4,001
Views
34
CrossRef citations to date
0
Altmetric
Research Article

Development of Lactobacillus kimchicus DCY51T-mediated gold nanoparticles for delivery of ginsenoside compound K: in vitro photothermal effects and apoptosis detection in cancer cells

, , , , , , , , , , & show all
Pages 30-44 | Received 15 Jun 2018, Accepted 13 Oct 2018, Published online: 19 Jan 2019

Figures & data

Figure 1. HR-TEM analysis of (A) DCY51T-AuCKNps and (B) DCY65-AuCKNps which were derived from two different Lactobacillus strains: L. kimchicus DCY51T and L. brevis DCY65, respectively. HR-TEM analysis of model drug compounds derived from Lactobacillus kimchicus DCY51T. (C) DCY51T-DoxorubicinNps. (D) DCY51T-RifaximinNps.

Figure 1. HR-TEM analysis of (A) DCY51T-AuCKNps and (B) DCY65-AuCKNps which were derived from two different Lactobacillus strains: L. kimchicus DCY51T and L. brevis DCY65, respectively. HR-TEM analysis of model drug compounds derived from Lactobacillus kimchicus DCY51T. (C) DCY51T-DoxorubicinNps. (D) DCY51T-RifaximinNps.

Figure 2. TEM analysis of (A) DCY51T-AuCKNps, (B) DCY51T-DoxorubicinNps and (C) DCY51T-RifaximinNps. TEM images of (a, b) DCY51T-AuCKNps, (f, g) DCY51T-DoxorubicinNps and (k, i) DCY51T-RifaximinNps. Gold distribution of (c) DCY51T-AuCKNps, (h) DCY51T-DoxorubicinNps and (m) DCY51T-RifaximinNps. SAED of (d) DCY51T-AuCKNps, (i) DCY51T-DoxorubicinNps and (n) DCY51T-RifaximinNps. EDX of (e) DCY51T-AuCKNps, (j) DCY51T-DoxorubicinNps and (o) DCY51T-RifaximinNps.

Figure 2. TEM analysis of (A) DCY51T-AuCKNps, (B) DCY51T-DoxorubicinNps and (C) DCY51T-RifaximinNps. TEM images of (a, b) DCY51T-AuCKNps, (f, g) DCY51T-DoxorubicinNps and (k, i) DCY51T-RifaximinNps. Gold distribution of (c) DCY51T-AuCKNps, (h) DCY51T-DoxorubicinNps and (m) DCY51T-RifaximinNps. SAED of (d) DCY51T-AuCKNps, (i) DCY51T-DoxorubicinNps and (n) DCY51T-RifaximinNps. EDX of (e) DCY51T-AuCKNps, (j) DCY51T-DoxorubicinNps and (o) DCY51T-RifaximinNps.

Figure 3. Particle size distribution (DLS) of the obtained nanoparticles with respect to intensity, number and volume. (A) DCY51T-AuCKNps. (B) DCY51T-DoxorubicinNps. (C) DCY51T-RifaximinNps.

Figure 3. Particle size distribution (DLS) of the obtained nanoparticles with respect to intensity, number and volume. (A) DCY51T-AuCKNps. (B) DCY51T-DoxorubicinNps. (C) DCY51T-RifaximinNps.

Figure 4. (A) UV–vis spectra of DCY51T-AuNps, with and without ginsenoside CK. (B) XRD spectrum of DCY51T-AuCKNps. (C) Photoluminescence of DCY51T-AuCKNps.

Figure 4. (A) UV–vis spectra of DCY51T-AuNps, with and without ginsenoside CK. (B) XRD spectrum of DCY51T-AuCKNps. (C) Photoluminescence of DCY51T-AuCKNps.

Figure 5. UV–vis absorbance spectra of the obtained nanoparticles depicting their high stability in various conditions due to protein capping. Stability of DCY51T-AuNps and DCY51T-AuCKNps against varied pH conditions (A, B) and electrolytic concentrations (C, D) and three-month storage (E, F), respectively.

Figure 5. UV–vis absorbance spectra of the obtained nanoparticles depicting their high stability in various conditions due to protein capping. Stability of DCY51T-AuNps and DCY51T-AuCKNps against varied pH conditions (A, B) and electrolytic concentrations (C, D) and three-month storage (E, F), respectively.

Figure 6. FTIR spectra of DCY51T-AuCKNps and proposed ginsenoside CK complexation onto the surface of DCY51T-AuNps. FT-IR spectra of DCY51T-AuCKNps revealed the presence of C–H bends (alkane groups) and C–O stretch (ethers) of ginsenoside CK.

Figure 6. FTIR spectra of DCY51T-AuCKNps and proposed ginsenoside CK complexation onto the surface of DCY51T-AuNps. FT-IR spectra of DCY51T-AuCKNps revealed the presence of C–H bends (alkane groups) and C–O stretch (ethers) of ginsenoside CK.

Figure 7. LC–MS spectra of ginsenoside CK released from DCY51T-AuCKNps after 24 h of sonication in DMSO-MeOH mixture (1:4, v/v). Inset distinguishes the ginsenoside CK released from the intracellular content of cell debris, supernatant, and purified DCY51T-AuCKNps.

Figure 7. LC–MS spectra of ginsenoside CK released from DCY51T-AuCKNps after 24 h of sonication in DMSO-MeOH mixture (1:4, v/v). Inset distinguishes the ginsenoside CK released from the intracellular content of cell debris, supernatant, and purified DCY51T-AuCKNps.

Figure 8. Cytotoxicity of DCY51T-AuNps after 48 h of incubation. (A) Macrophage cell line RAW264.7. (B) Keratinocyte cell line HaCaT. (C) Lung cancer cell line A549. (D) Colon cancer cell line HT29. Results are presented as means ± SDs.

Figure 8. Cytotoxicity of DCY51T-AuNps after 48 h of incubation. (A) Macrophage cell line RAW264.7. (B) Keratinocyte cell line HaCaT. (C) Lung cancer cell line A549. (D) Colon cancer cell line HT29. Results are presented as means ± SDs.

Figure 9. Antiproliferative activity (MTT assay) (A) of DCY51T-AuCkNPs on the cancer cell lines with and without treatment (B). Cytotoxicity of DCY51T-AuCKNps and ginsenoside CK after 48 h of incubation. (A) Macrophage cell line RAW264.7. (B) Lung cancer cell line A549. (C) Colon cancer cell line HT29. Results are presented as means ± SDs.

Figure 9. Antiproliferative activity (MTT assay) (A) of DCY51T-AuCkNPs on the cancer cell lines with and without treatment (B). Cytotoxicity of DCY51T-AuCKNps and ginsenoside CK after 48 h of incubation. (A) Macrophage cell line RAW264.7. (B) Lung cancer cell line A549. (C) Colon cancer cell line HT29. Results are presented as means ± SDs.

Figure 10. Detection of intercellular DCY51T-AuCKNps in the stomach cancer cell lines (AGS). (A) Control sample, (B) 800 nm laser treatment for 10 min. (C) DCY51T-AuCKNps + laser at 800 nm for 10 min.

Figure 10. Detection of intercellular DCY51T-AuCKNps in the stomach cancer cell lines (AGS). (A) Control sample, (B) 800 nm laser treatment for 10 min. (C) DCY51T-AuCKNps + laser at 800 nm for 10 min.

Figure 11. Fluorescent images of cells stained with Hoechst stain after an incubation time of Ankrum h. (A) Blank. (B) DCY51T-AuNps; 5 µg/mL. (C) DCY51T-AuCKNps; 1 µg/mL. (D) DCY51T-AuCKNps; 5 µg/mL. Apoptotic cells are indicated with white arrows. The scale bar equals 10 µm. There were more apoptotic cells in the DCY51T-AuCKNps groups than in blank or in the negative control (DCY51T-AuNps). The co-treatment groups of hyperthermia and chemotherapy exhibited the greatest number of apoptosis in cancer cells.

Figure 11. Fluorescent images of cells stained with Hoechst stain after an incubation time of Ankrum h. (A) Blank. (B) DCY51T-AuNps; 5 µg/mL. (C) DCY51T-AuCKNps; 1 µg/mL. (D) DCY51T-AuCKNps; 5 µg/mL. Apoptotic cells are indicated with white arrows. The scale bar equals 10 µm. There were more apoptotic cells in the DCY51T-AuCKNps groups than in blank or in the negative control (DCY51T-AuNps). The co-treatment groups of hyperthermia and chemotherapy exhibited the greatest number of apoptosis in cancer cells.

Figure 12. The proposed mechanism of cell internalization of DCY51T-AuCKNps by the EPR effect and in vitro photothermal therapy. DCY51T-AuCKNps reaches the cell membrane by exploiting the properties of the EPR effect. Following association with the cell by endocytosis, DCY51T-AuCKNps may aggregate on the anionic surface of cancer cells due to the cationic surface charge of the nanoparticles. In conjunction, DCY51T-AuCKNps can be irradiated to rapidly induce cell lysis.

Figure 12. The proposed mechanism of cell internalization of DCY51T-AuCKNps by the EPR effect and in vitro photothermal therapy. DCY51T-AuCKNps reaches the cell membrane by exploiting the properties of the EPR effect. Following association with the cell by endocytosis, DCY51T-AuCKNps may aggregate on the anionic surface of cancer cells due to the cationic surface charge of the nanoparticles. In conjunction, DCY51T-AuCKNps can be irradiated to rapidly induce cell lysis.
Supplemental material

Supplemental Material

Download ()