Share this post on:

Tive phage clones as possible. In this study, we randomly selected 60 phage clones from three fractions of recovered 3PO phages for further characterization after the fourth round of biopanning. We performed DNA sequencing of the selected phage clones prior to cellular ELISA, which helped to eliminate some unnecessary tests because many phage clones displayed the same sequence. After the 60 phage clones were sequenced, 18 different phage clones or peptide sequences were obtained (Table 1). Subsequently, the 18 phage clones were further tested using cellular ELISA to confirm their specific binding to CHO-K1/VPAC1 cells in vitro. Among these 18 phage clones, VP2 appeared to have the highest OD450 nm and selectivity value, and it bound more effectively than any of the other clones (Figure 3). Therefore, the phage clone VP2 and its displaying peptide were selected for further investigation. A multiple sequence alignment showed that the sequence did not exhibit homology to the sequences of any characterized proteins in various protein databases. This finding demonstrates that VP2 is a novel peptide and may mimic a complex epitope, which may get 58-49-1 explain why VP2 is not present in any database. Therefore, VPScreening of a VPAC1-Binding PeptideFigure 6. Binding of VP2 peptide to CHO-K1/VPAC1 cells and colorectal cancer cell lines (6200). The FITC-conjugated VP2 (FITC-VP2) was incubated with CHO-K1/VPAC1 (A), HT29 (B), SW480(C), SW620 (D) and control CHO-K1 cells (E). At the same time, the control FITC-conjugated unrelated peptide (FITC-URp) was incubated with CHO-K1/VPAC1 (a), HT29 (b), SW480 (c), SW620 (d) and CHO-K1 cells(e). The cells were observed under a fluorescence microscope. doi:10.1371/journal.pone.0054264.gwarrants further investigation regarding its biological characteristics. The results of a competitive inhibition assay demonstrated that the phage VP2 binds to CHO-K1/VPAC1 cells via the VP2 peptide (Figure 4). Although the VP2 peptide specifically binds to CHO-K1/VPAC1 cells, future studies should be performed to determine whether the peptide can specifically bind to the VPAC1 receptor. The results of two receptor-binding assays showed that the binding activity of VP2 was significantly inhibited when VIP was incubated with CHO-K1/VPAC1 cells, which also demonstrated that VIP negatively affects VP2 binding to CHO-K1/ VPAC1 cells (Figure 5). These results confirmed that VIP and VP2 compete for the same binding site, further indicating that VP2 may specifically bind to the VPAC1 receptor. The VP2 peptide inhibited the binding of phage VP2 to CHO-K1/VPAC1 cells with an IC50 of approximately 13.2 nM (Figure 4), which was significantly lower than the IC50 of VIP (Figure 5A). This finding may indicate that VP2 binds to the VPAC1 receptor with a higher affinity than VIP. A fluorescence microscopy assay was performedto directly observe the binding of VP2 to CHO-K1/VPAC1 cells and CRC cells. The results of this assay indicated that VP2 specifically bound to these cells and not the control CHO-K1 cells (Figure 6). Most importantly, the peptide not only bound to the membrane but was also internalized into CRC cells; thus, this peptide could be used as a targeting vector for radionuclide or chemotherapeutic agents and is potentially valuable for targeted imaging and the treatment of CRC. The results of flow cytometry, which were consistent with the results of the fluorescence microscopy assay, further confirmed that VP2 can specifically bind to CHO-K1/.Tive phage clones as possible. In this study, we randomly selected 60 phage clones from three fractions of recovered phages for further characterization after the fourth round of biopanning. We performed DNA sequencing of the selected phage clones prior to cellular ELISA, which helped to eliminate some unnecessary tests because many phage clones displayed the same sequence. After the 60 phage clones were sequenced, 18 different phage clones or peptide sequences were obtained (Table 1). Subsequently, the 18 phage clones were further tested using cellular ELISA to confirm their specific binding to CHO-K1/VPAC1 cells in vitro. Among these 18 phage clones, VP2 appeared to have the highest OD450 nm and selectivity value, and it bound more effectively than any of the other clones (Figure 3). Therefore, the phage clone VP2 and its displaying peptide were selected for further investigation. A multiple sequence alignment showed that the sequence did not exhibit homology to the sequences of any characterized proteins in various protein databases. This finding demonstrates that VP2 is a novel peptide and may mimic a complex epitope, which may explain why VP2 is not present in any database. Therefore, VPScreening of a VPAC1-Binding PeptideFigure 6. Binding of VP2 peptide to CHO-K1/VPAC1 cells and colorectal cancer cell lines (6200). The FITC-conjugated VP2 (FITC-VP2) was incubated with CHO-K1/VPAC1 (A), HT29 (B), SW480(C), SW620 (D) and control CHO-K1 cells (E). At the same time, the control FITC-conjugated unrelated peptide (FITC-URp) was incubated with CHO-K1/VPAC1 (a), HT29 (b), SW480 (c), SW620 (d) and CHO-K1 cells(e). The cells were observed under a fluorescence microscope. doi:10.1371/journal.pone.0054264.gwarrants further investigation regarding its biological characteristics. The results of a competitive inhibition assay demonstrated that the phage VP2 binds to CHO-K1/VPAC1 cells via the VP2 peptide (Figure 4). Although the VP2 peptide specifically binds to CHO-K1/VPAC1 cells, future studies should be performed to determine whether the peptide can specifically bind to the VPAC1 receptor. The results of two receptor-binding assays showed that the binding activity of VP2 was significantly inhibited when VIP was incubated with CHO-K1/VPAC1 cells, which also demonstrated that VIP negatively affects VP2 binding to CHO-K1/ VPAC1 cells (Figure 5). These results confirmed that VIP and VP2 compete for the same binding site, further indicating that VP2 may specifically bind to the VPAC1 receptor. The VP2 peptide inhibited the binding of phage VP2 to CHO-K1/VPAC1 cells with an IC50 of approximately 13.2 nM (Figure 4), which was significantly lower than the IC50 of VIP (Figure 5A). This finding may indicate that VP2 binds to the VPAC1 receptor with a higher affinity than VIP. A fluorescence microscopy assay was performedto directly observe the binding of VP2 to CHO-K1/VPAC1 cells and CRC cells. The results of this assay indicated that VP2 specifically bound to these cells and not the control CHO-K1 cells (Figure 6). Most importantly, the peptide not only bound to the membrane but was also internalized into CRC cells; thus, this peptide could be used as a targeting vector for radionuclide or chemotherapeutic agents and is potentially valuable for targeted imaging and the treatment of CRC. The results of flow cytometry, which were consistent with the results of the fluorescence microscopy assay, further confirmed that VP2 can specifically bind to CHO-K1/.

Share this post on:

Author: bet-bromodomain.