Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Int Immunopharmacol ; 19(1): 119-26, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24468679

ABSTRACT

Interactions between CD155 and nectins on tumor cells have been reported to potentially inhibit tumor growth. CD226, a receptor that recognizes CD155 and CD112, is an activation receptor of NK and T cells by which immune cells may attack a tumor. The purpose of this study is to explore whether soluble CD226 (sCD226) directly inhibits tumor growth by binding CD155 or CD112 on tumor cells. We expressed, purified and confirmed the identity of recombinant sCD226 (19aa-248aa) and then examined the effect of sCD226 on tumor cell growth using CD226 ligand (CD155 and CD112)-expressing cancer cell lines (K562, HeLa). After 3days of co-culture with sCD226, we found that the numbers of K562 and HeLa cells were significantly reduced but those of a CD226-blocking mAb specifically attenuated the inhibitory effects of sCD226. We also noted that the sCD226 protein could compete with a PE-conjugated anti-CD112 antibody in flow cytometric analysis and block the binding of the PE-conjugated anti-CD112 antibody to tumor cells. Mechanistic studies using flow cytometric analysis demonstrated that sCD226 inhibited the division of CFSE (carboxyfluorescein diacetate succinimidyl ester)-labeled K562 cells by delaying the cell cycle. In addition, we observed that sCD226 might have an impact on the metastatic potential of solid tumors in vitro. These results demonstrated that sCD226 molecule might be a potential biotherapy against tumor for further development.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/pharmacology , Antineoplastic Agents/pharmacology , Animals , Antigens, Differentiation, T-Lymphocyte/genetics , CHO Cells , Cell Cycle/drug effects , Cell Line , Cell Proliferation/drug effects , Cricetulus , HeLa Cells , Humans , K562 Cells , Neoplasms , Protein Structure, Tertiary , Recombinant Proteins/pharmacology
2.
J Biol Chem ; 289(10): 6969-6977, 2014 Mar 07.
Article in English | MEDLINE | ID: mdl-24451371

ABSTRACT

CD226, an activating receptor that interacts with the ligands CD155 and CD112, activates natural killer (NK) cells via its immunoreceptor tyrosine-based activatory motif (ITAM). There are two extracellular domains of CD226; however, the comparative functional relevance of these domains remains unknown. In this study, two different deletion mutants, rCD226-ECD1 (the first extracellular domain) and rCD226-ECD (full extracellular domains), were recombinantly expressed. We observed that rCD226-ECD1, similar to rCD226-ECD, specifically bound to ligand-positive cell lines and that this interaction could be competitively blocked by an anti-CD226 mAb. In addition, rCD226-ECD1 was able to block the binding of CD112 mAb to tumor cells in a competitive binding assay. Importantly, based on surface plasmon resonance (SPR), we determined that rCD226-ECD1, similar to rCD226-ECD, directly bound to its ligand CD155 on a protein chip. Functionally, NK cell cytotoxicity against K562 or HeLa cells was blocked by rCD226-ECD1 by reducing the expression of CD69 and granzyme B, indicating the critical role of ECD1 in NK cell activation. We also examined the role of rCD226-ECD1 in effector/target interactions by using rCD226-ECD to block these interactions. Using flow cytometry, we found that the number of conjugates between IL-2-dependent NKL cells and HeLa cells was reduced and observed that the formation of immune synapses was also decreased under confocal microscopy. In addition, we prepared two anti-rCD226-ECD1 agonistic antibodies, 2E6 and 3B9. Both 2E6 and 3B9 antibodies could induce the phosphorylation of ERK in NK-92 cells. Taken together, our results show that CD226 functions via its first extracellular domain.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Immunological Synapses/physiology , Killer Cells, Natural/immunology , Lymphocyte Activation , Amino Acid Sequence , Animals , Antigens, Differentiation, T-Lymphocyte/chemistry , Antigens, Differentiation, T-Lymphocyte/genetics , CD55 Antigens/metabolism , CHO Cells , Cricetulus , Cytotoxicity, Immunologic , HeLa Cells , Humans , Immunological Synapses/metabolism , K562 Cells , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Surface Plasmon Resonance
3.
Small ; 6(5): 670-8, 2010 Mar 08.
Article in English | MEDLINE | ID: mdl-20143348

ABSTRACT

Fluorescent quantum dots (QDs) have great potential for in vivo biomedical imaging and diagnostic applications. However, these nanoparticles are composed of heavy metals and are very small in diameter, and their possible toxicity must therefore be considered. As yet, no studies have reported the transfer of QDs between mother and fetus. The transfer of CdTe/CdS QDs of different sizes and dosages, and with different outer capping materials, from pregnant mice to fetuses is investigated. It is shown that QDs may be transferred from female mice to their fetuses across the placental barrier. Smaller QDs are more easily transferred than larger QDs and the number of QDs transferred increases with increasing dosage. Capping with an inorganic silica shell or organic polyethylene glycol reduces QD transfer but does not eliminate it. These results suggest that the clinical utility of QDs could be limited in pregnant women.


Subject(s)
Fetus/metabolism , Placenta/metabolism , Quantum Dots , Animals , Female , Mice , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Nanotechnology , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...