Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS Pathog ; 9(3): e1003226, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23555244

ABSTRACT

Both NK cells and CTLs kill virus-infected and tumor cells. However, the ways by which these killer cells recognize the infected or the tumorigenic cells are different, in fact almost opposite. CTLs are activated through the interaction of the TCR with MHC class I proteins. In contrast, NK cells are inhibited by MHC class I molecules. The inhibitory NK receptors recognize mainly MHC class I proteins and in this regard practically all of the HLA-C proteins are recognized by inhibitory NK cell receptors, while only certain HLA-A and HLA-B proteins interact with these receptors. Sophisticated viruses developed mechanisms to avoid the attack of both NK cells and CTLs through, for example, down regulation of HLA-A and HLA-B molecules to avoid CTL recognition, leaving HLA-C proteins on the cell surface to inhibit NK cell response. Here we provide the first example of a virus that through specific down regulation of HLA-C, harness the NK cells for its own benefit. We initially demonstrated that none of the tested HSV-2 derived microRNAs affect NK cell activity. Then we show that surprisingly upon HSV-2 infection, HLA-C proteins are specifically down regulated, rendering the infected cells susceptible to NK cell attack. We identified a motif in the tail of HLA-C that is responsible for the HSV-2-meduiated HLA-C down regulation and we show that the HLA-C down regulation is mediated by the viral protein ICP47. Finally we show that HLA-C proteins are down regulated from the surface of HSV-2 infected dendritic cells (DCs) and that this leads to the killing of DC by NK cells. Thus, we propose that HSV-2 had developed this unique and surprising NK cell-mediated killing strategy of infected DC to prevent the activation of the adaptive immunity.


Subject(s)
Cytotoxicity, Immunologic , Dendritic Cells/immunology , HLA-C Antigens/metabolism , Herpesviridae Infections/immunology , Herpesvirus 2, Human/immunology , Immediate-Early Proteins/immunology , Killer Cells, Natural/immunology , Adaptive Immunity , Cell Survival , Cloning, Molecular , Dendritic Cells/virology , Down-Regulation , Gene Expression Regulation, Viral , Herpesviridae Infections/metabolism , Herpesviridae Infections/virology , Herpesvirus 2, Human/genetics , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/metabolism , Killer Cells, Natural/metabolism , Lymphocyte Activation/immunology
2.
Biotechnol Prog ; 28(2): 573-80, 2012.
Article in English | MEDLINE | ID: mdl-22275241

ABSTRACT

Designing a chimeric protein and developing a procedure for its stable production as a biologically active protein, are key steps in its potential application to clinical trails. IL2-Caspase3 chimeric protein designed to target activated T lymphocytes was found to be a promising molecule for targeted treatment, however was found to be difficult to produce as a biological active molecule. Thus, we designed a new version of the molecule, IL2-Caspase3s, in which six amino acids (aa 29-34) from the N-terminus of the large subunit of caspase 3 were excluded. Repeated expressions, productions, and partial purifications of the IL2-Caspase3s yielded reproducible batches with consistent results. We found that IL2-Caspase3s causes cell death in a specific, dose-, and time-dependent manner. Cell death due to IL2-Caspase3s is caused by apoptosis. This improved and biologically stable IL2-Caspase3s chimeric protein may be developed in the future for clinical trails as a promising therapy for several pathologies involving activated T-cells. Moreover, this truncated caspase 3 sequence, lacking the N-terminal six amino acids of its large subunit, may be used in other caspase 3-based chimeric proteins targeted against various human diseases, using the appropriate targeting moiety.


Subject(s)
Caspase 3/chemistry , Caspase 3/metabolism , Interleukin-2/pharmacology , Amino Acid Motifs , Animals , Apoptosis/drug effects , Caspase 3/genetics , Caspase 3/pharmacology , Cell Cycle/drug effects , Cell Line , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Lymphocyte Activation/drug effects , Mice , Molecular Sequence Data , Protein Subunits/chemistry , Protein Subunits/genetics , Protein Subunits/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology
3.
Int J Cancer ; 128(8): 1966-80, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-20568105

ABSTRACT

One of the main problems of conventional anticancer therapy is multidrug resistance (MDR), whereby cells acquire resistance to structurally and functionally unrelated drugs following chemotherapeutic treatment. One of the main causes of MDR is overexpression of the P-glycoprotein transporter. In addition to extruding the chemotherapeutic drugs, it also inhibits apoptosis through the inhibition of caspases. To overcome MDR, we constructed a novel chimeric protein, interleukin (IL)-2 granzyme A (IGA), using IL-2 as a targeting moiety and granzyme A as a killing moiety, fused at the cDNA level. IL-2 binds to the high-affinity IL-2 receptor that is expressed in an array of abnormal cells, including malignant cells. Granzyme A is known to cause caspase 3-independent cell death. We show here that the IGA chimeric protein enters the target sensitive and MDR cancer cells overexpressing IL-2 receptor and induces caspase 3-independent cell death. Specifically, after its entry, IGA causes a decrease in the mitochondrial potential, triggers translocation of nm23-H1, a granzyme A-dependent DNase, from the cytoplasm to the nucleus, where it causes single-strand DNA nicks, thus causing cell death. Moreover, IGA is able to overcome MDR and kill cells resistant to chemotherapeutic drugs. We believe that overcoming MDR with targeted molecules such as IGA chimeric protein that causes caspase-independent apoptotic cell death could be applied to many other resistant types of tumors using the appropriate targeting moiety. Thus, this novel class of targeted molecules could open up new vistas in the fight against human cancer.


Subject(s)
Apoptosis , Caspase 3/metabolism , Drug Resistance, Multiple , Granzymes/metabolism , Interleukin-2/metabolism , Lymphoma/drug therapy , Recombinant Fusion Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Blotting, Western , Cell Nucleus/metabolism , Cell Proliferation , Cytoplasm/metabolism , DNA Damage , Drug Resistance, Neoplasm , Granzymes/genetics , Humans , Interleukin-2/genetics , Lymphoma/metabolism , Lymphoma/pathology , Membrane Potential, Mitochondrial , Mice , Mitochondria/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Protein Transport , Receptors, Interleukin-2/metabolism , Recombinant Fusion Proteins/genetics , Tumor Cells, Cultured
4.
Int J Oncol ; 27(1): 143-8, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15942653

ABSTRACT

Since early diagnosis of many types of cancer greatly improves the chances for successful treatment, high-quality methods for cancer detection are necessary. Our laboratory develops chimeric proteins for targeted therapy, such as gonadotropin releasing hormone (GnRH)-based chimeric proteins for the targeted therapy of adenocarcinomas in humans. For chimeric proteins to cause specific cell death, they must first recognize specific receptors/binding sites expressed on the surface of target cells. Thus, we examined whether we could exploit these binding sites not only as targets for the killing of specific cells but also as a diagnostic marker for identifying adenocarcinomas, using the same chimeric proteins. In this report, we show that one such GnRH-based chimeric protein, GnRH-Caspase3, can indeed serve as a diagnostic tool. GnRH-Caspase3 was able to specifically bind adenocarcinoma cells, as measured by FACS analysis and demonstrated with the aid of confocal microscopy and specific antibodies. Moreover, we found a correlation between cell sensitivity to treatment and the binding level of the chimeric protein to the cells. Hence, we suggest that in addition to their therapeutic potential, GnRH-based chimeric proteins can be used as a diagnostic tool for the detection of adenocarcinomas.


Subject(s)
Adenocarcinoma/diagnosis , Gonadotropin-Releasing Hormone/chemistry , Recombinant Fusion Proteins/chemistry , Adenocarcinoma/metabolism , Binding Sites , Caspase 3 , Caspases/metabolism , Cell Line, Tumor , Cell Separation , Cell Survival , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Gonadotropin-Releasing Hormone/metabolism , Humans , Microscopy, Confocal , Plasmids/metabolism , Protein Binding , Recombinant Fusion Proteins/metabolism , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...