We recently described a novel cytotoxicity assay, termed the Matador assay, which was based on marine luciferases and their engineered derivatives. In this study, we describe the development of a new cytotoxicity assay termed 'Matador-Glo assay' which takes advantage of a thermostable variant of Click Beetle Luciferase (Luc146-1H2). Matador-Glo assay utilizes Luc146-1H2 and D-luciferin as the luciferase-substrate pair for luminescence detection. The assay involves ectopic over-expression of Luc146-1H2 in the cytosol of target cells of interest. Upon damage to the membrane integrity, the Luc146-1H2 is either released from the dead and dying cells or its activity is preferentially measured in dead and dying cells. We demonstrate that this assay is simple, fast, specific, sensitive, cost-efficient, and not labor-intensive. We further demonstrate that the Matador-Glo assay can be combined with the marine luciferase-based Matador assay to develop a dual luciferase assay for cell death detection. Finally, we demonstrate that the Luc146-1H2 expressing target cells can also be used for in vivo bioluminescence imaging applications.


Subject(s)
Benzothiazoles , Coleoptera/enzymology , Cytotoxicity Tests, Immunologic , Luciferases , Animals , Humans , K562 Cells , Mice, Inbred NOD , Mice, SCID
2.
Sci Rep ; 10(1): 5712, 2020 03 31.
Article in English | MEDLINE | ID: mdl-32235878

ABSTRACT

Primary effusion lymphoma (PEL) is a subtype of non-Hodgkin lymphoma associated with infection by Kaposi sarcoma-associated herpes virus (KSHV). PEL is an aggressive disease with extremely poor prognosis when treated with conventional chemotherapy. Narciclasine, a natural product present in Amaryllidaceae family of flowering plants including daffodils, belongs to a class of molecules termed 'isocarbostyril alkaloid'. We have found that narciclasine displays preferential cytotoxicity towards PEL at low nanomolar concentrations and is approximately 10 and 100-fold more potent than its structural analogs lycoricidine and lycorine, respectively. Narciclasine arrested cell-cycle progression at the G1 phase and induced apoptosis in PEL, which is accompanied by activation of caspase-3/7, cleavage of PARP and increase in the surface expression of Annexin-V. Although narciclasine treatment resulted in a marked decrease in the expression of MYC and its direct target genes,time-course experiments revealed that MYC is not a direct target of narciclasine. Narciclasine treatment neither induces the expression of KSHV-RTA/ORF50 nor the production of infectious KSHV virions in PEL. Finally, narciclasine provides dramatic survival advantages to mice in two distinct mouse xenograft models of PEL. In conclusion, our results suggest that narciclasine could be a promising agent for the treatment of PEL.


Subject(s)
Amaryllidaceae Alkaloids/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Lymphoma, Primary Effusion/drug therapy , Phenanthridines/pharmacology , Plant Extracts/pharmacology , Amaryllidaceae Alkaloids/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Body Weight/drug effects , Cell Line, Tumor , Humans , Lymphoma, Primary Effusion/pathology , Mice , Phenanthridines/therapeutic use , Plant Extracts/therapeutic use , Xenograft Model Antitumor Assays
3.
Sci Rep ; 10(1): 2318, 2020 02 11.
Article in English | MEDLINE | ID: mdl-32047180

ABSTRACT

Success of immunotherapeutic approaches using genetically engineered antibodies and T cells modified with chimeric antigen receptors (CARs) depends, among other things, on the selection of antigen binding domains with desirable expression and binding characteristics. We developed a luciferase-based assay, termed Malibu-Glo Assay, which streamlines the process of optimization of an antigen binding domain with desirable properties and allows the sensitive detection of tumor antigens. The assay involves a recombinant immunoconjugate, termed Malibu-Glo reagent, comprising an immunoglobulin or a non-immunoglobulin based antigen binding domain genetically linked to a marine luciferase. Malibu-Glo reagent can be conveniently produced in mammalian cells as a secreted protein that retains the functional activity of both the antigen binding domain and the luciferase. Moreover, crude supernatant containing the secreted Malibu-Glo reagent can directly be used for detection of cell surface antigens obviating the laborious steps of protein purification and labeling. We further demonstrate the utility of Malibu-Glo assay for the selection of optimal single chain fragment variables (scFvs) with desired affinity characteristics for incorporation into CARs. In summary, Malibu-Glo assay is a fast, simple, sensitive, specific and economical assay for antigen detection with multiple applications in the fields of antibody engineering, antibody humanization and CAR-T cell therapy.


Subject(s)
Aquatic Organisms/enzymology , Genetic Engineering/methods , Luciferases/metabolism , Receptors, Antigen, T-Cell/immunology , Receptors, Chimeric Antigen/immunology , Recombinant Fusion Proteins/immunology , Single-Chain Antibodies/immunology , Animals , Humans , Luciferases/genetics , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , Recombinant Fusion Proteins/genetics , Single-Chain Antibodies/genetics
4.
Sci Rep ; 9(1): 1957, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760795

ABSTRACT

Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy has produced dramatic responses in hematologic malignancies. One of the challenges in the field is the lack of a simple assay for the detection of CARs on the surface of immune effector cells. In this study, we describe a novel luciferase-based assay, termed Topanga Assay, for the detection of CAR expression. The assay utilizes a recombinant fusion protein, called Topanga reagent, generated by joining the extra-cellular domain of a CAR-target in frame with one of the marine luciferases or their engineered derivatives. The assay involves incubation of CAR expressing cells with the Topanga reagent, a few washes and measurement of luminescence. The assay can detect CARs comprising either immunoglobulin- or non-immunoglobulin-based antigen binding domains. We further demonstrate that addition of epitope tags to the Topanga reagent not only allows its convenient one step purification but also extends its use for detection of CAR cells using flow cytometry. However, crude supernatant containing the secreted Topanga reagent can be directly used in both luminescence and flow-cytometry based assays without prior protein purification. Our results demonstrate that the Topanga assay is a highly sensitive, specific, convenient, economical and versatile assay for the detection of CARs.


Subject(s)
Immunotherapy, Adoptive/methods , Luciferases/metabolism , Receptors, Chimeric Antigen/analysis , Cell Line , Flow Cytometry/methods , Humans , Lymphocytes/metabolism , Receptors, Antigen/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , Recombinant Fusion Proteins/metabolism , T-Lymphocytes/immunology
5.
Sci Rep ; 8(1): 199, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29317736

ABSTRACT

A simple, accurate, sensitive and robust assay that can rapidly and specifically measure the death of target cells would have applications in many areas of biomedicine and particularly for the development of novel cellular- and immune-therapeutics. In this study, we describe a novel cytotoxicity assay, termed the Matador assay, which takes advantage of the extreme brightness, stability and glow-like characteristics of recently discovered novel marine luciferases and their engineered derivatives. The assay involves expression of a luciferase of interest in target cells in a manner so that it is preferentially retained within the healthy cells but is either released from dead and dying cells or whose activity can be preferentially measured in dead and dying cells. We demonstrate that this assay is highly sensitive, specific, rapid, and can be performed in a single-step manner without the need for any expensive equipment. We further validate this assay by demonstrating its ability to detect cytotoxicity induced by several cellular and immune-therapeutic agents including antibodies, natural killer cells, chimeric antigen receptor expressing T cells and a bispecific T cell engager.


Subject(s)
Luciferases/metabolism , Toxicity Tests/methods , Cell Line, Tumor , Cells, Cultured , HEK293 Cells , Humans , Luciferases/genetics
6.
J Virol ; 88(11): 6345-54, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24672029

ABSTRACT

UNLABELLED: Kaposi's sarcoma-associated herpesvirus-encoded viral FLICE inhibitory protein (vFLIP) K13 was originally believed to protect virally infected cells against death receptor-induced apoptosis by interfering with caspase 8/FLICE activation. Subsequent studies revealed that K13 also activates the NF-κB pathway by binding to the NEMO/inhibitor of NF-κB (IκB) kinase gamma (IKKγ) subunit of an IKK complex and uses this pathway to modulate the expression of genes involved in cellular survival, proliferation, and the inflammatory response. However, it is not clear if K13 can also induce gene expression independently of NEMO/IKKγ. The minimum region of NEMO that is sufficient for supporting K13-induced NF-κB has not been delineated. Furthermore, the contribution of NEMO and NF-κB to the protective effect of K13 against death receptor-induced apoptosis remains to be determined. In this study, we used microarray analysis on K13-expressing wild-type and NEMO-deficient cells to demonstrate that NEMO is required for modulation of K13-induced genes. Reconstitution of NEMO-null cells revealed that the N-terminal 251 amino acid residues of NEMO are sufficient for supporting K13-induced NF-κB but fail to support tumor necrosis factor alpha (TNF-α)-induced NF-κB. K13 failed to protect NEMO-null cells against TNF-α-induced cell death but protected those reconstituted with the NEMO mutant truncated to include only the N-terminal 251 amino acid residues [the NEMO(1-251) mutant]. Taken collectively, our results demonstrate that NEMO is required for modulation of K13-induced genes and the N-terminal 251 amino acids of NEMO are sufficient for supporting K13-induced NF-κB. Finally, the ability of K13 to protect against TNF-α-induced cell death is critically dependent on its ability to interact with NEMO and activate NF-κB. IMPORTANCE: Kaposi's sarcoma-associated herpesvirus-encoded vFLIP K13 is believed to protect virally infected cells against death receptor-induced apoptosis and to activate the NF-κB pathway by binding to adaptor protein NEMO/IKKγ. However, whether K13 can also induce gene expression independently of NEMO and the minimum region of NEMO that is sufficient for supporting K13-induced NF-κB remain to be delineated. Furthermore, the contribution of NEMO and NF-κB to the protective effect of K13 against death receptor-induced apoptosis is not clear. We demonstrate that NEMO is required for modulation of K13-induced genes and its N-terminal 251 amino acids are sufficient for supporting K13-induced NF-κB. The ability of K13 to protect against TNF-α-induced cell death is critically dependent on its ability to interact with NEMO and activate NF-κB. Our results suggest that K13-based gene therapy approaches may have utility for the treatment of patients with NEMO mutations and immunodeficiency.


Subject(s)
Apoptosis/genetics , Gene Expression Regulation, Viral/genetics , I-kappa B Kinase/metabolism , Receptors, Death Domain/metabolism , Viral Proteins/metabolism , Animals , Blotting, Western , DNA Primers/genetics , Fibroblasts , HEK293 Cells , Humans , Jurkat Cells , Luciferases , Mice , Microarray Analysis , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
7.
Clin Cancer Res ; 19(18): 5016-26, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23881928

ABSTRACT

PURPOSE: Kaposi sarcoma-associated herpes virus (KSHV)-associated primary effusion lymphomas (PEL) have extremely poor prognosis when treated with conventional chemotherapy. KSHV-encoded viral FLICE-inhibitory protein (vFLIP) K13 binds to the IkappaB kinase (IKK) complex to constitutively activate the NF-κB pathway, which has been shown to be essential for the survival and proliferation of PEL cells. The molecular chaperone HSP90 is a component of the IKK complex and is required for its activity. EXPERIMENTAL DESIGN: We have analyzed the effect of HSP90 inhibitors on the survival and proliferation of PEL cells and on the activity of the NF-κB pathway. RESULTS: We show that BIIB021, a purine scaffold-based orally administrable HSP90 inhibitor, shows preferential cytotoxicity toward PEL cells as compared with non-PEL cells. The cytotoxic effect of BIIB021 against PEL was associated with induction of cell-cycle arrest and apoptosis. BIIB021 blocked the expression of a number of cellular proteins involved in the regulation of cell cycle and apoptosis. BIIB021 also blocked constitutive NF-κB activity present in PEL cells, in part, by blocking the interaction of vFLIP K13 with the IKK complex subunits. In a xenograft model of PEL, BIIB021 significantly reduced tumor growth. CONCLUSION: BIIB021 blocks constitutive NF-κB activity in PEL and shows preferential antitumor activity against PEL in vitro and in vivo. BIIB021 may be a promising agent for treatment of PEL.


Subject(s)
Adenine/analogs & derivatives , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Herpesviridae Infections/drug therapy , Herpesvirus 8, Human/pathogenicity , Lymphoma, Primary Effusion/drug therapy , NF-kappa B/antagonists & inhibitors , Pyridines/pharmacology , Viral Proteins/antagonists & inhibitors , Adenine/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Cycle/drug effects , Cell Proliferation/drug effects , Herpesviridae Infections/complications , Herpesviridae Infections/metabolism , Humans , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Mice , Mice, Nude , NF-kappa B/metabolism , Poly(ADP-ribose) Polymerases/metabolism , Purines/chemistry , Tumor Cells, Cultured , Viral Proteins/metabolism , Xenograft Model Antitumor Assays
8.
J Virol ; 87(4): 2242-52, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23236068

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) has been linked to the development of Kaposi's sarcoma, primary effusion lymphoma, and multicentric Castleman's disease (MCD). We have characterized the role of KSHV-encoded viral FLICE inhibitory protein (vFLIP) K13 in the modulation of anti-IgM-induced growth arrest and apoptosis in B cells. We demonstrate that K13 protects WEHI 231, an immature B-cell line, against anti-IgM-induced growth arrest and apoptosis. The protective effect of K13 was associated with the activation of the NF-κB pathway and was deficient in a mutant K13 with three alanine substitutions at positions 58 to 60 (K13-58AAA) and a structural homolog, vFLIP E8, both of which lack NF-κB activity. K13 upregulated the expression of NF-κB subunit RelB and blocked the anti-IgM-induced decline in c-Myc and rise in p27(Kip1) that have been associated with growth arrest and apoptosis. K13 also upregulated the expression of Mcl-1, an antiapoptotic member of the Bcl2 family. Finally, K13 protected the mature B-cell line Ramos against anti-IgM-induced apoptosis through NF-κB activation. Inhibition of anti-IgM-induced apoptosis by K13 may contribute to the development of KSHV-associated lymphoproliferative disorders.


Subject(s)
Apoptosis , B-Lymphocytes/immunology , B-Lymphocytes/virology , Herpesvirus 8, Human/pathogenicity , Host-Pathogen Interactions , Transcription Factor RelB/metabolism , Viral Proteins/metabolism , Animals , Cell Line , Mice , Myeloid Cell Leukemia Sequence 1 Protein , NF-kappa B/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism
9.
PLoS One ; 7(5): e36601, 2012.
Article in English | MEDLINE | ID: mdl-22590573

ABSTRACT

BACKGROUND: Kaposi's sarcoma associated herpesvirus encoded viral FLICE inhibitory protein (vFLIP) K13 activates the NF-κB pathway by binding to the NEMO/IKKγ subunit of the IκB kinase (IKK) complex. However, it has remained enigmatic how K13-NEMO interaction results in the activation of the IKK complex. Recent studies have implicated TRAF6, TAK1 and linear ubiquitin chains assembled by a linear ubiquitin chain assembly complex (LUBAC) consisting of HOIL-1, HOIP and SHARPIN in IKK activation by proinflammatory cytokines. METHODOLOGY/PRINCIPAL FINDINGS: Here we demonstrate that K13-induced NF-κB DNA binding and transcriptional activities are not impaired in cells derived from mice with targeted disruption of TRAF6, TAK1 and HOIL-1 genes and in cells derived from mice with chronic proliferative dermatitis (cpdm), which have mutation in the Sharpin gene (Sharpin(cpdm/cpdm)). Furthermore, reconstitution of NEMO-deficient murine embryonic fibroblast cells with NEMO mutants that are incapable of binding to linear ubiquitin chains supported K13-induced NF-κB activity. K13-induced NF-κB activity was not blocked by CYLD, a deubiquitylating enzyme that can cleave linear and Lys63-linked ubiquitin chains. On the other hand, NEMO was required for interaction of K13 with IKK1/IKKα and IKK2/IKKß, which resulted in their activation by "T Loop" phosphorylation. CONCLUSIONS/SIGNIFICANCE: Our results demonstrate that K13 activates the NF-κB pathway by binding to NEMO which results in the recruitment of IKK1/IKKα and IKK2/IKKß and their subsequent activation by phosphorylation. Thus, K13 activates NF-κB via a mechanism distinct from that utilized by inflammatory cytokines. These results have important implications for the development of therapeutic agents targeting K13-induced NF-κB for the treatment of KSHV-associated malignancies.


Subject(s)
Herpesvirus 8, Human/metabolism , MAP Kinase Kinase Kinases/metabolism , Multiprotein Complexes/metabolism , NF-kappa B/metabolism , Sarcoma, Kaposi/metabolism , TNF Receptor-Associated Factor 6/metabolism , Viral Proteins/metabolism , Animals , Chronic Disease , Dermatitis/genetics , Dermatitis/metabolism , Dermatitis/pathology , Embryo, Mammalian/metabolism , Embryo, Mammalian/pathology , Fibroblasts/metabolism , Fibroblasts/pathology , Herpesvirus 8, Human/genetics , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Jurkat Cells , MAP Kinase Kinase Kinases/genetics , Mice , Mice, Knockout , Multiprotein Complexes/genetics , NF-kappa B/genetics , Sarcoma, Kaposi/genetics , Sarcoma, Kaposi/pathology , TNF Receptor-Associated Factor 6/genetics , Viral Proteins/genetics
10.
PLoS One ; 7(5): e37498, 2012.
Article in English | MEDLINE | ID: mdl-22624040

ABSTRACT

Infection with Kaposi's sarcoma associated herpesvirus (KSHV) has been linked to the development of primary effusion lymphoma (PEL), a rare lymphoproliferative disorder that is characterized by loss of expression of most B cell markers and effusions in the body cavities. This unique clinical presentation of PEL has been attributed to their distinctive plasmablastic gene expression profile that shows overexpression of genes involved in inflammation, adhesion and invasion. KSHV-encoded latent protein vFLIP K13 has been previously shown to promote the survival and proliferation of PEL cells. In this study, we employed gene array analysis to characterize the effect of K13 on global gene expression in PEL-derived BCBL1 cells, which express negligible K13 endogenously. We demonstrate that K13 upregulates the expression of a number of NF-κB responsive genes involved in cytokine signaling, cell death, adhesion, inflammation and immune response, including two NF-κB subunits involved in the alternate NF-κB pathway, RELB and NFKB2. In contrast, CD19, a B cell marker, was one of the genes downregulated by K13. A comparison with K13-induced genes in human vascular endothelial cells revealed that although there was a considerable overlap among the genes induced by K13 in the two cell types, chemokines genes were preferentially induced in HUVEC with few exceptions, such as RANTES/CCL5, which was induced in both cell types. Functional studies confirmed that K13 activated the RANTES/CCL5 promoter through the NF-κB pathway. Taken collectively, our results suggest that K13 may contribute to the unique gene expression profile, immunophenotype and clinical presentation that are characteristics of KSHV-associated PEL.


Subject(s)
Gene Expression Regulation, Neoplastic/physiology , Herpesviridae Infections/complications , Herpesviridae Infections/metabolism , Herpesvirus 8, Human/metabolism , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/virology , Viral Proteins/metabolism , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Humans , Luciferases , Lymphoma, Primary Effusion/etiology , Lymphoma, Primary Effusion/genetics , Microarray Analysis , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Viral Proteins/genetics
11.
J Biol Chem ; 286(32): 27988-97, 2011 Aug 12.
Article in English | MEDLINE | ID: mdl-21705340

ABSTRACT

Myeloma cells are dependent on IL6 for their survival and proliferation during the early stages of disease, and independence from IL6 is associated with disease progression. The role of the NF-κB pathway in the IL6-independent growth of myeloma cells has not been studied. Because human herpesvirus 8-encoded K13 selectively activates the NF-κB pathway, we have used it as a molecular tool to examine the ability of the NF-κB pathway to confer IL6 independence on murine plasmacytomas. We demonstrated that ectopic expression of K13, but not its NF-κB-defective mutant or a structural homolog, protected plasmacytomas against IL6 withdrawal-induced apoptosis and resulted in emergence of IL6-independent clones that could proliferate long-term in vitro in the absence of IL6 and form abdominal plasmacytomas with visceral involvement when injected intraperitoneally into syngeneic mice. These IL6-independent clones were dependent on NF-κB activity for their survival and proliferation but were resistant to dexamethasone and INCB018424, a selective Janus kinase 1/2 inhibitor. Ectopic expression of human T cell leukemia virus 1-encoded Tax protein, which resembles K13 in inducing constitutive NF-κB activation, similarly protected plasmacytoma cells against IL6 withdrawal-induced apoptosis. Although K13 is known to up-regulate IL6 gene expression, its protective effect was not due to induction of endogenous IL6 production but instead was associated with sustained expression of several antiapoptotic members of the Bcl2 family upon IL6 withdrawal. Collectively, these results demonstrate that NF-κB activation cannot only promote the emergence of IL6 independence during myeloma progression but can also confer resistance to dexamethasone and INCB018424.


Subject(s)
Antineoplastic Agents, Hormonal/pharmacology , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/drug effects , Interleukin-6/metabolism , MAP Kinase Kinase 7/antagonists & inhibitors , Mitogen-Activated Protein Kinase 8/antagonists & inhibitors , NF-kappa B/metabolism , Plasmacytoma/metabolism , Pyrazoles/pharmacology , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , HEK293 Cells , Humans , Interleukin-6/genetics , Interleukin-6/pharmacology , MAP Kinase Kinase 7/genetics , MAP Kinase Kinase 7/metabolism , Mice , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Mutation , NF-kappa B/genetics , Neoplasm Transplantation , Nitriles , Plasmacytoma/drug therapy , Plasmacytoma/genetics , Plasmacytoma/pathology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Pyrimidines , Transplantation, Isogeneic
12.
J Biol Chem ; 286(24): 21555-64, 2011 Jun 17.
Article in English | MEDLINE | ID: mdl-21531730

ABSTRACT

Expression of A20, a negative regulator of the NF-κB pathway, is frequently lost in several subtypes of Hodgkin and non-Hodgkin lymphoma. We report that A20 is expressed in Kaposi sarcoma-associated herpesvirus (KSHV)-infected primary effusion lymphoma cell lines, and its expression correlates closely with the expression of KSHV-encoded viral FLICE inhibitory protein K13. Ectopic expression of K13 induced A20 expression through NF-κB-mediated activation of A20 promoter. In turn, A20 blocked K13-induced NF-κB activity and up-regulation of proinflammatory cytokines CCL20 and IL-8 in a negative feedback fashion. Both the N-terminal deubiquitinating domain and the C-terminal zinc finger domain of A20 were involved in the inhibition of K13-induced NF-κB activity. Overexpression of A20 blocked K13-induced IκBα phosphorylation, NF-κB nuclear translocation, and cellular transformation. Consistent with the above, K13-induced IκBα phosphorylation and NF-κB transcriptional activation were enhanced in A20-deficient cells. Finally, A20 was found to interact physically with K13. Taken collectively, these results demonstrate that K13 is a key determinant of A20 expression in KSHV-infected cells, and A20 is a key negative regulator of K13-induced NF-κB activity. A20 might serve to control the inflammatory response to KSHV infection and protect KSHV-infected cells from apoptosis.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Gene Expression Regulation , Herpesvirus 8, Human/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , Viral Proteins/metabolism , Apoptosis , Cell Line , Chemokine CCL20/metabolism , DNA-Binding Proteins , Humans , Inflammation , Interleukin-8/metabolism , Phosphorylation , Protein Structure, Tertiary , Signal Transduction , Tumor Necrosis Factor alpha-Induced Protein 3
13.
Cancer Biol Ther ; 10(10): 1033-40, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20818173

ABSTRACT

Primary effusion lymphoma (PEL) is an aggressive form of lymphoma that is associated with infection by Kaposi's sarcoma-associated herpesvirus (KSHV). One of the KSHV genes expressed in PEL cells is K13, a potent activator of the NF-κB pathway. K13 transgenic mice develop lymphomas, but after a long period of latency. A possible candidate that could cooperate with K13 in the development of PEL is c-Myc, whose expression is frequently dysregulated in PEL cells. To study the cooperative interaction between K13 and c-Myc in the pathogenesis of PEL, we crossed the K13 transgenic mice to iMyc(Eµ) transgenic mice that overexpress Myc. We report that lymphomas in the K13/iMyc(Eµ) double transgenic mice developed with shorter latency and were histologically distinct from those observed in the iMyc(Eµ) mice. Lymphomas in the K13/iMyc(Eµ) mice also lacked the expression of B- and T-cell markers, thus resembling the immunophenotype of PEL. The accelerated development of lymphoma in the K13/iMyc(Eµ) mice was associated with increased expression of K13, elevated NF-κB activity and decrease in apoptosis. Taken collectively, our results demonstrate a cooperative interaction between the NF-κB and Myc pathways in lymphomagenesis.


Subject(s)
Apoptosis , Herpesvirus 8, Human/genetics , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Proto-Oncogene Proteins c-myc/physiology , Viral Proteins/physiology , Animals , Blotting, Western , Electrophoretic Mobility Shift Assay , Female , Flow Cytometry , Immunoenzyme Techniques , Lymphoma, Primary Effusion/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transfection
14.
Clin Cancer Res ; 16(4): 1140-8, 2010 Feb 15.
Article in English | MEDLINE | ID: mdl-20145163

ABSTRACT

PURPOSE: The X-linked ectodermal dysplasia receptor (XEDAR) is a novel receptor of the tumor necrosis factor receptor family that binds to ectodysplasin-A2 (EDA-A2) and induces cell death. The purpose of this study was to determine the tumor-suppressive potential of XEDAR in the development of breast cancer. EXPERIMENTAL DESIGN: We analyzed the expression of XEDAR in breast cancer cell lines and tumor samples using quantitative real-time PCR analysis and immunoblotting. We analyzed the human XEDAR gene promoter for the presence of any CpG island and examined its methylation status using methylation-specific real-time PCR. We examined the effect of 5-aza-2'-deoxycytidine on the expression of XEDAR and sensitivity to EDA-A2-induced apoptosis in breast cancer cell lines. RESULTS: Expression of XEDAR, but not EDA-A2, was downregulated in most tumorigenic breast cancer cell lines and tumor samples. Loss of XEDAR expression correlated with the hypermethylation of its promoter. Ectopic expression of XEDAR in MDA-MB-231 cells resulted in significant induction of apoptosis and reduction in colony formation. Treatment with 5-aza-2'-deoxycytidine restored XEDAR expression in breast cancer cell lines with methylated XEDAR promoter and sensitized them to EDA-A2-induced cell death. CONCLUSIONS: Our results suggest that XEDAR expression is downregulated in most breast cancers via promoter methylation, which may contribute to accelerated tumor development by blocking EDA-A2-induced cell death. XEDAR may represent a novel breast tumor suppressor gene, and restoration of its expression by treatment with DNA demethylating agents may represent an attractive approach for the treatment of breast cancer.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation , Xedar Receptor/genetics , 5' Flanking Region , Azacitidine/pharmacology , Cell Line, Tumor , Down-Regulation , Ectodysplasins/genetics , Female , Humans , Promoter Regions, Genetic
15.
BMC Med Genomics ; 2: 50, 2009 Aug 06.
Article in English | MEDLINE | ID: mdl-19660139

ABSTRACT

BACKGROUND: Kaposi's sarcoma (KS) associated herpesvirus (KSHV) is the etiological agent of KS, a neoplasm characterized by proliferating spindle cells, extensive neoangiogenesis and a prominent inflammatory infiltrate. Infection of blood vascular endothelial cells with KSHV in vitro results in their spindle cell transformation, which is accompanied by increased expression of inflammatory chemokines and cytokines, and acquisition of lymphatic endothelial markers. Mimicking the effect of viral infection, ectopic expression of KSHV-encoded latent protein vFLIP K13 is sufficient to induce spindle transformation of vascular endothelial cells. However, the effect of K13 expression on global gene expression and induction of lymphatic endothelial markers in vascular endothelial cells has not been studied. METHODS: We used gene array analysis to determine change in global gene expression induced by K13 in human vascular endothelial cells (HUVECs). Results of microarray analysis were validated by quantitative RT-PCR, immunoblotting and a multiplex cytokine array. RESULTS: K13 affected the expression of several genes whose expression is known to be modulated by KSHV infection, including genes involved in immune and inflammatory responses, anti-apoptosis, stress response, and angiogenesis. The NF-kappaB pathway was the major signaling pathway affected by K13 expression, and genetic and pharmacological inhibitors of this pathway effectively blocked K13-induced transcriptional activation of the promoter of CXCL10, one of the chemokines whose expression was highly upregulated by K13. However, K13, failed to induce expression of lymphatic markers in blood vascular endothelial cells. CONCLUSION: While K13 may account for change in the expression of a majority of genes observed following KSHV infection, it is not sufficient for inducing lymphatic reprogramming of blood vascular endothelial cells.

16.
Blood ; 113(22): 5660-8, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19324905

ABSTRACT

Kaposi sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8, is the etiologic agent of Kaposi sarcoma (KS), an angioproliferative lesion characterized by dramatic angiogenesis and inflammatory infiltration. In this study, we report that expression of chemokine CCL20, a potent chemoattractant of dendritic cells and lymphocytes, is strongly induced in cultured cells either by KSHV infection or on ectopic expression of viral FLICE inhibitory protein K13. This induction is caused by transcriptional activation of CCL20 gene, which is mediated by binding of the p65, p50, and c-Rel subunits of the transcription factor nuclear factor-kappaB (NF-kappaB) to an atypical NF-kappaB-binding site present in the CCL20 gene promoter. The CCL20 gene induction is defective in K13 mutants that lack NF-kappaB activity, and can be blocked by specific genetic and pharmacologic inhibitors of the NF-kappaB pathway. CCR6, the specific receptor for CCL20, is also induced in cultured cells either by KSHV infection or on K13 expression. Finally, expression of CCL20 and CCR6 is increased in clinical samples of KS. These results suggest that KSHV and K13-mediated induction of CCL20 and CCR6 may contribute to the recruitment of dendritic cells and lymphocytes into the KS lesions, and to tumor growth and metastases.


Subject(s)
Chemokine CCL20/genetics , Herpesvirus 8, Human/physiology , NF-kappa B/metabolism , Viral Proteins/physiology , Binding Sites , CASP8 and FADD-Like Apoptosis Regulating Protein/antagonists & inhibitors , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/physiology , Cells, Cultured , Chemokine CCL20/metabolism , Dendritic Cells/pathology , Herpesvirus 8, Human/genetics , Humans , K562 Cells , Lymphocytes/pathology , NF-kappa B/antagonists & inhibitors , NF-kappa B/physiology , Promoter Regions, Genetic , Protein Binding , RNA, Small Interfering/pharmacology , Receptors, CCR6/genetics , Receptors, CCR6/metabolism , Sarcoma, Kaposi/immunology , Sarcoma, Kaposi/pathology , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/genetics , Viral Proteins/antagonists & inhibitors , Viral Proteins/genetics
17.
PLoS One ; 2(10): e1067, 2007 Oct 24.
Article in English | MEDLINE | ID: mdl-17957251

ABSTRACT

BACKGROUND: Accumulating evidence suggests that dysregulated expression of lytic genes plays an important role in KSHV (Kaposi's sarcoma associated herpesvirus) tumorigenesis. However, the molecular events leading to the dysregulation of KSHV lytic gene expression program are incompletely understood. METHODOLOGY/PRINCIPAL FINDINGS: We have studied the effect of KSHV-encoded latent protein vFLIP K13, a potent activator of the NF-kappaB pathway, on lytic reactivation of the virus. We demonstrate that K13 antagonizes RTA, the KSHV lytic-regulator, and effectively blocks the expression of lytic proteins, production of infectious virions and death of the infected cells. Induction of lytic replication selects for clones with increased K13 expression and NF-kappaB activity, while siRNA-mediated silencing of K13 induces the expression of lytic genes. However, the suppressive effect of K13 on RTA-induced lytic genes is not uniform and it fails to block RTA-induced viral IL6 secretion and cooperates with RTA to enhance cellular IL-6 production, thereby dysregulating the lytic gene expression program. CONCLUSIONS/SIGNIFICANCE: Our results support a model in which ongoing KSHV lytic replication selects for clones with progressively higher levels of K13 expression and NF-kappaB activity, which in turn drive KSHV tumorigenesis by not only directly stimulating cellular survival and proliferation, but also indirectly by dysregulating the viral lytic gene program and allowing non-lytic production of growth-promoting viral and cellular genes. Lytic Replication-Induced Clonal Selection (LyRICS) may represent a general mechanism in viral oncogenesis.


Subject(s)
Herpesvirus 8, Human/metabolism , Interleukin-6/metabolism , Neoplasms/virology , Virus Replication , Cell Line , DNA Replication , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Expression Regulation, Viral , Gene Silencing , Genes, Viral , Humans , Immediate-Early Proteins/chemistry , RNA, Small Interfering/metabolism , Viral Proteins/chemistry
18.
Br J Haematol ; 138(4): 487-501, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17659053

ABSTRACT

Primary effusion lymphoma (PEL) is a rare, distinct subtype of non-Hodgkin lymphoma, which is associated with Kaposi sarcoma-associated herpesvirus (KSHV) infection. Although MYB levels are high in most neoplastic B cells, we found that, unexpectedly, both PEL cells and uncultured PEL patients' samples contained very low levels of MYB mRNA when compared to B-cell leukaemia samples obtained from KSHV(-) patients. These results were further confirmed at the protein level. Both latent viral FLICE inhibitory protein (v-FLIP) and early lytic viral G protein coupled receptor (v-GPCR) KSHV proteins were found to activate nuclear factor (NF)-kappaB and transrepress a MYB promoter reporter construct. In contrast, a dominant negative inhibitor of NF-kappaB (IkappaB-alpha) mutant prevented v-FLIP and v-GPCR from inhibiting MYB functions while a v-GPCR mutant that was impaired for NF-kappaB activation could not repress the MYB construct. Transduction of a v-FLIP expressing vector or stable transfection of v-GPCR both resulted in a marked downregulation of the endogenous MYB protein expression. However, MYB expression transactivated the lytic switch Replication and Transcription Activator (RTA) promoter in transient transfection assays. Taken together, our results demonstrate that, contrary to a number of other haematological malignancies, MYB expression is not required for PEL cell proliferation. Repressing MYB expression also helps in maintaining the virus in latency.


Subject(s)
Gene Expression Regulation, Viral , Genes, myb , Herpesvirus 8, Human/physiology , Lymphoma, AIDS-Related/virology , Lymphoma, Non-Hodgkin/virology , Sarcoma, Kaposi/virology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Line, Tumor , Cell Transformation, Viral , Gene Expression , Humans , Immediate-Early Proteins/metabolism , Lymphoma, AIDS-Related/metabolism , Lymphoma, Non-Hodgkin/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins c-myb/analysis , Receptors, G-Protein-Coupled/metabolism , Sarcoma, Kaposi/metabolism , Trans-Activators/metabolism , Transcription, Genetic , Transduction, Genetic , Transfection , Viral Proteins/metabolism , Virus Activation , Virus Latency
19.
J Biol Chem ; 282(34): 24858-65, 2007 Aug 24.
Article in English | MEDLINE | ID: mdl-17597077

ABSTRACT

Kaposi's sarcoma herpesvirus oncoprotein vFLIP K13 is a potent activator of NF-kappaB and plays a key role in viral pathogenesis. K13 contains a putative TRAF-interacting motif, which is reportedly required for its interaction with TRAF2. The K13-TRAF2 interaction is believed to be essential for the recruitment of K13 to the I-kappaB kinase (IKK) complex and for K13-induced NF-kappaB and JNK activation. In addition, TRAF3 has been reported to be required for K13-induced NF-kappaB and JNK activation. We have re-examined the role of the TRAFs in K13 signaling and report that mutations in the putative TRAF-interacting motif of K13 have no deleterious effect on its ability to interact with the IKK complex or activation of the NF-kappaB pathway. Furthermore, endogenously expressed TRAF2 and TRAF3 do not interact with K13 and play no role in K13-induced NF-kappaB activation or its interaction with the IKK complex. Finally, K13 does not activate the JNK pathway. Our results support a model in which K13 bypasses the upstream components of the tumor necrosis factor receptor signaling pathway and directly interacts with the IKK complex to selectively activate the NF-kappaB pathway without affecting the JNK pathway. Selective NF-kappaB activation by K13 might represent a novel strategy employed by the virus to promote latency.


Subject(s)
Herpesvirus 8, Human/metabolism , I-kappa B Kinase/metabolism , MAP Kinase Kinase 4/metabolism , Viral Proteins/chemistry , Viral Proteins/physiology , Amino Acid Motifs , Cell Line , Humans , Immunoprecipitation , Models, Molecular , NF-kappa B/metabolism , Plasmids/metabolism , Protein Conformation , RNA Interference , Signal Transduction , U937 Cells
20.
Proc Natl Acad Sci U S A ; 102(36): 12885-90, 2005 Sep 06.
Article in English | MEDLINE | ID: mdl-16120683

ABSTRACT

Human herpesvirus 8 (HHV-8, also called Kaposi's sarcoma-associated herpes virus) has been linked to Kaposi's sarcoma and primary effusion lymphoma. HHV-8-encoded viral Fas-associated death domain-like IL-1-converting enzyme inhibitory protein (vFLIP) is one of the few viral proteins to be expressed in latently infected cells and plays a key role in the survival and proliferation of primary effusion lymphoma cells. Two main functions have been ascribed to HHV-8 vFLIP, inhibition of caspase 8/Fas-associated death domain-like IL-1-converting enzyme and activation of NF-kappaB. In this article, we demonstrate that vFLIP-expressing transgenic mice lack any of the features seen in mice deficient in caspase 8 or Fas-associated death domain protein and are not resistant to Fas-induced apoptosis. On the other hand, these mice display constitutive activation of classical and alternative NF-kappaB pathways, enhanced response to mitogenic stimuli, and increased incidence of lymphoma. Collectively, our results demonstrate that HHV-8 vFLIP is an oncogenic protein that mimics the signaling activities of caspase 8 during antigen receptor signaling and could contribute to the development of lymphoproliferative disorders via constitutive NF-kappaB activation independent of inhibition of Fas-induced apoptosis.


Subject(s)
Apoptosis , Lymphoma/metabolism , Lymphoma/pathology , NF-kappa B/metabolism , Viral Proteins/metabolism , fas Receptor/metabolism , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Transformation, Neoplastic , Cells, Cultured , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/physiology , Homeostasis , Lymphoma/genetics , Mice , Mice, Transgenic , Signal Transduction , Spleen/cytology , Spleen/metabolism , Survival Rate , T-Lymphocytes/metabolism , Transgenes/genetics , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...