Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
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.
Mol Cell Biochem ; 377(1-2): 163-76, 2013 May.
Article in English | MEDLINE | ID: mdl-23397134

ABSTRACT

Hepatocellular carcinoma (HCC) incidence rates are increasing in many parts of the world. HCC's limited treatment remedies and the poor prognosis emphasize the importance in developing an effective chemoprevention for this disease. Here, we investigated the molecular mechanisms involved in the chemoprevention of silymarin in N-nitrosodiethylamine (NDEA)-induced rat model of HCC. Liver of the rats treated with NDEA showed higher proliferation index and glycoconjugates. NDEA treatment also increased the level of anti-apoptotic proteins with simultaneous decrease in the level of pro-apoptotic proteins along with increased accumulation of Cytochrome c in mitochondria. The carcinogenic insult also increased microsomal phase I metabolizing enzymes with a simultaneous decrease in the Phase II detoxifying enzyme glutathione-S-transferase (GST). Whereas dietary silymarin administration along with NDEA treatment significantly decreased the proliferation and down regulated the expression of anti-apoptotic proteins with simultaneously increased expression of pro-apoptotic proteins along with the release of Cytochrome c to cytosol there by activating the intrinsic apoptotic pathway. Silymarin administration also decreased the level of glycoproteins and activated the phase II detoxifying enzyme GST. These results demonstrate that suppression of HCC by silymarin in vivo involves inhibition of proliferation, activation of apoptosis, and efficient detoxification.


Subject(s)
Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Liver Neoplasms, Experimental/drug therapy , Silymarin/administration & dosage , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Caspase 3/metabolism , Cells, Cultured , Cyclin D1/metabolism , Dietary Supplements , Drug Screening Assays, Antitumor , Glycoproteins/metabolism , Hexosamines/metabolism , Hexoses/metabolism , Liver/drug effects , Liver/metabolism , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Metabolic Detoxication, Phase II , Microtubule-Associated Proteins/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Rats , Rats, Wistar , Sialic Acids/metabolism , Survivin , Tumor Suppressor Protein p53/metabolism , beta Catenin/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.
Thromb Haemost ; 107(5): 951-61, 2012 May.
Article in English | MEDLINE | ID: mdl-22370814

ABSTRACT

Recent in vitro studies have shown that the zymogen and activated form of factor (F)VII bind to endothelial cell protein C receptor (EPCR). At present, there is no evidence that FVIIa binds to EPCR on vascular endothelium in vivo in the presence of circulating protein C, a primary ligand for EPCR. The present study was carried out to investigate the interaction of murine and human ligands with murine EPCR both in vivo and in vitro . Measurement of endogenous plasma levels of FVII in wild-type, EPCR-deficient and EPCR-over expressing mice showed slightly lower levels of FVII in EPCR-over expressing mice. However, infusion of high concentrations of competing ligands, either human APCi or FVIIai, to EPCR-over expressing mice failed to increase plasma levels of mouse FVII whereas they increased the plasma levels of protein C by two- to three-fold. Examining the association of exogenously administered mouse FVIIa or human FVIIa by immunohistochemistry revealed that human, but not murine FVIIa, binds to the murine endothelium in an EPCR-dependent manner. In vitro binding studies performed using surface plasmon resonance and endothelial cells revealed that murine FVIIa binds murine EPCR negligibly. Human FVIIa binding to EPCR, particularly to mouse EPCR, is markedly enhanced by availability of Mg2+ ions. In summary, our data show that murine FVIIa binds poorly to murine EPCR, whereas human FVIIa binds efficiently to both murine and human EPCR. Our data suggest that one should consider the use of human FVIIa in mouse models to investigate the significance of FVIIa and EPCR interaction.


Subject(s)
Antigens, CD/metabolism , Endothelium, Vascular/metabolism , Factor VIIa/metabolism , Glycoproteins/metabolism , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Animals , Blood Coagulation Factor Inhibitors/metabolism , Cells, Cultured , Endothelial Protein C Receptor , Factor VIIa/administration & dosage , Glycoproteins/deficiency , Glycoproteins/genetics , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Immunohistochemistry , Ligands , Magnesium/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Protein Binding , Protein C/metabolism , Protein C Inhibitor/metabolism , Radioligand Assay , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Species Specificity , Surface Plasmon Resonance
12.
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
13.
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
14.
Blood ; 117(11): 3199-208, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21252088

ABSTRACT

Recent studies have shown that factor VIIa (FVIIa) binds to the endothelial cell protein C receptor (EPCR), a cellular receptor for protein C and activated protein C, but the physiologic significance of this interaction is unclear. In the present study, we show that FVIIa, upon binding to EPCR on endothelial cells, activates endogenous protease activated receptor-1 (PAR1) and induces PAR1-mediated p44/42 mitogen-activated protein kinase (MAPK) activation. Pretreatment of endothelial cells with FVIIa protected against thrombin-induced barrier disruption. This FVIIa-induced, barrier-protective effect was EPCR dependent and did not involve PAR2. Pretreatment of confluent endothelial monolayers with FVIIa before thrombin reduced the development of thrombin-induced transcellular actin stress fibers, cellular contractions, and paracellular gap formation. FVIIa-induced p44/42 MAPK activation and the barrier-protective effect are mediated via Rac1 activation. Consistent with in vitro findings, in vivo studies using mice showed that administration of FVIIa before lipopolysaccharide (LPS) treatment attenuated LPS-induced vascular leakage in the lung and kidney. Overall, our present data provide evidence that FVIIa bound to EPCR on endothelial cells activates PAR1-mediated cell signaling and provides a barrier-protective effect. These findings are novel and of great clinical significance, because FVIIa is used clinically for the prevention of bleeding in hemophilia and other bleeding disorders.


Subject(s)
Antigens, CD/metabolism , Endothelial Cells/metabolism , Factor VIIa/metabolism , Receptor, PAR-1/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Capillary Permeability/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Endothelial Protein C Receptor , Enzyme Activation/drug effects , Factor VIIa/administration & dosage , Factor VIIa/pharmacology , Humans , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3/metabolism , Protein Binding/drug effects , Signal Transduction/drug effects , Thrombin/pharmacology , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
15.
Blood ; 114(9): 1974-86, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19587380

ABSTRACT

Although the binding of endothelial cell protein C receptor (EPCR) to its ligands is well characterized at the biochemical level, it remains unclear how EPCR interaction with its ligands at the cell surface impacts its cellular trafficking. We characterized the cellular localization and trafficking of EPCR in endothelial cells and a heterologous expression system. Immunofluorescence confocal microscopy studies revealed that a majority of EPCR is localized on the cell surface in membrane microdomains that are positive for caveolin-1. A small fraction of EPCR is also localized intracellularly in the recycling compartment. Factor VIIa (FVIIa) or activated protein C binding to EPCR promoted the internalization of EPCR. EPCR and EPCR-bound ligands were endocytosed rapidly via a dynamin- and caveolar-dependent pathway. The endocytosed receptor-ligand complexes were accumulated in a recycling compartment before being targeted back to the cell surface. EPCR-mediated FVIIa endocytosis/recycling also resulted in transport of FVIIa from the apical to the basal side. In vivo studies in mice showed that blockade of EPCR with EPCR-blocking antibodies impaired the early phase of FVIIa clearance. Overall, our results show that FVIIa or activated protein C binding to EPCR promotes EPCR endocytosis, and EPCR-mediated endocytosis may facilitate the transcytosis of FVIIa and its clearance from the circulation.


Subject(s)
Blood Coagulation Factors/biosynthesis , Factor VIIa/metabolism , Protein C/metabolism , Receptors, Cell Surface/biosynthesis , Animals , CHO Cells , Cricetinae , Cricetulus , Endocytosis , Endothelium, Vascular/cytology , Humans , Ligands , Mice , Microscopy, Fluorescence/methods , Models, Biological , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...