Nothing Special   »   [go: up one dir, main page]

You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (5)

Search Parameters:
Keywords = thymopentin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
10 pages, 3008 KiB  
Communication
Entire Encapsulation of Thymopentin by Extended Biphen[3]arene Carboxylate for Improving Plasma Stability
by Keming Ren, Junyi Chen and Chunju Li
Molecules 2025, 30(2), 314; https://doi.org/10.3390/molecules30020314 - 15 Jan 2025
Viewed by 366
Abstract
Peptide-based therapy is appealing in modern medicine owing to its high activity and excellent biocompatibility. Poor stability, leading to unacceptable bioavailability, severely constrains its clinical application. Here, we proposed a general supramolecular approach for improving the plasma resistance of a commercially available peptide [...] Read more.
Peptide-based therapy is appealing in modern medicine owing to its high activity and excellent biocompatibility. Poor stability, leading to unacceptable bioavailability, severely constrains its clinical application. Here, we proposed a general supramolecular approach for improving the plasma resistance of a commercially available peptide agent, thymopentin. The 1H NMR results indicated that the large-sized extended biphen[3]arene carboxylate (ExBP3C) can entirely encapsulate this peptide at its main chain with a binding stoichiometry of 1:1 and Ka value of (1.87 ± 0.15) × 105 M−1, which varied radically from recognizing specific amino acid residues by carboxylatopillar[5]arene (CP5A). Notably, host–guest complexation by ExBP3C could maintain 24.85% of the original thymopentin amount for 60 min in the presence of rat plasma, whereas free thymopentin, or co-dosed with CP5A and cucurbit[7]uril, underwent rapid degradation and became undetectable within just 30 min. In addition, cytotoxicity and hemolysis assays preliminary demonstrated that the employment of ExBP3C as a supplementary material was relatively nontoxic at a cellular level. Full article
Show Figures

Figure 1

Figure 1
<p><sup>1</sup>H NMR spectra (600 MHz, D<sub>2</sub>O) of (<b>a</b>) thymopentin (5.0 mM), (<b>b</b>) thymopentin (5.0 mM) with the addition of ExBP3C (5.0 mM), and (<b>c</b>) ExBP3C (5.0 mM).</p>
Full article ">Figure 2
<p><sup>1</sup>H NMR spectra (400 MHz, D<sub>2</sub>O) of (<b>a</b>) thymopentin (5.0 mM), (<b>b</b>) thymopentin (5.0 mM) with the addition of CP5A (5.0 mM), and (<b>c</b>) CP5A (5.0 mM).</p>
Full article ">Figure 3
<p>ITC isotherm for the titration of thymopentin/ExBP3C in aqueous solution at room temperature (<b>top</b>: raw data; <b>bottom</b>: net reaction heat obtained from integration of calorimetric traces).</p>
Full article ">Figure 4
<p>Possible scheme for degradation of thymopentin in plasma.</p>
Full article ">Figure 5
<p>(<b>a</b>) Calibration curve obtained by HPLC and used to calculate thymopentin concentration in plasma stability. (<b>b</b>) Residual percentage of thymopentin (1.0 mM) in rat plasma in the absence and presence of ExBP3C (2.0 mM), CP5A (2.0 mM), and CB[7] (2.0 mM) as determined by HPLC.</p>
Full article ">Figure 6
<p>(<b>a</b>) Relative viability of mouse fibroblast cell line (L929) after treatment with ExBP3C for 24 h at the indicated concentrations (mean ± SD, <span class="html-italic">n</span> = 5). (<b>b</b>) Hemolysis of ExBP3C toward rabbit red blood cells (rRBCs) after 24 h of incubation time, and the hemolytic rate of rRBCs incubated with 1% Triton X-100 was defined as 100% (mean ± SD, <span class="html-italic">n</span> = 3).</p>
Full article ">Scheme 1
<p>Chemical structures of thymopentin, extended biphen[3]arene carboxylate (ExBP3C), and carboxylatopillar[5]arene (CP5A), and schematic illustration of the impact of different recognition modes by macrocycles on plasma stability of thymopentin.</p>
Full article ">
23 pages, 8398 KiB  
Article
Cellular Uptake and Transport Mechanism Investigations of PEGylated Niosomes for Improving the Oral Delivery of Thymopentin
by Mengyang Liu, Darren Svirskis, Thomas Proft, Jacelyn Loh, Yuan Huang and Jingyuan Wen
Pharmaceutics 2024, 16(3), 397; https://doi.org/10.3390/pharmaceutics16030397 - 14 Mar 2024
Cited by 1 | Viewed by 1742
Abstract
Background: Although its immunomodulatory properties make thymopentin (TP5) appealing, its rapid metabolism and inactivation in the digestive system pose significant challenges for global scientists. PEGylated niosomal nanocarriers are hypothesized to improve the physicochemical stability of TP5, and to enhance its intestinal permeability for [...] Read more.
Background: Although its immunomodulatory properties make thymopentin (TP5) appealing, its rapid metabolism and inactivation in the digestive system pose significant challenges for global scientists. PEGylated niosomal nanocarriers are hypothesized to improve the physicochemical stability of TP5, and to enhance its intestinal permeability for oral administration. Methods: TP5-loaded PEGylated niosomes were fabricated using the thin film hydration method. Co-cultured Caco-2 and HT29 cells with different ratios were screened as in vitro intestinal models. The cytotoxicity of TP5 and its formulations were evaluated using an MTT assay. The cellular uptake and transport studies were investigated in the absence or presence of variable inhibitors or enhancers, and their mechanisms were explored. Results and Discussion: All TP5 solutions and their niosomal formulations were nontoxic to Caco-2 and HT-29 cells. The uptake of TP5-PEG-niosomes by cells relied on active endocytosis, exhibiting dependence on time, energy, and concentration, which has the potential to significantly enhance its cellular uptake compared to TP5 in solution. Nevertheless, cellular transport rates were similar between TP5 in solution and its niosomal groups. The cellular transport of TP5 in solution was carried out mainly through MRP5 endocytosis and a passive pathway and effluxed by MRP5 transporters, while that of TP5-niosomes and TP5-PEG-niosomes was carried out through adsorptive- and clathrin-mediated endocytosis requiring energy. The permeability and transport rate was further enhanced when EDTA and sodium taurocholate were used as the penetration enhancers. Conclusions: This research has illustrated that PEG-niosomes were able to enhance the cellular uptake and maintain the cellular transport of TP5. This study also shows this formulation’s potential to serve as an effective carrier for improving the oral delivery of peptides. Full article
(This article belongs to the Special Issue Advances in Oral Administration)
Show Figures

Figure 1

Figure 1
<p>The chemical structure of TP5 and the amino acid sequences of thymopoietin and thymopentin.</p>
Full article ">Figure 2
<p>Schematic diagrams of transcellular transport and different transporters in Caco-2 and HT-29 co-cultured cell monolayers.</p>
Full article ">Figure 3
<p>The micrographs of Caco-2 and HT-29 cells after 80% confluence with ratios of (<b>a</b>) 1:0, (<b>b</b>) 0:1, (<b>c</b>) 1:1, (<b>d</b>) 1:2, (<b>e</b>) 1:3, (<b>f</b>) 3:1, (<b>g</b>) 5:1, (<b>h</b>) 7:1, and (<b>i</b>) 9:1.</p>
Full article ">Figure 4
<p>The micrographs of Caco-2 and HT-29 cells after 72 h, (<b>a</b>) Caco-2 and HT-29 cells without any TP5 formulations; (<b>b</b>) Caco-2 and HT-29 cells treated with TP5 solution; (<b>c</b>) Caco-2 and HT-29 cells treated with blank PEG-niosomes suspension; (<b>d</b>) Caco-2 and HT-29 cells treated with TP5-PEG-niosomes.</p>
Full article ">Figure 5
<p>MTT viability assay showing the effect of TP5 formulations on co-cultured Caco-2 and HT-29 cells. (<b>a</b>) Effect of increasing TP5 concentrations in solution; (<b>b</b>) effect of increasing PEG-TP5-niosome concentrations. Cells were incubated with drug at 37 °C for 24, 48, and 72 h (<span class="html-italic">p</span> value ˂ 0.05 * and 0.01 ** to control) (Mean ± S.D., <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 6
<p>Fluorescence microscopy images of Caco-2 and HT-29 cells treated with FITC-TP5 (green) at 0.5, 1, 2, and 3 h in contrast to cell nuclei (blue), cytoplasm (red), and merged (green, blue, and red) (magnification = 500×).</p>
Full article ">Figure 7
<p>Confocal laser scanning microscopy 3D cross-section images of Caco-2 and HT-29 cells treated with FITC-TP5 (green) at 0.5, 1, 2, and 3 h in contrast to cell nuclei (blue), cytoplasm (red), and merged (green, blue, and red) (magnification = 500×).</p>
Full article ">Figure 8
<p>Fluorescence microscopy images of Caco-2 and HT-29 cells treated with FITC-TP5-PEG-niosomes (green) at 0.5, 1, 2, 3, 4, and 5 h in contrast to cell nuclei (blue), cytoplasm (red), and merged (green, blue, and red) (magnification = 500×).</p>
Full article ">Figure 9
<p>Confocal laser scanning microscopy 3D cross-section images of Caco-2 and HT-29 cells treated with FITC-TP5-PEG-niosomes (green) at 0.5, 1, 2, 3, 4, and 5 h in contrast to cell nuclei (blue), cytoplasm (red), and merged (green, blue, and red) (magnification = 500×).</p>
Full article ">Figure 10
<p>(<b>a</b>) Fluorescence-intensity-derived cellular uptake of Caco-2 and HT-29 cells treated with FITC-TP5-PEG-niosomes (green) at 0.5, 1, 2, 3, 4, and 5 h; (<b>b</b>) HPLC and BCA quantitative cellular uptake of Caco-2 and HT-29 cells treated with FITC-TP5-PEG-niosomes (green) at 0.5, 1, 2, 3, 4, and 5 h (Mean ± S.D., <span class="html-italic">n</span> = 3) (0.05 * and 0.01 ** &lt; 0.05).</p>
Full article ">Figure 11
<p>(<b>a</b>) The effect of TP5 concentration on TP5 cellular uptake values; (<b>b</b>) the effect of niosomes’ concentration on TP5 cellular uptake values (mean ± S.D., <span class="html-italic">n</span> = 5).</p>
Full article ">Figure 12
<p>The effect of inhibitors and low temperature on TP5 and its formulation cellular uptake for 4 h on Caco-2 and HT-29 co-cultured cell monolayers (Mean ± S.D., <span class="html-italic">n</span> = 5; *** <span class="html-italic">p</span> &lt; 0.005, ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 comparing to control).</p>
Full article ">Figure 13
<p>(<b>a</b>) Transepithelial transport of cumulative amount of TP5 from TP5 solution, TP5-niosomes, and TP5-PEG-niosomes; (<b>b</b>) transepithelial transport of flux rate of TP5 from TP5 solution, TP5-niosomes, and TP5-PEG-niosomes (Mean ± S.D., <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 14
<p>(<b>a</b>) Effect of treatment concentrations on the Caco-2 and HT-29 cells’ viability. Cells were incubated with different transporters, inhibitors, and enhancers at 37 °C for 72 h (Mean ± S.D., <span class="html-italic">n</span> = 6); (<b>b</b>) TP5 cumulative amount in the absence or presence of variable inhibitors and penetration enhancers for 6 h transport study at 37 °C (Mean ± S.D., <span class="html-italic">n</span> = 6).</p>
Full article ">Figure 15
<p>(<b>a</b>) TP5 cumulative amount with or without niosomes in the absence or presence of variable inhibitors and penetration enhancers for 6 h transport study at 37 °C (Mean ± S.D., <span class="html-italic">n</span> = 6); (<b>b</b>) TP5 cumulative amount with or without PEG-niosomes in the absence or presence of variable inhibitors and penetration enhancers for 6 h transport study at 37 °C (Mean ± S.D., <span class="html-italic">n</span> = 6).</p>
Full article ">
14 pages, 9840 KiB  
Article
The Protective Effects of Water-Soluble Alginic Acid on the N-Terminal of Thymopentin
by Haiyu Ji, Yuting Fan, Xiaoji Gao, Youshun Gong, Keyao Dai, Zhenhua Wang, Bo Xu and Juan Yu
Molecules 2023, 28(18), 6445; https://doi.org/10.3390/molecules28186445 - 5 Sep 2023
Cited by 5 | Viewed by 1724
Abstract
Thymopentin (TP5) has exhibited strong antitumor and immunomodulatory effects in vivo. However, the polypeptide is rapidly degraded by protease and aminopeptidase within a minute at the N-terminal of TP5, resulting in severe limitations for further practical applications. In this study, the protective effects [...] Read more.
Thymopentin (TP5) has exhibited strong antitumor and immunomodulatory effects in vivo. However, the polypeptide is rapidly degraded by protease and aminopeptidase within a minute at the N-terminal of TP5, resulting in severe limitations for further practical applications. In this study, the protective effects of water-soluble alginic acid (WSAA) on the N-terminal of TP5 were investigated by establishing an H22 tumor-bearing mice model and determining thymus, spleen, and liver indices, immune cells activities, TNF-α, IFN-γ, IL-2, and IL-4 levels, and cell cycle distributions. The results demonstrated that WSAA+TP5 groups exhibited the obvious advantages of the individual treatments and showed superior antitumor effects on H22 tumor-bearing mice by effectively protecting the immune organs, activating CD4+ T cells and CD19+ B cells, and promoting immune-related cytokines secretions, finally resulting in the high apoptotic rates of H22 cells through arresting them in S phase. These data suggest that WSAA could effectively protect the N-terminal of TP5, thereby improving its antitumor and immunoregulatory activities, which indicates that WSAA has the potential to be applied in patients bearing cancer or immune deficiency diseases as a novel immunologic adjuvant. Full article
(This article belongs to the Special Issue Recent Advances in Bioactive Macromolecules from Natural Plants)
Show Figures

Figure 1

Figure 1
<p>Preparation technology (<b>A</b>), UV spectrum (<b>B</b>), and HPGPC profile (<b>C</b>) of WSAA.</p>
Full article ">Figure 2
<p>Schematic diagrams of TP5 degradation (<b>A</b>) and WSAA protecting TP5 (<b>B</b>).</p>
Full article ">Figure 3
<p>Tumor inhibitory rates (<b>A</b>) and organs indices (<b>B</b>) of mice in these groups. Note: α, <span class="html-italic">p</span> &lt; 0.05 compared with blank group; β, <span class="html-italic">p</span> &lt; 0.05 compared with model group.</p>
Full article ">Figure 4
<p>Distributions and proportions of CD3<sup>+</sup> T and CD19<sup>+</sup> B lymphocytes and CD4<sup>+</sup> T and CD8<sup>+</sup> T cells in peripheral bloods.</p>
Full article ">Figure 5
<p>Proliferative activities of splenic T and B lymphocytes (<b>A</b>) and antibodies expressions (<b>B</b>) in sera of mice in these groups. Note: α, <span class="html-italic">p</span> &lt; 0.05 compared with blank group; β, <span class="html-italic">p</span> &lt; 0.05 compared with model group.</p>
Full article ">Figure 6
<p>The activities of peritoneal macrophages and splenic NK cells in mice of these groups. Note: α, <span class="html-italic">p</span> &lt; 0.05 compared with blank group; β, <span class="html-italic">p</span> &lt; 0.05 compared with model group.</p>
Full article ">Figure 7
<p>Expression levels of IL-2 and IL-4 (<b>A</b>) and TNF-α and IFN-γ (<b>B</b>) in mice sera. Note: α, <span class="html-italic">p</span> &lt; 0.05 compared with blank group; β, <span class="html-italic">p</span> &lt; 0.05 compared with model group.</p>
Full article ">Figure 8
<p>Cell cycle distributions and apoptosis rates of H22 solid tumor cells in mice of these groups.</p>
Full article ">
18 pages, 10337 KiB  
Article
Altered Membrane Expression and Function of CD11b Play a Role in the Immunosuppressive Effects of Morphine on Macrophages at the Nanomolar Level
by Peng-Cheng Yu, Cui-Yun Hao, Ying-Zhe Fan, Di Liu, Yi-Fan Qiao, Jia-Bao Yao, Chang-Zhu Li and Ye Yu
Pharmaceuticals 2023, 16(2), 282; https://doi.org/10.3390/ph16020282 - 13 Feb 2023
Cited by 7 | Viewed by 3069
Abstract
Morphine, one of the most efficacious analgesics, is effective in severe pain, especially in patients with concomitant painful cancers. The clinical use of morphine may be accompanied by increased immunosuppression, susceptibility to infection and postoperative tumor metastatic recurrence, and the specific mechanisms and [...] Read more.
Morphine, one of the most efficacious analgesics, is effective in severe pain, especially in patients with concomitant painful cancers. The clinical use of morphine may be accompanied by increased immunosuppression, susceptibility to infection and postoperative tumor metastatic recurrence, and the specific mechanisms and clinical strategies to alleviate this suppression remain to be investigated. Expression of CD11b is closely associated with the macrophage phagocytosis of xenobiotic particles, bacteria or tumor cells. Here, we find that morphine at 0.1–10 nM levels inhibited CD11b expression and function on macrophages via a μ-opioid receptor (MOR)-dependent mechanism, thereby reducing macrophage phagocytosis of tumor cells, a process that can be reversed by thymopentin (TP5), a commonly used immune-enhancing adjuvant in clinical practice. By knocking down or overexpressing MOR on macrophages and using naloxone, an antagonist of the MOR receptor, and LA1, a molecule that promotes macrophage CD11b activation, we suggest that morphine may regulate macrophage phagocytosis by inhibiting the surface expression and function of macrophage CD11b through the membrane expression and activation of MOR. The CD47/SIRPα axis, which is engaged in macrophage-tumor immune escape, was not significantly affected by morphine. Notably, TP5, when combined with morphine, reversed the inhibition of macrophage phagocytosis by morphine through mechanisms that promote membrane expression of CD11b and modulate its downstream signaling (e.g., NOS2, IFNG, IL1B and TNFA, as well as AGR1, PDGFB, IL6, STAT3, and MYC). Thus, altered membrane expression and function of CD11b may mediate the inhibition of macrophage phagocytosis by therapeutic doses of morphine, and the reversal of this process by TP5 may provide an effective palliative option for clinical immunosuppression by morphine. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Nanomolar levels of morphine inhibit phagocytosis of A549 cells by THP-1-derived macrophages (<b>A</b>,<b>B</b>) Representative fluorescence micrographs of phagocytic activity (<b>A</b>) and statistical analysis of the mean fluorescence intensity signal in confocal images (<b>B</b>). Scale bar = 50 μm. (<b>C</b>) Flow cytometric quantification of macrophage phagocytosis under different concentrations of morphine culture (0, 0.001, 0.01, 0.1, 1, 10, 100, 1000 and 10,000 nM). (<b>D</b>–<b>G</b>) A549 cells and macrophages being labeled and analyzed for CD47- and SIRPα-expression, respectively, (<b>D</b>,<b>F</b>) by flow cytometry, and pooled data of cell surface markers of CD47 (<b>E</b>) and SIRPα (<b>G</b>). (<b>H</b>) Viability of A549 cells measured by MTT assay at different concentrations of morphine. (<b>I</b>) Macrophages viability measured by MTT assay at different concentrations of morphine (n = 3–4 independent experiments). All data are expressed as mean ± SEM.; ** <span class="html-italic">p</span> &lt; 0.01 versus control, unpaired <span class="html-italic">t</span>-test (<b>B</b>,<b>E</b>,<b>G</b>), one-way ANOVA with Dunnett’s post-hoc test (<b>H</b>,<b>I</b>), (<b>H</b>), (F (7, 16) = 0.4882, <span class="html-italic">p</span> = 0.8295), (<b>I</b>), (F (7, 24) = 1.563, <span class="html-italic">p</span> = 0.1945. ns, not significant.</p>
Full article ">Figure 2
<p>Morphine reduces the gene expression of CD11b and μ-opioid receptor (MOR) on macrophages. (<b>A</b>,<b>B</b>) RT-qPCR analysis of relative mRNA expression of CD11b (<b>A</b>) and OPRM (<b>B</b>) in macrophages. (<b>C</b>–<b>E</b>) Macrophages being labeled and analyzed for CD11b-expression (<b>C</b>) by flow cytometry, and pooled data of cell surface marker CD11b (<b>D</b>,<b>E</b>) (n = 3 independent experiments, and the solid line is fitted to <span class="html-italic">hill 1</span> equation). All data are expressed as mean ± SEM.; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 versus control, one-way ANOVA with Dunnett’s post-hoc test, (<b>A</b>), (F (5, 18) = 17.83, <span class="html-italic">p</span> &lt; 0.0001), (<b>B</b>), (F (5, 18) = 9.865, <span class="html-italic">p</span> = 0.0001), (<b>D</b>), (F (4, 10) = 13.14, <span class="html-italic">p</span> = 0.0005); ns, not significant.</p>
Full article ">Figure 3
<p>Leukadherin-1 (LA1) promotes the activation of CD11b on macrophages and alleviates the inhibitory effect of morphine on macrophage phagocytosis. (<b>A</b>,<b>B</b>) RT-qPCR analysis of relative mRNA expression of anti-inflammatory and pro-inflammatory factors ARG1, PDGFB, IL6, STAT3 and MYC (<b>A</b>), as well as NOS2, IFNG, IL1B and TNFA (<b>B</b>) in macrophages. (<b>C</b>,<b>D</b>) Flow cytometry analysis of CD11b surface expression on macrophages (<b>C</b>), and pooled data of cell surface marker CD11b (<b>D</b>). (<b>E</b>,<b>F</b>) Representative fluorescence photomicrographs of the phagocytic activity (<b>E</b>) and statistical analysis of the mean fluorescence intensity signal in confocal images (<b>F</b>). Scale bar = 50 μm. (<b>G</b>,<b>H</b>) Quantitative flow cytometry analysis of macrophage phagocytosis after incubation with different concentrations of LA1 (4 and 8 μM) and morphine (10 nM) (n = 3 independent experiments). All data are expressed as mean ± SEM.; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 versus control, one-way ANOVA with Dunnett’s post-hoc test, (<b>D</b>), (F (5, 12) = 42.96, <span class="html-italic">p</span> &lt; 0.05), (<b>F</b>), (F (3, 8) = 20.34, <span class="html-italic">p</span> = 0.0004), (<b>H</b>), (F (3, 12) = 39.07, <span class="html-italic">p</span> &lt; 0.001); # <span class="html-italic">p</span> &lt; 0.05 and ### <span class="html-italic">p</span> &lt; 0.001 versus control; ns, not significant.</p>
Full article ">Figure 4
<p>Surface expression of MOR and CD11b on macrophages is interrelated. (<b>A</b>). Flow cytometry analysis of MOR surface expression on macrophages after cells were incubated with different concentrations of morphine (the solid line is fitted to <span class="html-italic">hill 1</span> equation). (<b>B</b>) RT-qPCR analysis of relative mRNA expressions of CD11b and OPRM in macrophages. In transfected THP-1-derived macrophages, siRNA-mediated knockdown of OPRM also decreased the gene expression of both CD11b. (<b>C</b>,<b>D</b>) Flow cytometric analysis of siRNA-mediated alterations in CD11b expression on the surface of macrophages (<b>C</b>) and pooled data of cell surface marker CD11b (<b>D</b>). (<b>E</b>) Plasmid-mediated overexpression of OPRM in transfected THP-1-derived macrophages. (<b>F</b>,<b>G</b>) Flow cytometric analysis of the effect of morphine on CD11b surface expression in OPRM-overexpressing macrophages (<b>F</b>), and pooled data for the cell surface marker CD11b (<b>G</b>) (n = 3–4 independent experiments). All data are expressed as mean ± SEM.; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 versus control, unpaired <span class="html-italic">t</span>-test (<b>B</b>,<b>D</b>,<b>E</b>), one-way ANOVA with Dunnett’s post-hoc test (<b>G</b>), (F (3, 8) = 110.1, <span class="html-italic">p</span> &lt; 0.001); ### <span class="html-italic">p</span> &lt; 0.001 versus control; ns, not significant.</p>
Full article ">Figure 5
<p>The MOR antagonist naloxone (NLX) reverses the inhibitory effect of morphine on membrane expression and function of CD11b on macrophages. (<b>A</b>,<b>B</b>) Effect (<b>A</b>) and pooled data (<b>B</b>) of NLX (10 μM) on CD11b surface expression in macrophages treated with morphine (10 nM). Macrophages were labeled and analyzed for CD11b-expression by flow cytometry. (<b>C</b>,<b>D</b>) Representative fluorescence photomicrographs of the phagocytic activity (<b>C</b>) and statistical analysis of the mean fluorescence intensity signal in confocal images (<b>D</b>). Scale bar = 50 μm. (<b>E</b>,<b>F</b>) Quantitative flow cytometric analysis of macrophage phagocytosis cultured with different concentrations of NLX (10 and 20 μM) and morphine (10 nM) (<b>E</b>), and the pooled data (<b>F</b>). (<b>G</b>,<b>H</b>) RT-qPCR analysis of relative mRNA expression of anti-inflammatory factors ARG1, PDGFB, IL6, STAT3 and MYC (<b>G</b>), and pro-inflammatory factors of NOS2, IFNG, IL1B and TNFA (<b>H</b>) in macrophages (n = 3 or 4 independent experiments). All data are expressed as mean ± SEM.; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 versus control, one-way ANOVA with Dunnett’s post-hoc test, (<b>B</b>), (F (2, 9) = 47.07, <span class="html-italic">p</span> &lt; 0.05), (<b>D</b>), (F (3, 8) = 54.22, <span class="html-italic">p</span> &lt; 0.001), (<b>F</b>), (F (3, 12) = 31.64, <span class="html-italic">p</span> &lt; 0.01); # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 versus control; ns, not significant.</p>
Full article ">Figure 6
<p>TP5 promotes the surface expression and function of CD11b on macrophages. (<b>A</b>,<b>B</b>) RT-qPCR analysis of relative mRNA expression of OPRM (<b>A</b>) and CD11b (<b>B</b>) in macrophages incubated with different treatments of TP5 (0, 50, 100 and 200 μM). (<b>C</b>,<b>D</b>) Macrophages labeled and analyzed for CD11b-expression (<b>C</b>) by flow cytometry, as well as pooled data for the cell surface marker CD11b (<b>D</b>). (<b>E</b>,<b>F</b>) Quantitative flow cytometry analysis (<b>E</b>) of macrophages phagocytosis after incubation with different treatments of TP5 (50, 100, 200 μM) and morphine (10 nM), as well as the pooled data (<b>F</b>). (<b>G</b>,<b>H</b>) Flow cytometry analysis (<b>G</b>) and pooled data (<b>H</b>) of CD11b surface expression in macrophages incubated with different treatments of TP5 (50, 100, 200 μM) and morphine (10 nM). (<b>I</b>,<b>J</b>) RT-qPCR analysis of relative mRNA expression of OPRM (<b>I</b>) and CD11b (<b>J</b>) in macrophages. (<b>K</b>,<b>L</b>) RT-qPCR analysis of relative mRNA expression of anti-inflammatory factors ARG1, PDGFB, IL6, STAT3 and MYC (<b>K</b>), and pro- inflammatory factors NOS2, IFNG, IL1B and TNFA (<b>L</b>) in macrophages (n = 3–5 independent experiments). All data are expressed as mean ± SEM.; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 versus control, one-way ANOVA with Dunnett’s post-hoc test, (<b>A</b>), (F (2, 6) = 5.212, <span class="html-italic">p</span> = 0.0487), (<b>B</b>), (F (2, 9) = 20.07, <span class="html-italic">p</span> = 0.0005), (<b>D</b>), (F (3, 8) = 7.761, <span class="html-italic">p</span> = 0.0094), (<b>F</b>), (F (2, 9) = 14.65, <span class="html-italic">p</span> = 0.0015), (<b>H</b>), (F (4, 10) = 14.49, <span class="html-italic">p</span> = 0.0004), (<b>I</b>), (F (3, 12) = 29.04, <span class="html-italic">p</span> &lt; 0.0001), (<b>J</b>), (F (3, 16) = 11.11, <span class="html-italic">p</span> = 0.0003); # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 versus control; ns, not significant.</p>
Full article ">Figure 7
<p>TP5 reverses the inhibitory effect of morphine on macrophage phagocytosis. (<b>A</b>,<b>B</b>) Representative fluorescence photomicrographs of the phagocytic activity (<b>A</b>) and statistical analysis of the mean fluorescence intensity signal in confocal images (<b>B</b>). Scale bar = 50 μm. (<b>C</b>,<b>D</b>) Quantitative flow cytometric analysis (<b>C</b>) of macrophage phagocytosis incubated with morphine (10 nM) and TP5 (200 μM), as well as pooled data (<b>D</b>). (<b>E</b>,<b>F</b>) Flow cytometry analysis (<b>E</b>) of CD11b surface expression and pooled date (<b>F</b>) on macrophages treated with morphine (10 nM) and TP5 (200 μM) or in combination with the AchR inhibitor D-Tubocurarine chloride pentahydrate (TUB, 150 and 200 μg/mL) (n = 3–4 independent experiments). All data are expressed as mean ± SEM.; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. control, one-way ANOVA with Dunnett’s post-hoc test, (<b>B</b>), (F (4, 10) = 27.15, <span class="html-italic">p</span> &lt; 0.0001), (<b>D</b>), (F (2, 9) = 27.44, <span class="html-italic">p</span> = 0.0001), (<b>F</b>), (F (4, 10) = 61.76, <span class="html-italic">p</span> &lt; 0.01); # <span class="html-italic">p</span> &lt; 0.05 and ### <span class="html-italic">p</span> &lt; 0.001 versus control; ns, not significant.</p>
Full article ">
23 pages, 6487 KiB  
Article
Novel Hybrid Peptide Cathelicidin 2 (1-13)-Thymopentin (TP5) and Its Derived Peptides with Effective Antibacterial, Antibiofilm, and Anti-Adhesion Activities
by He-Nan Guo, Yu-Cui Tong, Hui-Li Wang, Jing Zhang, Zhong-Xuan Li, Zaheer Abbas, Tian-Tian Yang, Meng-Yao Liu, Pei-Yao Chen, Zheng-Chang Hua, Xiao-Na Yan, Qiang Cheng, Marhaba Ahmat, Jun-Yong Wang, Lu-Lu Zhang, Xu-Biao Wei, Xiu-Dong Liao and Ri-Jun Zhang
Int. J. Mol. Sci. 2021, 22(21), 11681; https://doi.org/10.3390/ijms222111681 - 28 Oct 2021
Cited by 7 | Viewed by 3062
Abstract
The increasing numbers of infections caused by multidrug-resistant (MDR) pathogens highlight the urgent need for new alternatives to conventional antibiotics. Antimicrobial peptides have the potential to be promising alternatives to antibiotics because of their effective bactericidal activity and highly selective toxicity. The present [...] Read more.
The increasing numbers of infections caused by multidrug-resistant (MDR) pathogens highlight the urgent need for new alternatives to conventional antibiotics. Antimicrobial peptides have the potential to be promising alternatives to antibiotics because of their effective bactericidal activity and highly selective toxicity. The present study was conducted to investigate the antibacterial, antibiofilm, and anti-adhesion activities of different CTP peptides (CTP: the original hybrid peptide cathelicidin 2 (1-13)-thymopentin (TP5); CTP-NH2: C-terminal amidated derivative of cathelicidin 2 (1-13)-TP5; CTPQ: glutamine added at the C-terminus of cathelicidin 2 (1-13)-TP5) by determining the minimal inhibitory concentrations (MICs), minimal bactericidal concentrations (MBCs), propidium iodide uptake, and analysis by scanning electron microscopy, transmission electron microscopy, and confocal laser scanning microscopy). The results showed that CTPs had broad-spectrum antibacterial activity against different gram-positive and gram-negative bacteria, with MICs against the tested strains varying from 2 to 64 μg/mL. CTPs at the MBC (2 × MIC 64 μg/mL) showed strong bactericidal effects on a standard methicillin-resistant Staphylococcus aureus strain ATCC 43300 after co-incubation for 6 h through disruption of the bacterial membrane. In addition, CTPs at 2 × MIC also displayed effective inhibition activity of several S. aureus strains with a 40–90% decrease in biofilm formation by killing the bacteria embedded in the biofilms. CTPs had low cytotoxicity on the intestinal porcine epithelial cell line (IPEC-J2) and could significantly decrease the rate of adhesion of S. aureus ATCC 43300 on IPEC-J2 cells. The current study proved that CTPs have effective antibacterial, antibiofilm, and anti-adhesion activities. Overall, this study contributes to our understanding of the possible antibacterial and antibiofilm mechanisms of CTPs, which might be an effective anti-MDR drug candidate. Full article
Show Figures

Figure 1

Figure 1
<p>Helical wheel diagram of peptides (<b>A</b>) CTP (cathelicidin 2 (CATH2) and thymopentin (TP5)) and (<b>B</b>) CTPQ (addition of glutamine at the C-terminal of cathelicidin 2 (CATH2) and thymopentin (TP5)) using a website (<a href="https://www.bioinformatics.nl/cgi-bin/emboss/pepwheel" target="_blank">https://www.bioinformatics.nl/cgi-bin/emboss/pepwheel</a> (accessed on 12 February 2021)). The hydrophobic residues were shown in green, and the hydrophilic residues were shown in red. In addition, the red line separated the hydrophilic and hydrophobic residues.</p>
Full article ">Figure 2
<p>(<b>A</b>) The inhibition zone of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs at 1, 2, and 4 × MIC (32, 64 and 128 μg/mL) and (<b>B</b>) their diameters. Data were given as the mean value ± SD from three biological replicates. <sup>a,b</sup>: Different lowercase letters mean significantly different (<span class="html-italic">p</span> &lt; 0.05) values were expressed as mean ± SEM; NS: not significant (<span class="html-italic">p</span> &gt; 0.05) (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 3
<p>Time-kill curves of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH<sub>2</sub>, and CTPQ) at the MIC (32 μg/mL) and 2 × MIC (64 μg/mL). Data were given as the mean value ± SD from three biological replicates.</p>
Full article ">Figure 4
<p>Spot plates of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH2, and CTPQ) treatment at the MIC (32 μg/mL) and the MBC (64 μg/mL) for 6 h. 10<sup>0</sup>–10<sup>5</sup>: dilution ratio of bacteria suspension.</p>
Full article ">Figure 5
<p>CRA plate of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH<sub>2</sub>, and CTPQ) treatment at the MIC (32 μg/mL) and the MBC (64 μg/mL) for 9 h.</p>
Full article ">Figure 6
<p>PI uptake of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH2, and CTPQ) treatment at the MIC (32 μg/mL) and the MBC (64 μg/mL). Results are expressed in arbitrary units (a. u.).</p>
Full article ">Figure 7
<p>SEM assay of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH<sub>2</sub>, and CTPQ) at the MIC (32 μg/mL).</p>
Full article ">Figure 8
<p>TEM assay of <span class="html-italic">S. aureus</span> ATCC 43300 treated with CTPs (CTP, CTP-NH2, and CTPQ) at the MIC (32 μg/mL).</p>
Full article ">Figure 9
<p>Antibiofilm activity of CTPs (CTP, CTP-NH2, and CTPQ) against (<b>A</b>) <span class="html-italic">P. aeruginosa</span> ATCC 9027, (<b>B</b>) <span class="html-italic">P. aeruginosa</span> ATCC 27853, (<b>C</b>) <span class="html-italic">S. aureus</span> ATCC 6538, (<b>D</b>) <span class="html-italic">S. aureus</span> ATCC 25923, (<b>E</b>) <span class="html-italic">S. aureus</span> CVCC 1882, and (<b>F</b>) <span class="html-italic">S. aureus</span> ATCC 43300.</p>
Full article ">Figure 10
<p>Optical microscope observation of <span class="html-italic">S. aureus</span> ATCC 43300 biofilm treated by CTPs (CTP, CTP-NH<sub>2</sub>, and CTPQ).</p>
Full article ">Figure 11
<p>CLSM assay of <span class="html-italic">S. aureus</span> ATCC 43300 biofilm treated by CTPs (CTP, CTP-NH<sub>2</sub>, and CTPQ) at the MBC (64 μg/mL) with 40 × oil lens.</p>
Full article ">Figure 12
<p>The qRT-PCR results of <span class="html-italic">S. aureus</span> ATCC 43300 genes related to biofilm treated with CTPs. NS: not significant (<span class="html-italic">p</span> &gt; 0.05) (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 13
<p>(<b>A</b>) Cell viability of intestinal porcine epithelial cell line (IPEC-J2) upon exposure to CTPs and (<b>B</b>) inhibitory effect of CTPs against the adherence of <span class="html-italic">S. aureus</span> ATCC 43300 to IPEC-J2 cells. <sup>a–d</sup>: Different lowercase letters mean significantly different (<span class="html-italic">p</span> &lt; 0.05) values were expressed as mean ± SEM (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 14
<p>The hypothesized model of antibacterial and antibiofilm effects of CTPs against <span class="html-italic">S. aureus</span> ATCC 43300.</p>
Full article ">
Back to TopTop