[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
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
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (127)

Search Parameters:
Keywords = L-Selectin

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
9 pages, 273 KiB  
Communication
Compartmentalization of the Inflammatory Response in the Pericardial Cavity in Patients Undergoing Cardiac Surgery
by Mohammad M. El-Diasty, Javier Rodríguez, Luis Pérez, Souhayla Souaf, Sonia Eiras and Angel L. Fernández
Int. J. Mol. Sci. 2024, 25(24), 13720; https://doi.org/10.3390/ijms252413720 - 23 Dec 2024
Viewed by 384
Abstract
The systemic inflammatory response after cardiopulmonary bypass has been widely studied. However, there is a paucity of studies that focus on the local inflammatory changes that occur in the pericardial cavity. The purpose of this study is to assess the inflammatory mediators in [...] Read more.
The systemic inflammatory response after cardiopulmonary bypass has been widely studied. However, there is a paucity of studies that focus on the local inflammatory changes that occur in the pericardial cavity. The purpose of this study is to assess the inflammatory mediators in the pericardial fluid of patients undergoing cardiac surgery. We conducted a prospective cohort study on patients undergoing aortic valve replacement. Pericardial fluid and peripheral venous blood samples were collected after the opening of the pericardium. Additional samples were obtained from peripheral blood and the pericardial fluid shed through mediastinal drains 24 and 48 h after surgery. Levels of interleukin 1α (IL-1α), interleukin 1β (IL-1β), interleukin 2 (IL-2), interleukin 4 (IL-4), interleukin 6 (IL-6), interleukin 8 (IL-8), interleukin 10 (IL-10), tumor necrosis factor-α (TNF-α), interferon-γ (IFN-γ), vascular endothelial growth factor (VEGF), monocyte chemotactic protein-1 (MCP-1), epidermal growth factor (EGF), soluble E-selectin, L-selectin, P-selectin, intercellular adhesion molecule-1 (ICAM-1), and vascular cell adhesion molecule-1 (VCAM-1) were determined in all pericardial fluid and serum samples. A total of 45 patients with a mean age of 74 years were included, of which 66% were males. Serum levels of IL-6, IL-8, and MCP-1 were significantly increased at 24 and 48 h after surgery. No significant changes were observed in the serum levels of the remaining mediators. A significant increase of postoperative pericardial fluid levels of IL-1α, IL-1β, IL-6, IL-8, IL-10, IFN-γ, VEGF, MCP-1, VCAM-1, and P-selectin was observed at 24 and 48 h after surgery. There is a robust systemic and pericardial inflammatory response after cardiac surgery on cardiopulmonary bypass. However, postoperative pericardial inflammatory activity shows a distinct pattern and is more marked than at the systemic level. These findings suggest that there is a compartmentalization of the inflammatory response within the pericardial cavity after cardiac surgery. Full article
(This article belongs to the Section Molecular Immunology)
31 pages, 3608 KiB  
Article
Reversal of Endothelial Cell Anergy by T Cell-Engaging Bispecific Antibodies
by Márcia Gonçalves, Karsten M. Warwas, Marten Meyer, Reinhard Schwartz-Albiez, Nadja Bulbuc, Inka Zörnig, Dirk Jäger and Frank Momburg
Cancers 2024, 16(24), 4251; https://doi.org/10.3390/cancers16244251 - 20 Dec 2024
Viewed by 358
Abstract
Objectives: Reduced expression of adhesion molecules in tumor vasculature can limit infiltration of effector T cells. To improve T cell adhesion to tumor endothelial cell (EC) antigens and enhance transendothelial migration, we developed bispecific, T-cell engaging antibodies (bsAb) that activate T cells after [...] Read more.
Objectives: Reduced expression of adhesion molecules in tumor vasculature can limit infiltration of effector T cells. To improve T cell adhesion to tumor endothelial cell (EC) antigens and enhance transendothelial migration, we developed bispecific, T-cell engaging antibodies (bsAb) that activate T cells after cross-linking with EC cell surface antigens. Methods: Recombinant T-cell stimulatory anti-VEGFR2–anti-CD3 and costimulatory anti-TIE2–anti-CD28 or anti-PD-L1–anti-CD28 bsAb were engineered and expressed. Primary lines of human umbilical vein endothelial cells (HUVEC) that constitutively express VEGFR2 and TIE2 growth factor receptors and PD-L1, but very low levels of adhesion molecules, served as models for anergic tumor EC. Results: In cocultures with HUVEC, anti-VEGFR2–anti-CD3 bsAb increased T cell binding and elicited rapid T cell activation. The release of proinflammatory cytokines TNF-α, IFN-γ, and IL-6 was greatly augmented by the addition of anti-TIE2–anti-CD28 or anti-PD-L1–anti-CD28 costimulatory bsAb. Concomitantly, T cell-released cytokines upregulated E-selectin, ICAM1, and VCAM1 adhesion molecules on HUVEC. HUVEC cultured in breast cancer cell-conditioned medium to mimic the influence of tumor-secreted factors were similarly activated by T cell-engaging bsAb. Migration of T cells in transwell assays was significantly increased by anti-VEGFR2–anti-CD3 bsAb. The combination with costimulatory anti-TIE2–anti-CD28 bsAb augmented activation and proliferation of migrated T cells and their cytotoxic capacity against spheroids of the MCF-7 breast cancer cell line seeded in the lower transwell chamber. Conclusions: T cells activated by anti-VEGFR2–anti-CD3 and costimulatory EC-targeting bsAb can reverse the energy of quiescent EC in vitro, resulting in improved T cell migration through an EC layer. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Binding of bsAb to CD3<sup>+</sup> T cells and to endothelial cells. (<b>A</b>) Schematic representation of the tetravalent bispecific bsAb format used in this work. A single chain variable fragment (scFv) antibody 1 specific for tumor endothelium surface receptors is linked to the hinge–CH2–CH3 domains of hIgG1 via a flexible glycine-serine linker (GSL). The Fc domain contains multiple point mutations to abrogate the Fc receptor and complement binding. At the C-terminal end of the CH3 domain, a Strep-tag II is added for immunoaffinity purification, followed by an scFv antibody 2 recognizing either CD3ε or CD28. (<b>B</b>) SDS-PAGE analysis (10%) and Coomassie staining of purified bispecific antibodies under non-reducing and reducing conditions. (<b>C</b>) Freshly isolated T cells and (<b>D</b>) HUVEC were incubated with 5 mg/mL of produced constructs, αVEGFR2–αCD3ε, αTIE2–αCD28, αPD-L1–αCD28, and binding was detected by goat anti-human-IgG1-PE secondary antibody which was also used alone for a negative staining control (grey histograms).</p>
Full article ">Figure 2
<p>T cell activation induced by αEC bsAbs. Freshly isolated T cells were cocultured with HUVEC in the presence of different bsAb, αVEGFR2–αCD3ε (1 nM) alone or in combination with αTIE2–αCD28 (1 nM) or αPD-L1–αCD28 (1 nM) for 24 h. After three washing steps, firmly EC-bound T cells were collected, and the induction of activation surface markers (CD69, CD25, 4-1BB, and OX40) was studied on (<b>A</b>) CD4<sup>+</sup> and (<b>B</b>) CD8<sup>+</sup> T cells using flow cytometry. Data are presented as means of percentages of T cells expressing the indicated activation markers ± SEM from 6 independent experiments. (<b>C</b>) Supernatants from the 24 h coculture were collected, and TNF-α and IFN-γ secretion was quantified by sandwich ELISA. Data are presented as mean values ± SEM from 3 independent experiments. (<b>D</b>) T-cell cytotoxic effects in HUVEC cocultures were studied at the indicated time points (4 h–24 h) by quantification of LDH released to cell culture supernatants. Data is presented as mean ± SEM from 3 independent experiments. (<b>E</b>) Firm T cell binding to EC was quantified using precision counting beads. Data are presented as means of percentages of bound cells from input ± SEM from 3 independent experiments. Statistical analysis by one-way ANOVA test followed by Tukey’s multiple comparison test (<b>A</b>,<b>B</b>,<b>D</b>,<b>E</b>) or two-way ANOVA followed by Sidak’s multiple comparison test (<b>C</b>); ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Upregulation of adhesion molecules by HUVEC in the presence of bsAb-activated T cells. (<b>A</b>) After 24 h of HUVEC–T cell coculture in the presence of the indicated bsAb, EC was dissociated from plates, and expression of adhesion molecules E-selectin, VCAM1, and ICAM1 was evaluated by flow cytometry. (<b>B</b>) Supernatants from HUVEC–T cell cocultures were collected after 24 h and transferred to an HUVEC monolayer. Expression of adhesion molecules E-selectin, VCAM1, and ICAM by HUVEC was evaluated after 24 h by flow cytometry. Data are presented as mean values ± SEM from 3 independent experiments. Statistical analysis by one-way ANOVA followed by Tukey’s multiple comparison test; ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Rapid enhancement of T cell adhesion to HUVEC by αVEGFR2–αCD3ε bsAb. Freshly isolated T cells were cocultured with adherent HUVEC with or without bispecific antibodies αVEGFR2–αCD3ε (1 nM) alone or in combination with αTIE2–αCD28 (1 nM) or αPD-L1–αCD28 (1 nM) for 4 h. After extensive washing, firmly bound T cells were collected by cell dissociation buffer and analyzed by flow cytometry. (<b>A</b>) Percentage of firm T cell binding from T cell input number was quantified using precision counting beads. (<b>B</b>,<b>C</b>) CD69 expression, which was used as a T cell early activation marker, was assessed by flow cytometry for CD4<sup>+</sup> T cells (<b>B</b>) and CD8<sup>+</sup> T cells (<b>C</b>). (<b>D</b>) Upregulation of cell surface expression of adhesion molecules E-selectin, VCAM1, and ICAM1 on HUVEC after 4 h of coculture was measured by flow cytometry. Data are presented as mean ± SEM from 3 independent experiments. Statistical analysis was done by one-way ANOVA test followed by Tukey’s multiple comparison test; ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>T cell migration and activation are increased by αVEGFR2–αCD3ε and αTIE2–αCD28 bsAb. (<b>A</b>) Scheme of the 2-layer transwell model. 2.5 × 10<sup>4</sup> HUVEC were seeded in transwell inserts coated with 1% human fibronectin and left for 48 h until reaching a tight confluent monolayer. Purified CD3<sup>+</sup> T cells (0.5 × 10<sup>6</sup> cells) were added to the insert together with αVEGFR2–Fc (1 nM), αTIE2–αCD28 (1 nM) alone, αVEGFR2–αCD3ε alone (1 nM), αVEGFR2–αCD3ε + αTIE2–αCD28 (1 nM each), or left untreated for control (NT). After 24 h, cells were collected from the lower and upper chambers and migrated CD3<sup>+</sup> T cells (<b>B</b>) (<span class="html-italic">n</span> = 6), and non-migrated lymphocytes (<b>C</b>) (<span class="html-italic">n</span> = 6) were quantified by flow cytometry using counting beads. (<b>D</b>) Non-adherent cells from the insert were studied for cell death by uptake of ZombieAqua live/dead stain. CTV-labeled T cells and non-labeled HUVEC were analyzed separately (<span class="html-italic">n</span> = 5). The activation of migrated CD3<sup>+</sup>/CD4<sup>+</sup> (<b>E</b>) and CD3<sup>+</sup>/CD8<sup>+</sup> (<b>F</b>) subpopulations was analyzed by measuring expression of CD69, CD25, and 4-1BB by flow cytometry (<span class="html-italic">n</span> = 6). A gating scheme is shown in <a href="#app1-cancers-16-04251" class="html-app">Supplementary Figure S7C</a>. (<b>G</b>) Adherent HUVEC harvested from the insert membrane were analyzed for E-selectin (<span class="html-italic">n</span> = 6), VCAM1 (<span class="html-italic">n</span> = 5), and ICAM1 (<span class="html-italic">n</span> = 3) expression by flow cytometry. Data are presented as mean ± SEM from <span class="html-italic">n</span> independent experiments done in duplicates. Statistical analysis by one-way ANOVA test (<b>B</b>,<b>D</b>–<b>G</b>) or two-way ANOVA test (<b>C</b>) followed by Tukey’s multiple comparison test; ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Migrated T cells kill MCF-7 tumor spheroids in the presence of αTAA bsAb. (<b>A</b>) Scheme of the 3-layer transwell model. Transwell inserts were coated with 1% human fibronectin, and 2.5 × 10<sup>4</sup> HUVEC were seeded and left for 48 h until reaching a tight confluent monolayer. Purified CD3<sup>+</sup> T cells (0.5 × 10<sup>6</sup> cells) were added to the insert together with αVEGFR2–Fc (1 nM) for control, αTIE2–αCD28 (1 nM), αVEGFR2–αCD3ε (1 nM), or αVEGFR2–αCD3ε + αTIE2–αCD28 (1 nM each). Preformed MCF-7 spheroids were transferred to the lower chamber together with αEpCAM–αCD3 (10 nM) or αEpCAM–αCD28 (10 nM). After 24 h of migration, the inserts were removed, and migrated T cells were kept in culture with the MCF-7 spheroids for an additional 48 h. (<b>B</b>) CD4<sup>+</sup> and (<b>C</b>) CD8<sup>+</sup> T cell activation was analyzed by cell surface staining of migrated cells for CD69, CD25, 4-1BB, and OX40 (<span class="html-italic">n</span> = 5). (<b>D</b>) CD4<sup>+</sup> and CD8<sup>+</sup> T cell proliferation was evaluated by determining intracellular CTV dilution by flow cytometry (<span class="html-italic">n</span> = 4). (<b>E</b>) Cytotoxicity was assessed by measuring LDH release into the coculture supernatant after 72 h (<span class="html-italic">n</span> = 3). Data are presented as mean values ± SEM from 3–6 independent experiments. Statistical analysis by two-way ANOVA test followed by Tukey’s multiple comparison test; ns, not significant; * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
13 pages, 1246 KiB  
Systematic Review
Combined Radiotherapy and Hyperthermia: A Systematic Review of Immunological Synergies for Amplifying Radiation-Induced Abscopal Effects
by Loïc Van Dieren, Tom Quisenaerts, Mackenzie Licata, Arnaud Beddok, Alexandre G. Lellouch, Dirk Ysebaert, Vera Saldien, Marc Peeters and Ivana Gorbaslieva
Cancers 2024, 16(21), 3656; https://doi.org/10.3390/cancers16213656 - 30 Oct 2024
Viewed by 1187
Abstract
Introduction: The abscopal effect is a systemic immune response characterized by metastases regression at sites distant from the irradiated lesion. This systematic review aims to explore the immunological mechanisms of action underlying the abscopal effect and to investigate how hyperthermia (HT) can increase [...] Read more.
Introduction: The abscopal effect is a systemic immune response characterized by metastases regression at sites distant from the irradiated lesion. This systematic review aims to explore the immunological mechanisms of action underlying the abscopal effect and to investigate how hyperthermia (HT) can increase the chances of radiotherapy (RT) triggering systemic anti-tumor immune responses. Methods: This review is created in accordance with the PRISMA guidelines. Results and Conclusion: HT and RT have both complementary and synergistic immunological effects. Both methods trigger danger signal release, promoting cytokine and chemokine secretion, which increases T-cell infiltration and facilitates cell death. Both treatments upregulate extracellular tumor HSP70, which could amplify DAMP recognition by macrophages and DCs, leading to stronger tumor antigen presentation and CTL-mediated immune responses. Additionally, the combined increase in cell adhesion molecules (VCAM-1, ICAM-1, E-selectin, L-selectin) could enhance leukocyte adhesion to tumors, improving lymphocyte trafficking and boosting systemic anti-tumor effects. Lastly, HT causes vasodilation and improves blood flow, which might exacerbate those distant effects. We suggest the combination of local radiotherapy with fever-range whole-body hyperthermia to optimally enhance the chances of triggering the abscopal effect mediated by the immune system. Full article
(This article belongs to the Topic Anti-Tumor Immune Responses 2.0)
Show Figures

Figure 1

Figure 1
<p>Prisma flowchart of the included studies.</p>
Full article ">Figure 2
<p>DAMP release induced by RT and its subsequent recognition by an immune cell. RT (indicated by the lightning bolt) triggers the presentation of ATP, calreticulin (CRT) and high-mobility group box 1 (HMG1) from the tumor cell. The released DAMPs bind to purinergic receptors on the surface of the immune cell, initiating a signaling cascade that enhances the immune cell’s ability to recognize and respond to the tumor cell. This interaction is crucial for the activation and recruitment of immune cells to the tumor microenvironment, thereby boosting the anti-tumor immune response.</p>
Full article ">Figure 3
<p>Activation of the cGAS-STING pathway by radiation therapy. Radiation therapy (indicated by the lightning bolt) causes the release of cytosolic DNA within the cell. This DNA is detected by the cyclic GMP-AMP synthase (cGAS), which then produces cyclic GMP-AMP (cGAMP). cGAMP binds to the stimulator of interferon genes (STING) on the endoplasmic reticulum, leading to its activation. Activated STING recruits and activates TANK-binding kinase 1 (TBK1), which in turn phosphorylates interferon regulatory factor 3 (IRF-3). Phosphorylated IRF-3 translocates to the nucleus, where it promotes the production of type I interferons (IFNs). These interferons are then secreted from the cell, enhancing the immune response by promoting the recruitment and activation of immune cells. Additionally, TREX1, a DNA exonuclease that degrades cytosolic DNA regulates this pathway.</p>
Full article ">Figure 4
<p>Schematic representation of the immune response induced by radiation and hyperthermia therapy. This figure illustrates the synergistic effects of hyperthermia (indicated by the little flames) and radiation therapy (indicated by the black and yellow circles) in enhancing the immune response within the tumor microenvironment and beyond. Radiation therapy activates the cGAS/STING pathway, enhancing immune recognition, T-cell trafficking, T-cell recruitment, and chemokine secretion. Radiation also normalizes blood vasculature, induces the expression of pro-inflammatory cytokines, stimulates antigen presentation, and increases the sensitivity of tumor cells to immune responses. Hyperthermia further amplifies these effects by increasing immune recognition and T-cell trafficking, as well as promoting the secretion of chemokines. The combination of hyperthermia and radiation therapy leads to an enhanced presentation of tumor antigens, which activates natural killer (NK) cells, T-cells, and helper T-cells (Th-cells). This combined approach effectively strengthens the anti-tumor immune response, providing a more robust attack on the tumor cells.</p>
Full article ">
21 pages, 4616 KiB  
Article
Targeted Delivery to Dying Cells Through P-Selectin–PSGL-1 Axis: A Promising Strategy for Enhanced Drug Efficacy in Liver Injury Models
by Te-Sheng Lien, Der-Shan Sun and Hsin-Hou Chang
Cells 2024, 13(21), 1778; https://doi.org/10.3390/cells13211778 - 27 Oct 2024
Viewed by 936
Abstract
To minimize off-target adverse effects and improve drug efficacy, various tissue-specific drug delivery systems have been developed. However, even in diseased organs, both normal and stressed, dying cells coexist, and a targeted delivery system specifically for dying cells has yet to be explored [...] Read more.
To minimize off-target adverse effects and improve drug efficacy, various tissue-specific drug delivery systems have been developed. However, even in diseased organs, both normal and stressed, dying cells coexist, and a targeted delivery system specifically for dying cells has yet to be explored to mitigate off-target effects within the same organ. This study aimed to establish such a system. By examining the surfaces of dying cells in vitro, we identified P-selectin glycoprotein ligand-1 (PSGL-1) as a universal marker for dying cells, positioning it as a potential target for selective drug delivery. We demonstrated that liposomes conjugated with the PSGL-1 binding protein P-selectin had significantly greater binding efficiency to dying cells compared to control proteins such as E-selectin, L-selectin, galectin-1, and C-type lectin-like receptor 2. Using thioacetamide (TAA) to induce hepatitis and hepatocyte damage in mice, we assessed the effectiveness of our P-selectin-based delivery system. In vivo, P-selectin-conjugated liposomes effectively delivered fluorescent dye and the apoptosis inhibitor z-DEVD to TAA-damaged livers in wild-type mice, but not in PSGL-1 knockout mice. In TAA-treated wild-type mice, unconjugated liposomes required a 100-fold higher z-DEVD dose compared to P-selectin-conjugated liposomes to achieve a comparable, albeit less effective, therapeutic outcome in lowering plasma alanine transaminase levels and alleviating thrombocytopenia. This emphasizes that P-selectin conjugation enhances drug delivery efficiency by approximately 100-fold in mice. These results suggest that P-selectin-based liposomes could be a promising strategy for targeted drug delivery, enabling both diagnosis and treatment by specifically delivering cell-labeling agents and rescue agents to dying cells via the P-selectin–PSGL-1 axis at the individual cell level. Full article
(This article belongs to the Special Issue Nanofluidics, Nanopores, and Nanomaterials for Understanding Biology)
Show Figures

Figure 1

Figure 1
<p>Measurement of relative surface PSGL-1 expression after F4/80<sup>+</sup> splenocytes, Huh-7, B16F10, and J774A.1 cells were treated with cell death inducers. (<b>A</b>) Human hepatoma cell line Huh-7, (<b>B</b>) mouse melanoma cell line B16F10, (<b>C</b>) mouse macrophage cell line J774A.1, and (<b>D</b>) primary F4/80<sup>+</sup> splenocytes were treated with inducers of apoptosis (staurosporine, 10 μM), autophagy (rapamycin, 2 μM), ferroptosis (erastin, 20 μM), necroptosis (TNF-α, 0.2 μM), and pyroptosis (nigericin, 10 μM) for 4 h. Surface PSGL-1 expression levels were then measured by flow cytometry. Vehicle control groups were normalized to 1-fold. ** <span class="html-italic">p</span> &lt; 0.01 indicates markedly increased vs. respective vehicle-treated groups. The study included three experiments with two replicates per group [<span class="html-italic">N</span> = 3 (3 independent experiments), with <span class="html-italic">n</span> = 2 (2 technical replicates per experiment)].</p>
Full article ">Figure 2
<p>Apoptosis inducers induced surface PSGL-1 expression in Huh-7 cells. (<b>A</b>) Apoptosis inducers like staurosporine (STS; 10 μM), cisplatin (50 μM), and 5-FU (50 μM) increased surface PSGL-1 expression on Huh-7 cells after 4 h. (<b>B</b>) A dose-dependent increase in PSGL-1 expression on the surfaces of Huh-7 cells was observed with STS treatments ranging from 0 to 10 μM over 4 h. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle-treated groups (<b>A</b>,<b>B</b>). The vehicle groups (<b>A</b>,<b>B</b>) were set to 1-fold. Three experiments were performed with two replicates per group [<span class="html-italic">N</span> = 3 (3 independent experiments), with <span class="html-italic">n</span> = 2 (2 technical replicates per experiment)].</p>
Full article ">Figure 3
<p>Huh-7 cell binding levels with fluorescent-dye-loaded liposomes were analyzed. Huh-7 cells were treated with or without the apoptosis inducer staurosporine, and their surface PSGL-1 expression (<b>A</b>) and engagement with fluorescein-loaded liposomes conjugated with various lectins and proteins (<b>B</b>) were measured using flow cytometry. Staurosporine treatment induced a dose-dependent increase in P-selectin (P-sel) liposome–Huh-7 cell interaction (<b>A</b>). ** <span class="html-italic">p</span> &lt; 0.01 indicates a significant increase compared to the respective IgG-Fc–liposome groups (<b>A</b>). The IgG–liposome plus 2.5 μM STS-treated group (<b>A</b>) was set to 1-fold. (<b>B</b>) The conjugated lectins and proteins included IgG-Fc, P-selectin (P-sel), E-selectin (E-sel), L-selectin (L-sel), galectin-1 (Gal-1), and CLEC2. As these recombinant proteins contained an IgG-Fc portion, IgG-Fc was used as a control. (<b>B</b>) Both vehicle-treated and staurosporine-treated IgG-Fc groups were normalized to 1-fold. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 indicates a significant increase compared to the respective IgG-Fc groups. The study comprised three experiments, with two replicates per group [<span class="html-italic">N</span> = 3 (3 independent experiments), <span class="html-italic">n</span> = 2 (2 technical replicates per experiment)].</p>
Full article ">Figure 4
<p>Surface PSGL-1 expression and engagement with liposomes conjugated to various lectins and proteins in primary ASGR1<sup>+</sup> mouse hepatocytes. Primary ASGR1<sup>+</sup> hepatocytes were treated with or without inducers of apoptosis (staurosporine, 10 μM), autophagy (rapamycin, 2 μM), ferroptosis (erastin, 20 μM), necroptosis (TNF-α, 0.2 μM), and pyroptosis (nigericin, 10 μM) for 4 h in vitro, followed by exposure to fluorescein-loaded liposomes conjugated with different lectins and proteins. Surface PSGL-1 expression levels (<b>A</b>) and cell–liposome engagement levels (<b>B</b>) were analyzed by flow cytometry. The conjugated lectins and proteins included IgG-Fc, P-sel, E-sel, L-sel, Gal-1, and CLEC2. Since these recombinant proteins all contained an IgG-Fc portion, an isotype-matched IgG-Fc was used as a control. Vehicle-treated groups (<b>A</b>) and IgG-Fc-treated hepatocyte groups (<b>B</b>) were normalized to 1-fold. ** <span class="html-italic">p</span> &lt; 0.01 vs. vehicle-treated groups (<b>A</b>), ** <span class="html-italic">p</span> &lt; 0.01 indicated a significant increase vs. respective IgG-Fc groups (<b>B</b>). The study was conducted with three independent experiments, each including two replicates per group [<span class="html-italic">N</span> = 3 (3 independent experiments), with <span class="html-italic">n</span> = 2 (2 technical replicates per experiment)].</p>
Full article ">Figure 5
<p>RCD profiling and PSGL-1 levels in hepatocytes from TAA-treated mice. (<b>A</b>) The RCD profile in ASGR1<sup>+</sup> hepatocytes was assessed following TAA treatment using flow cytometry to evaluate changes in RCD levels. (<b>B</b>) Surface expression levels of PSGL-1 were measured in RCD-marker<sup>+</sup>ASGR1<sup>+</sup> double-positive hepatocytes by flow cytometry. For example, in the apoptosis group, PSGL-1 levels were determined by identifying PSGL-1<sup>+</sup> cells within the active caspase-3<sup>+</sup>ASGR1<b><sup>+</sup></b> double-positive population. The “overall” groups were analyzed for PSGL-1 expression in ASGR1<sup>+</sup> hepatocytes, independent of RCD markers. Vehicle control groups were normalized to a 1-fold change. ** <span class="html-italic">p</span> &lt; 0.01 vs. respective vehicle-treated groups. The study was conducted with three independent experiments, each including two replicates per group [<span class="html-italic">N</span> = 3 (3 independent experiments), with <span class="html-italic">n</span> = 2 (2 technical replicates per experiment)]. A total of 12 mice were used in the study.</p>
Full article ">Figure 6
<p>Dying-cell-specific targeting via the P-selectin–PSGL-1 axis demonstrated by fluorescent dye delivery to injured mouse liver. Following TAA-induced liver injury, wild-type (WT) (<b>A</b>) and PSGL-1 null (<span class="html-italic">Selplg</span><sup>−/−</sup>) (<b>B</b>) mice were injected with fluorescein-loaded liposomes without protein conjugation (vehicle; 1), or conjugated to IgG-Fc (2), P-sel (3), E-sel (4), L-sel (5), Gal-1 (6), CLEC2 (7), and anti-PSGL-1 antibody (8). Fluorescent liver images are shown (A, B), and relative fluorescence intensities were quantified using ImageJ (<b>C</b>), in which the fluorescence levels of mouse liver in the IgG-Fc groups were normalized to 1-fold (<b>C</b>). (<b>D</b>) Pseudo-color imaging revealed that the system selectively delivered fluorescein to injured liver tissue while avoiding non-target organs like the lungs and spleen, where nanoparticles are typically retained. * <span class="html-italic">p</span> &lt; 0.05 vs. IgG-Fc groups. Three experiments with two mice per group (<span class="html-italic">n</span> = 6). A total of 106 (53 WT, 53 <span class="html-italic">Selplg</span><sup>−/−</sup>) mice were used in the study.</p>
Full article ">Figure 7
<p>Rescue of damaged liver through dying-cell-specific targeting via the P-selectin–PSGL-1 axis with delivery of apoptosis inhibitor z-DEVD in mice. After TAA-induced liver injury, wild-type and <span class="html-italic">Selplg</span><sup>−/−</sup> mice were injected with caspase-3 inhibitor z-DEVD-loaded liposomes (5 × 10<sup>7</sup> liposomes, containing 2 μM z-DEVD/mouse) conjugated to various proteins, including IgG-Fc, P-sel, E-sel, L-sel, Gal-1, CLEC2, and anti-PSGL-1 antibody (anti-PSGL-1). Plasma ALT levels (<b>A</b>) and platelet (PLT) counts (<b>B</b>) were measured. ** <span class="html-italic">p</span> &lt; 0.01, * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle groups; ## <span class="html-italic">p</span> &lt; 0.01, # <span class="html-italic">p</span> &lt; 0.05 vs. IgG-Fc groups. Three experiments, with two mice per group (<span class="html-italic">n</span> = 6). A total of 108 (54 WT, 54 <span class="html-italic">Selplg</span><sup>−/−</sup>) mice were used in the study.</p>
Full article ">Figure 8
<p>P-selectin-conjugated liposomes exert a rescue effect that is associated with the suppression of caspase-3 activities in mice. (<b>A</b>) Elevated surface PSGL-1 expression and (<b>B</b>) activated caspase-3 levels in TAA-treated mouse (wild type and <span class="html-italic">Selplg</span><sup>−/−</sup>) hepatocytes in vivo were markedly rescued by treatments with P-selectin-conjugated liposomes (P-sel groups) loaded with caspase-3 inhibitor z-DEVD (5 × 10<sup>7</sup> liposomes, containing 2 μM z-DEVD/mouse). The measurements of untreated groups were normalized to 1-fold. ** <span class="html-italic">p</span> &lt; 0.01, indicated markedly exacerbated vs. respective untreated control groups; # <span class="html-italic">p</span> &lt; 0.05 indicated markedly ameliorated vs. respective IgG-Fc control groups. Three experiments with two mice per group (<span class="html-italic">n</span> = 6) were performed. A total of 48 (24 WT, 24 <span class="html-italic">Selplg</span><sup>−/−</sup>) mice were used in the study.</p>
Full article ">Figure 9
<p>Markedly enhanced rescue efficiency was observed when Red-DEVD was loaded into P-selectin-conjugated liposomes in mice. (<b>A</b>) Quantification of Red-DEVD dosages was performed by measuring the fluorescence intensity of the fluorescent Red-DEVD. (<b>B</b>,<b>C</b>) In vivo testing of unconjugated liposomes (unc) loaded with Red-DEVD, with injection doses ranging from 30 ng to 900 ng per mouse [e.g., unc (30) represents unconjugated liposomes with 30 ng Red-DEVD], was performed to evaluate their ability to reduce TAA-induced increases in circulating ALT levels (<b>B</b>) and thrombocytopenia ((<b>C</b>), reduced platelet counts). The rescue effects on ALT levels (<b>D</b>) and thrombocytopenia (<b>E</b>) in TAA-treated mice, using unconjugated liposomes with Red-DEVD doses of 0–1000 ng per mouse, were compared to those using P-selectin-conjugated liposomes with Red-DEVD doses of 5–10 ng per mouse [e.g., P-sel (5) for P-selectin-conjugated liposomes with 5 ng Red-DEVD]. The untreated groups were normalized to 1-fold. ** <span class="html-italic">p</span> &lt; 0.01 vs. respective TAA-treated unc (0) groups. Three experiments with two replicates per group (<span class="html-italic">n</span> = 6; (<b>A</b>–<b>C</b>)), and four experiments with three mice per group (<span class="html-italic">n</span> = 12; (<b>D</b>,<b>E</b>)) were conducted.</p>
Full article ">
19 pages, 2028 KiB  
Article
Elevated Platelet Aggregation in Patients with Ovarian Cancer: More than Just Increased Platelet Count
by Zitha Redempta Isingizwe, Brooke A. Meelheim and Doris Mangiaracina Benbrook
Cancers 2024, 16(21), 3583; https://doi.org/10.3390/cancers16213583 - 24 Oct 2024
Viewed by 929
Abstract
Background: Patients with ovarian cancer have high platelet counts, which correlate with disease burden, incidence, and lethality of blood clots (thrombosis). We hypothesized that elevated aggregation is associated with both increased platelet number and altered behavior of platelets in patients with ovarian cancer. [...] Read more.
Background: Patients with ovarian cancer have high platelet counts, which correlate with disease burden, incidence, and lethality of blood clots (thrombosis). We hypothesized that elevated aggregation is associated with both increased platelet number and altered behavior of platelets in patients with ovarian cancer. Methods: Healthy controls and patients with suspected or diagnosed ovarian cancer were evaluated for complete blood counts. To evaluate the effects of platelet count versus platelet behavior, equal platelet-rich plasma (PRP) volumes versus equal platelet numbers were used in platelet aggregation assays. Arachidonic acid, adenosine diphosphate, and collagen platelet agonists were used to induce aggregation. Volunteers were grouped into healthy controls (23), benign/borderline cases (7), and cancer cases (25 ovarian, 1 colorectal, and 2 endometrial). Results: The rate and amount of platelet aggregation were higher in patients compared to healthy controls regardless of whether the same platelet number or PRP volume was used. Compared to healthy controls, patients with untreated ovarian cancer exhibited high levels of platelet activation markers, P-selectin (27.06 vs. 31.06 ng/mL, p = 0.03), and beta-thromboglobulin (3.073 vs. 4.091 µg/mL, p = 0.02) in their plasma. The significance of the elevation and its correlations with platelet number or PRP volume varied depending on the agonist. Platelet (305.88 vs. 134.12, p < 0.0001) and white blood cell (8.459 vs. 5.395, p < 0.01) counts (×109/L) were elevated pre-chemotherapy and decreased post-chemotherapy, respectively. Conclusions: Elevated platelet aggregation is caused by both altered platelet number and behavior in patients with ovarian cancer. These results support the study of antiplatelet agents for thrombosis prevention in these patients. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of this study's inclusion and exclusion criteria. Healthy participants were included based on age, lack of recent use of antiplatelet drugs, and ability to physically give blood samples. Patients with benign ovarian tumors or borderline diseases were included in the benign group. Ovarian cancer participants were included based on the diagnosis of epithelial ovarian cancer. Primary comparisons are indicated by asterisks. All other comparisons were exploratory. The bottom boxes show the numbers (n) of patient specimens evaluated using equal numbers of platelets (Behavior) and equal volumes of platelet-rich plasma (Number).</p>
Full article ">Figure 2
<p>Comparison of CBCs in citrated whole blood between healthy controls and patients with untreated ovarian cancer. (<b>A</b>) white blood cells, (<b>B</b>) red blood cells, (<b>C</b>) platelets. Venous blood (8.5 mL) was collected using acid citrate dextrose (1.5 mL) anticoagulant. A VetScan HM5 Hematology Analyzer was used to count blood cells in each sample. (Healthy: Healthy controls, Untreated: patients with ovarian cancer before surgery or chemotherapy) Mann-Whitney or <span class="html-italic">t</span>-test; ns = not significant, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Platelet aggregation and aggregation slope comparing platelet behavior and platelet count. (<b>A</b>) Platelet aggregation comparing platelet behavior, (<b>B</b>) aggregation slope comparing platelet behavior, (<b>C</b>) platelet aggregation comparing platelet count, (<b>D</b>) aggregation slope comparing platelet count: Arachidonic acid, ADP, or collagen platelet aggregation agonists were used to induce platelet. Kruskal-Wallis test * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Concentrations of <span class="html-italic">P</span>-selectin, PF4, and beta-TG in PRP samples from healthy controls and patients with untreated ovarian cancer. <span class="html-italic">t</span>-test; ns = not significant, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
12 pages, 1880 KiB  
Article
Association of Endothelial Cell Activation with Acute Kidney Injury during Coronary Angiography and the Influence of Recombinant Human C1 Inhibitor—A Secondary Analysis of a Randomized, Placebo-Controlled, Double-Blind Trial
by Stephan Moser, Laura Araschmid, Anneza Panagiotou, Leo H. Bonati, Tobias Breidthardt, Gregor Fahrni, Christoph Kaiser, Raban Jeger, Marten Trendelenburg and Michael Osthoff
Biomedicines 2024, 12(9), 1956; https://doi.org/10.3390/biomedicines12091956 - 27 Aug 2024
Viewed by 905
Abstract
Background: Acute kidney injury (AKI) as a result of iodinated contrast media (CM) has been linked to CM-induced renal ischemia and toxic effects on endothelial cells (EC). The recombinant human C1 inhibitor (rhC1INH) has been shown to influence EC activation. Methods: Secondary analysis [...] Read more.
Background: Acute kidney injury (AKI) as a result of iodinated contrast media (CM) has been linked to CM-induced renal ischemia and toxic effects on endothelial cells (EC). The recombinant human C1 inhibitor (rhC1INH) has been shown to influence EC activation. Methods: Secondary analysis of 74/77 (96%) participants of a double-blind, randomized, and placebo-controlled study that assessed the effect of rhC1INH on AKI. E-selectin, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule (VCAM-1), and CC-chemokin-ligand-5 (CCL5) were determined in frozen blood samples over 48 h and analyzed according to the treatment group and renal outcomes. Results: The mean age was 76.7 years, and 37 patients each received rhC1INH and placebo, respectively. In the entire study population, minor differences in median EC activation markers/CCL5 concentrations during the first 48 h compared to baseline were observed (e.g., E-selectin 27.5 ng/mL at baseline vs. 29.7 ng/mL on day 1, CCL5: 17.7 ng/mL at baseline vs. 32.2 ng/mL on day 2). Absolute changes in ICAM-1/E-selectin concentrations correlated with a higher peak change in urinary NGAL concentrations. However, AKI was not associated with significant changes in EC markers/CCL5. Last, no significant differences in serum concentrations of EC activation markers/CCL5 were evident between the placebo and the rhC1INH group. Conclusions: CM administration during coronary angiography only mildly activated ECs within the first 48 h, which does not explain subsequent AKI. The administration of rhC1INH was not associated with a reduction of EC activation or CCL5. Full article
(This article belongs to the Section Molecular and Translational Medicine)
Show Figures

Figure 1

Figure 1
<p>Serum concentrations of CCL5, E-Selectin, ICAM-1, and VCAM-1 (ng/mL) in the overall study population (n = 74) over time. Asterisks denote statistical significance (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Comparison of relative changes in serum concentrations of endothelial activation markers/CCL5 in patients according to the treatment with percutaneous coronary intervention (PCI) vs. coronary angiography only. Asterisks denote statistical significance (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Comparison of relative changes in serum concentrations of endothelial activation markers/CCL5 in patients according to the treatment rhC1INH or placebo.</p>
Full article ">
27 pages, 3141 KiB  
Article
Preclinical Efficacy of VTX-0811: A Humanized First-in-Class PSGL-1 mAb Targeting TAMs to Suppress Tumor Growth
by Tatiana Novobrantseva, Denise Manfra, Jessica Ritter, Maja Razlog, Brian O’Nuallain, Mohammad Zafari, Dominika Nowakowska, Sara Basinski, Ryan T. Phennicie, Phuong A. Nguyen, Michael A. Brehm, Stephen Sazinsky and Igor Feldman
Cancers 2024, 16(16), 2778; https://doi.org/10.3390/cancers16162778 - 6 Aug 2024
Viewed by 2469
Abstract
Omnipresent suppressive myeloid populations in the tumor microenvironment limit the efficacy of T-cell-directed immunotherapies, become more inhibitory after administration of T-cell checkpoint inhibitors, and are overall associated with worse survival of cancer patients. In early clinical trials, positive outcomes have been demonstrated for [...] Read more.
Omnipresent suppressive myeloid populations in the tumor microenvironment limit the efficacy of T-cell-directed immunotherapies, become more inhibitory after administration of T-cell checkpoint inhibitors, and are overall associated with worse survival of cancer patients. In early clinical trials, positive outcomes have been demonstrated for therapies aimed at repolarizing suppressive myeloid populations in the tumor microenvironment. We have previously described the key role of P-selectin glycoprotein ligand-1 (PSGL-1) in maintaining an inhibitory state of tumor-associated macrophages (TAMs), most of which express high levels of PSGL-1. Here we describe a novel, first-in-class humanized high-affinity monoclonal antibody VTX-0811 that repolarizes human macrophages from an M2-suppressive phenotype towards an M1 inflammatory phenotype, similar to siRNA-mediated knockdown of PSGL-1. VTX-0811 binds to PSGL-1 of human and cynomolgus macaque origins without inhibiting PSGL-1 interaction with P- and L-Selectins or VISTA. In multi-cellular assays and in patient-derived human tumor cultures, VTX-0811 leads to the induction of pro-inflammatory mediators. RNAseq data from VTX-0811 treated ex vivo tumor cultures and M2c macrophages show similar pathways being modulated, indicating that the mechanism of action translates from isolated macrophages to tumors. A chimeric version of VTX-0811, consisting of the parental murine antibody in a human IgG4 backbone, inhibits tumor growth in a humanized mouse model of cancer. VTX-0811 is exceptionally well tolerated in NHP toxicology assessment and is heading into clinical evaluation after successful IND clearance. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Generation of the anti-PSGL-1 antibody, 19L04. (<b>A</b>) Structure of PSGL-1: Human PSGL-1 is a homodimer linked by a disulfide-linker (green). The PSGL-1 N-terminus is responsible for binding to P-selectin, L-selectin, and VISTA and is the site of several post-translational modifications critical for ligand binding: tyrosine sulfation at residues 46, 48, and 51 and the addition of sialyl-Lewis x (inset). Between the N-terminus and the transmembrane region is the set of mucin-like domains. (<b>B</b>) Binding of anti-PSGL-1 antibodies 20I15, 18F02, and 19L04 to plate immobilized human PSGL-1-His protein, and biotinylated 21-mer peptides comprising the N-terminal amino acids of PSGL-1, residues 42–62. Sulfated peptides contain sulfated tyrosines at positions 46, 48, and 51. (<b>C</b>) Competition of anti-PSGL-1 antibodies 20I15, 18F02, and 19L04 with P-selectin and L-selectin. Serially diluted anti-PSGL-1 antibody was incubated with human PSGL-1-Fc-coated plates, and then biotinylated P-selectin or L-selectin was added at their ~EC50 of binding to PSGL-1 and detected with streptavidin-HRP. Experiments were performed at pH 7.4. (<b>D</b>) Competition of anti-PSGL-1 antibodies 20I15, 18F02, and 19L04 with VISTA. Serially diluted anti-PSGL-1 antibody was incubated with human PSGL-1-Fc-coated plates, and then biotinylated VISTA was added at its ~EC50 of binding to PSGL-1 and detected with streptavidin-HRP. Experiments were performed at pH 6.0.</p>
Full article ">Figure 2
<p>19L04c induced macrophage repolarization and pro-inflammatory activation in multicellular assays. (<b>A</b>) M2c repolarization. Fold-change relative to IgG4 isotype for each donor of secreted pro-inflammatory proteins TNFa, IL-6, IL-1b, and GM-CSF measured by Luminex in the supernatants from LPS stimulated M2c macrophages treated with 19L04c (n = 6 for GM-CSF, TNFa, and IL-1b; n = 4 for IL-6). (<b>B</b>) SEB-activated PBMC assay. Fold change relative to IgG4 isotype for each donor of secreted pro-inflammatory proteins IL-1b, IL-2, TNFa, and IFNg measured by Luminex in the supernatants from SEB-stimulated PBMCs treated with 19L04c. (n = 6 donors) (<b>C</b>) MLR assay. Fold change relative to IgG4 isotype for each donor of secreted proteins from an MLR assay. M0 macrophages were treated with 19L04c throughout the 9-day differentiation and polarization of M0 macrophages. The macrophages were then washed to remove residual 19L04c prior to co-incubation with allogeneic T cells for 4 days. On Day 13, Luminex was used to measure secreted mediators from macrophages (<b>a</b>–<b>c</b>) and T cells (<b>d</b>) in the supernatants, and flow cytometry was used for T-cell proliferation and activation (<b>e</b>,<b>f</b>). n = 6 donor pairs (three monocyte donors; two T-cell donors). * denotes the levels of significance. <span class="html-italic">p</span>-value ≤0.01 (**), ≤0.001 (***), ≤0.0001 (****).</p>
Full article ">Figure 3
<p>19L04c induces a pro-inflammatory microenvironment and leads to the suppression of tumor growth in a humanized mouse model. Tumor volume (<b>A</b>) and tumor weights in grams (<b>B</b>) of melanoma tumors in NGS-SGM3-BLT mice treated with 19L04c, Pembrolizumab, 19L04c plus Pembrolizumab combination or an Isotype control. Data are presented as the mean tumor volume ± standard deviation of the mean for all dose groups. Isotype n = 6, 19L04c n = 6, Pembrolizumab n = 5, 19L04c + Pembrolizumab n = 5. Dosing was initiated on Day 0. “ns” indicates no significance. (<b>C</b>) Total leukocytes per milligram of tumor calculated as the number of CD45 leukocytes divided by the weight of the tumor from each mouse. (<b>D</b>) The absolute number of leukocytes per tumor is calculated as the percent of viable CD45-positive leukocytes measured by flow cytometry multiplied by the total dissociated cell values in cells/mL. (<b>E</b>) MFI of the M2 marker CD163 on the total population of CD45<sup>+</sup>CD11b<sup>+</sup>CD14<sup>+</sup>CD3<sup>−</sup> macrophages identified using UMAP dimensionality reduction. The percentage of (<b>F</b>) granulocytic MDSCs identified as CD33<sup>+</sup> CD11blow MHCII low SSC high, (<b>G</b>) M1-like macrophages identified as CD33<sup>+</sup> CD86<sup>+</sup> MCHII<sup>+</sup> CD163<sup>−</sup>, (<b>H</b>) effector T cells identified as CD8<sup>+</sup> CD3<sup>+</sup>, (<b>I</b>) regulatory T cells identified as CD8<sup>−</sup>CD25<sup>+</sup>CD3<sup>+</sup> by flow cytometry. (<b>J</b>) The ratio of effector to regulatory T cells based on the percentage of each subset.</p>
Full article ">Figure 4
<p>19L04c induces a pro-inflammatory signature in ex vivo human tumor cultures. Percent induction of a (<b>A</b>) myeloid-associated pro-inflammatory cytokine signature (TNFα, IL-1β, GM-CSF), (<b>B</b>) an effector T-cell signature (IFNγ), and (<b>C</b>) a chemokine signature (CCL3, CCL4, CCL5, CXCL9, CXCL10) in human tumors treated with 19L04c or Pembrolizumab compared to an isotype control (n = 11 tumors). Solid horizontal lines (and numbers above the data sets) indicate the mean values in each treatment group. The dotted lines mark 30% induction levels. (<b>D</b>) Total tumor inflammation signature (IFN-γ, TNFa, IL1b, GM-CSF, CCL3, CCL4, CCL5, CXCL9, CXCL10) in two individual tumors: M1191174A1 (uterine), a 19L04c non-responder with response to Pembrolizumab (<b>left</b>) and WD-75441 (omentum), a Pembrolizumab non-responder with response to 19L04c (<b>right</b>).</p>
Full article ">Figure 5
<p>19L04 treatment leads to inflammatory shifts in primary M2c macrophages. M2c macrophages obtained from PBMCs of four donors treated with 19L04c or isotype control were subjected to RNAseq profiling. Differential gene expression was completed between the two treatment groups paired by the donor. Gene Set Enrichment Analysis was used to analyze the gene expression differences between the treatment groups. (<b>A</b>) 19L04c treatment mostly upregulated genes enriched in general inflammatory responses and TNFα signaling. (<b>B</b>) 19L04 treatment led to the downregulation of metabolic pathways, such as fatty acid metabolism and oxidative phosphorylation, and Myc targets pathways, all known to be associated with suppressive macrophages.</p>
Full article ">Figure 6
<p>VTX-0811 does not elicit cytokine release from naïve unstimulated PBMCs. Secreted cytokine levels (pg/mL) in response to treatment of unstimulated human PBMC with soluble (<b>A</b>) or plate-bound (<b>B</b>) 19L04c, anti-CD28, or an isotype control measured by Luminex. For the plate-bound method, tissue culture treated, flat bottom, 96-well plates were coated with 100 µL/well of F(ab′)<sub>2</sub> goat anti-human IgG, IgM (H + L) (10 µg/mL) at 4 °C for 24 h, washed, and then treated with VTX-0811, IgG4 isotype control, or anti-CD28 for 24 h prior to adding the cells. 19L04h and isotype control were evaluated at 10, 100, and 1000 µg/mL; anti-CD28 positive control was evaluated at 1, 10, and 50 µg/mL. Data shown are mean values ± standard deviation from six donors.</p>
Full article ">Figure 7
<p>VTX-0811 has minimal direct impact on T-cell function. Purified human T cells, activated with CD3 and CD28, were co-incubated with VTX-0811 antibody. Changes in IFNγ in the supernatant of the cultures (<b>A</b>) as well as changes in the MFI of CD69 staining intensity on CD8+ T cells (<b>B</b>) were compared to control-treated cells. Data are shown as mean values ± standard deviation from six donors.</p>
Full article ">Figure 8
<p>VTX-0811 has a modest effect on phagocytic activity and does not have an effect on human neutrophil activation. (<b>A</b>) M2c macrophages were differentiated and polarized from human blood monocytes. After polarization, the M2c macrophages were pre-incubated for 30 min with 19L04c, anti-CD47, or their respective isotype controls and then cultured with pHrodo-stained SK-MEL-5 cells for 2 h. The fold change was calculated based on the percentage of pHrodo+ gated from CD163+CD86- macrophages compared to the isotype control for 19L04 or the isotype control for anti-CD47 for each donor. The data represent an average of values obtained across 3 donors with 2 technical replicates per donor. (<b>B</b>) Whole blood was incubated with Isotype control or 19L04c or left untreated. One set of samples remained unstimulated (No stimulation). A second set was pre-activation for 15 min with fMLP prior to the addition of the antibody for 1 h (Pre-stimulation), and the third set was incubated with the antibody for 1 h prior to activation for 15 min with fMLP (Post-stimulation). The MFI of the indicated markers was assessed by flow cytometry and is representative of 3 donors.</p>
Full article ">Figure 9
<p>PK and RO of VTX-0811-CHO in cynomolgus monkeys. (<b>A</b>) PK of VTX-0811 in NHP. In the single-dose non-GLP compliant PK study, 3 male and 3 female cynomolgus monkeys (n = 1/group) received a single IV dose of VTX-0811 at 3, 10, or 30 mg/kg administered as a 30 min infusion. The concentration of the drug in the blood is plotted. Cmax was observed at 0.5 h post dose initiation for all doses. Cmax increased in a dose-proportional manner. (<b>B</b>) Target engagement for PSGL-1 was determined using a receptor occupancy method. VTX-0811 presented essentially dose-dependent binding to PSGL-1 on the surface of both T-cell (CD3<sup>+</sup>) and leukocyte (CD11b<sup>+</sup>) in whole blood samples collected in the single-dose PK study. For both cell types, PSGL-1 RO was near the peak or peaked at 0.5 h post infusion. RO was high at 79.24–100% and remained high at 0.5–168 h post-dosing for all dose levels assessed in both cell populations. For 3, 10, and 30 mg/kg doses, receptor occupancy returned to pre-dose levels at 648 h. Receptor occupancy data correlated with serum concentration, both declining at roughly 168 h, though the decline in RO was slower than the decline in exposure.</p>
Full article ">
20 pages, 2341 KiB  
Article
Anti-Platelet Activity of Sea Buckthorn Seeds and Its Relationship with Thermal Processing
by Natalia Sławińska, Jerzy Żuchowski, Anna Stochmal and Beata Olas
Foods 2024, 13(15), 2400; https://doi.org/10.3390/foods13152400 - 29 Jul 2024
Viewed by 855
Abstract
Sea buckthorn (Hippophae rhamnoides L.) is a tree or shrub with small, orange berries. Sea buckthorn seeds have shown many properties beneficial to human health, including antioxidant, anti-hypertensive, anti-hyperlipidemic, and retinoprotective activities. Seeds, as a component of food, are often exposed to [...] Read more.
Sea buckthorn (Hippophae rhamnoides L.) is a tree or shrub with small, orange berries. Sea buckthorn seeds have shown many properties beneficial to human health, including antioxidant, anti-hypertensive, anti-hyperlipidemic, and retinoprotective activities. Seeds, as a component of food, are often exposed to high temperatures, which can increase or decrease their biological activity. In our previous study, we showed that both raw and roasted sea buckthorn seeds had significant antioxidant activity, which was measured in human plasma in vitro. In this paper, we evaluated the effect of extracts from raw and roasted sea buckthorn seeds on several parameters of hemostasis in vitro, including thrombus formation in full blood (measured by the Total Thrombus formation Analysis System—T-TAS), blood platelet activation (based on the exposition of P-selectin, the active form of GPIIb/IIIa on their surface and platelet-derived microparticles formation), aggregation (measured with impedance aggregometry), adhesion to fibrinogen and collagen, arachidonic acid metabolism in washed platelets stimulated by thrombin, and COX-1 activity. We also measured the levels of free 8-isoprostane in plasma and the total non-enzymatic antioxidant status of plasma. The extract from roasted seeds (50 µg/mL) significantly prolonged the time of occlusion measured by T-TAS—the AUC10 (area under the curve) value was decreased by approximately 18%. Both extracts decreased the exposition of the active form of GPIIb/IIIa on the surface of platelets activated with 10 μM ADP (by 38.4–62.2%) and 20 μM ADP (by 39.7–51.3%). Moreover, the extract from raw seeds decreased the exposition of P-selectin on the surface of platelets stimulated with 20 μM ADP (by 31.2–34.9%). The adhesion of thrombin-stimulated platelets to fibrinogen and collagen was inhibited only by the extract from roasted sea buckthorn seeds (by 20–30%). Moreover, the extract from raw seeds inhibited the level of TBARS (thiobarbituric acid-reactive substances, an indicator of enzymatic peroxidation of arachidonic acid) in washed platelets stimulated with thrombin; the activity of COX-1 was inhibited by both extracts, although the effect of the extract from raw seeds was stronger. These results indicate that sea buckthorn seeds have anti-platelet activity that is not decreased by thermal processing, but more research is needed to determine which exact chemical compounds and mechanisms are responsible for this phenomenon. Full article
(This article belongs to the Section Food Engineering and Technology)
Show Figures

Figure 1

Figure 1
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on thrombus formation in whole blood (<b>A</b>,<b>B</b>) (<span class="html-italic">n</span> = 6) and blood platelet aggregation in whole blood (<b>C</b>) (<span class="html-italic">n</span> = 6). The samples (<b>A</b>,<b>B</b>) were analyzed with the T-TAS PL chip at the shear stress rates of 1500/s. The results (<b>A</b>,<b>B</b>) are calculated as AUC<sub>10</sub> (area under the curve). In the graph (<b>A</b>), AUC<sub>10</sub> is expressed as a percentage of the control sample (blood without the tested extract). The data are expressed as means ± SD (<b>A</b>) or median and interquartile range (<b>C</b>). The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05). The number above the significant result is the % of inhibition of thrombus formation (<b>A</b>). (<b>B</b>) demonstrates a selected diagram of the pressure recorded inside the PL chip for 10 min (OT—occlusion time) (for the extract from roasted seeds).</p>
Full article ">Figure 2
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the exposition of the active form of GPIIb/IIIa on 10 µM ADP-stimulated blood platelets (<b>A</b>,<b>B</b>) and the exposition of P-selectin on 10 µM ADP-stimulated blood platelets (<b>C</b>,<b>D</b>) in whole blood. Blood platelets were gated based on their size and the exposition of CD61. In each sample, 5000 CD61-positive objects were acquired. To assess the exposition of GPIIb/IIIa, fluorescently conjugated monoclonal antibody PAC-1/FITC was used. Results are expressed as the percentage of platelets binding PAC-1/FITC. To assess the exposition of P-selectin, a fluorescently conjugated monoclonal antibody CD62P/PE was used. Results are expressed as the percentage of platelets binding CD62P/PE. Data represent the means ± SD. The blood samples were drawn from 5–6 healthy volunteers. The activity of the tested extract was compared to the control sample. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns—not significant.</p>
Full article ">Figure 3
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the exposition of the active form of GPIIb/IIIa on 20 µM ADP-stimulated blood platelets (<b>A</b>,<b>B</b>), and the exposition of P-selectin on 20 µM ADP-stimulated blood platelets (<b>C</b>,<b>D</b>) in whole blood. Blood platelets were gated based on their size and the exposition of CD61. For each sample, 5000 CD61-positive objects were acquired. To assess the exposition of GPIIb/IIIa, a fluorescently conjugated monoclonal antibody PAC-1/FITC was used. Results are expressed as the percentage of platelets binding PAC-1/FITC. To assess the exposition of P-selectin, fluorescently conjugated monoclonal antibody CD62P/PE was used. Results are expressed as the percentage of platelets binding CD62P/PE. Data represent the means ± SD. The blood samples were drawn from 5–6 healthy volunteers. The activity of the tested extract was compared to the control samples. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01); ns—not significant.</p>
Full article ">Figure 4
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on the adhesion of thrombin-activated platelets to fibrinogen (<b>A</b>), ADP-activated platelets to fibrinogen (<b>B</b>), thrombin-activated platelets to collagen (<b>C</b>), and the adhesion of unstimulated platelets to collagen (<b>D</b>) (<span class="html-italic">n</span> = 7). In the graphs, platelet adhesion is expressed as a percentage of the control sample (blood platelets without the tested extract). The data are expressed as medians and interquartile ranges. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns—not significant.</p>
Full article ">Figure 5
<p>Effect of the extracts from raw and roasted sea buckthorn seeds (at concentrations of 0.5–50 μg/mL) on arachidonic acid metabolism in blood platelets stimulated by thrombin (5 U/mL) (<span class="html-italic">n</span> = 8) (<b>A</b>), and the activity of COX-1 (<b>B</b>). In (<b>A</b>), the results are presented as TBARS/2 × 10<sup>8</sup> blood platelets (percent of the control sample). The data are expressed as medians and interquartile ranges. The results were considered significant at <span class="html-italic">p</span> &lt; 0.05 (*** <span class="html-italic">p</span> &lt; 0.001). The differences between negative and positive control were statistically significant (<span class="html-italic">p</span> &lt; 0.001). In (<b>B</b>), the results are expressed as % of COX-1 inhibition (in comparison to 100% initial activity sample—sample where inhibitor vehicle (50% DMSO) was added instead of the extracts). The extracts were incubated with COX-1 for 10 or 30 min at the concentrations of 1, 10, 50, and 100 μg/mL. The data are shown as the means and SD of two dilutions of the sample (each measured in duplicate).</p>
Full article ">
16 pages, 4748 KiB  
Article
Prunus yedoensis Bark Downregulates the Expression of Cell Adhesion Molecules in Human Endothelial Cell Lines and Relaxes Blood Vessels in Rat Aortic Rings
by Ye Eun Choi, Jung Mo Yang, Chae Won Jeong, Sujin Shin, Junkyu Park, Kyungjin Lee and Ju Hyun Cho
Pharmaceuticals 2024, 17(7), 926; https://doi.org/10.3390/ph17070926 - 10 Jul 2024
Viewed by 991
Abstract
The incidence of cardiovascular diseases, such as high blood pressure, is increasing worldwide, owing to population aging and irregular lifestyle habits. Previous studies have reported the vasorelaxant effects of Prunus yedoensis bark methanol extract. However, various solvent extracts of P. yedoensis bark and [...] Read more.
The incidence of cardiovascular diseases, such as high blood pressure, is increasing worldwide, owing to population aging and irregular lifestyle habits. Previous studies have reported the vasorelaxant effects of Prunus yedoensis bark methanol extract. However, various solvent extracts of P. yedoensis bark and their vascular relaxation mechanisms have not been sufficiently studied. We prepared extracts of P. yedoensis bark using various solvents (water, 30% ethanol, and 70% ethanol). P. yedoensis bark 30% ethanol extract (PYB-30E) decreased the expression of vascular cell adhesion molecule-1 (VCAM-1), intercellular adhesion molecule-1 (ICAM-1), and E-selectin in human umbilical vein endothelial cells (HUVECs) activated with 200 ng/mL TNF-α. Additionally, PYB-30E showed vasodilatory effects on isolated rat aortic rings. This was confirmed to be the result of the activation of the NO/cGMP pathway, regulation of non-selective calcium-activated K+ channels, and calcium channel blockade. Additionally, PYB-30E significantly reduced systolic and diastolic blood pressure in spontaneously hypertensive rats (SHR). Taken together, our results indicated that PYB-30E is a candidate functional material with preventive and therapeutic effects against hypertension. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of <span class="html-italic">Prunus yedoensis</span> bark extracts (PYB) and prunetin on the cell viability in human umbilical vein endothelial cells (HUVECs). The HUVECs were treated with PYB extracts of 100 and 200 μg/mL and prunetin from 1 to 10 μg/mL for 24 h. Cell viability was measured by MTT assay. Data were obtained from three independent experiments and are expressed as mean ± SEM. PYB-W, <span class="html-italic">P. yedoensis</span> water extract; PYE-30E, <span class="html-italic">P. yedoensis</span> 30% ethanol extract; PYE-70E, <span class="html-italic">P. yedoensis</span> 70% ethanol extract.</p>
Full article ">Figure 2
<p>Effects of PYB extracts by solvent and prunetin on protein expression level of vascular cell adhesion molecule-1 (VCAM-1). (<b>a</b>) intercellular adhesion molecule-1 (ICAM-1), (<b>b</b>) E-selectin, (<b>c</b>) in HUVECs. HUVECs were pretreated with PYB of 100 and 200 μg/mL and prunetin from 1 to 10 μg/mL and TNF-α for 6 h. Protein expression levels were measured via western blot and analyzed using Image J software (1.8.0 version). Data were obtained from three independent experiments and are expressed as mean ± SEM. * <span class="html-italic">p</span> &lt; 0.5, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. CON. ### <span class="html-italic">p</span> &lt; 0.001 vs. Nor. PYB-W, <span class="html-italic">P. yedoensis</span> water extract; PYE-30E, <span class="html-italic">P. yedoensis</span> 30% ethanol extract; PYE-70E, <span class="html-italic">P. yedoensis</span> 70% ethanol extract.</p>
Full article ">Figure 3
<p>The relaxant effect of PYB-30E on PE (1 μM)-induced aortic ring contraction in vascular endothelium-intact [(Endo+)] (<b>a</b>), or -denuded [(Endo-)] rings (<b>c</b>). Representative trace under the indicated conditions (<b>b</b>,<b>d</b>). Data points represent mean ± SEM (<span class="html-italic">n</span> = 4). *** <span class="html-italic">p</span> &lt; 0.001 vs. control. PE, Phenylephrine; Ach, Acetylcholine; PYB-30E, <span class="html-italic">P. yedoensis</span> 30% ethanol extract.</p>
Full article ">Figure 3 Cont.
<p>The relaxant effect of PYB-30E on PE (1 μM)-induced aortic ring contraction in vascular endothelium-intact [(Endo+)] (<b>a</b>), or -denuded [(Endo-)] rings (<b>c</b>). Representative trace under the indicated conditions (<b>b</b>,<b>d</b>). Data points represent mean ± SEM (<span class="html-italic">n</span> = 4). *** <span class="html-italic">p</span> &lt; 0.001 vs. control. PE, Phenylephrine; Ach, Acetylcholine; PYB-30E, <span class="html-italic">P. yedoensis</span> 30% ethanol extract.</p>
Full article ">Figure 4
<p>The relaxant effect of PYB-30E on PE (1 μM)-induced endothelial-intact aortic rings contraction in the presence or absence (Control) of L-NAME (100 μM), indomethacin (10 μM), ODQ (10 μM), and MB (10 μM). (<b>a</b>) Representative trace under the indicated conditions. (<b>b</b>) Data points represent mean ± SEM (n = 4–5). *** <span class="html-italic">p</span> &lt; 0.001 vs. control. PE, Phenylephrine; L-NAME, NO synthase inhibitor; ODQ, 1-H-[1,2,4]-oxadiazolo[4,3-α]quinoxalin-1-one; MB, methylene blue. *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 5
<p>Effect of K<sup>+</sup> channel blockers on PYB-30E-induced relaxation. The relaxant effect of PYB-30E on PE (1 μM)-induced contraction endothelial-intact aortic rings in the presence or absence (Control) of BaCl<sub>2</sub> (10 μM), 4-AP (10 mM), TEA (1 mM). (<b>a</b>) Representative trace under the indicated conditions. (<b>b</b>) Data points represent mean ± SEM (n = 5–7). *** <span class="html-italic">p</span> &lt; 0.001 vs. control. PE, Phenylephrine; BaCl<sub>2</sub>, Barium chloride; 4-AP, 4-aminopyridine; TEA, tetraethylammonium chloride.</p>
Full article ">Figure 6
<p>Effect of blocking Ca<sup>2+</sup> channel PYB-30E-induced relaxation. The inhibitory effect of PYB-30E on CaCl<sub>2</sub>-induced contraction in endothelium-intact aortic rings pre-contracted by PE. (<b>a</b>) Representative trace under the indicated conditions. (<b>b</b>) Data points represent mean ± SEM (<span class="html-italic">n</span> = 4–5). ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control. PE, Phenylephrine.</p>
Full article ">Figure 7
<p>The Inhibitory effect of PYB-30E on endothelial-intact aortic rings constricted by angiotensin II (10<sup>−9</sup>, 10<sup>−8</sup>, 10<sup>−7</sup>, 10<sup>−6</sup> M) (<b>a</b>). Representative trace under the indicated conditions (<b>b</b>). Data points represent mean ± SEM (<span class="html-italic">n</span> = 4–5). * <span class="html-italic">p</span> &lt; 0.5, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. control.</p>
Full article ">Figure 8
<p>Blood pressure-lowering effect of PYB-30E in a spontaneously hypertensive rat (SHR). (<b>a</b>) Systolic blood pressure (SBP), (<b>b</b>) diastolic blood pressure (DBP), (<b>c</b>) percent changes in SBP, (<b>d</b>) percent changes in DBP. Data points represent mean ± SEM (<span class="html-italic">n</span> = 5–6). * <span class="html-italic">p</span> &lt; 0.5, ** <span class="html-italic">p</span> &lt; 0.01 vs. control.</p>
Full article ">
12 pages, 1726 KiB  
Article
Antithrombotic Effect of Oil from the Pulp of Bocaiúva—Acrocomia aculeata (Jacq.) Lodd. ex Mart. (Arecaceae)
by Isabelly Teixeira Espinoça, Denise Caroline Luiz Soares Basilio, Anna Júlia Papa de Araujo, Rafael Seiji Nakano Ota, Kamylla Fernanda Souza de Souza, Nadla Soares Cassemiro, Davi Campos Lagatta, Edgar Julian Paredes-Gamero, Maria Lígia Rodrigues Macedo, Denise Brentan Silva, Janaina de Cássia Orlandi Sardi, Danilo Wilhelm-Filho, Ana Cristina Jacobowski and Eduardo Benedetti Parisotto
Nutrients 2024, 16(13), 2024; https://doi.org/10.3390/nu16132024 - 26 Jun 2024
Viewed by 1664
Abstract
The study aimed to evaluate the antithrombotic action of Acrocomia aculeata pulp oil (AAPO) in natura, in an in vitro experimental model. AAPO was obtained by solvent extraction, and its chemical characterization was performed by gas chromatography coupled to a mass spectrometer (GC-MS). [...] Read more.
The study aimed to evaluate the antithrombotic action of Acrocomia aculeata pulp oil (AAPO) in natura, in an in vitro experimental model. AAPO was obtained by solvent extraction, and its chemical characterization was performed by gas chromatography coupled to a mass spectrometer (GC-MS). In vitro toxicity was evaluated with the Trypan Blue exclusion test and in vivo by the Galleria mellonella model. ADP/epinephrine-induced platelet aggregation after treatment with AAPO (50, 100, 200, 400, and 800 μg/mL) was evaluated by turbidimetry, and coagulation was determined by prothrombin activity time (PT) and activated partial thromboplastin time (aPTT). Platelet activation was measured by expression of P-selectin on the platelet surface by flow cytometry and intraplatelet content of reactive oxygen species (ROS) by fluorimetry. The results showed that AAPO has as major components such as oleic acid, palmitic acid, lauric acid, caprylic acid, and squalene. AAPO showed no toxicity in vitro or in vivo. Platelet aggregation decreased against agonists using treatment with different concentrations of AAPO. Oil did not interfere in PT and aPTT. Moreover, it expressively decreased ROS-induced platelet activation and P-selectin expression. Therefore, AAPO showed antiplatelet action since it decreased platelet activation verified by the decrease in P-selectin expression as well as in ROS production. Full article
(This article belongs to the Special Issue Effects of Plant Extracts on Human Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Toxicity in human platelets and systemic in <span class="html-italic">G. mellonella</span>. (<b>A</b>) Percentage (%) of human platelet viability obtained in PRP treated with <span class="html-italic">Acrocomia aculeata</span> (AAPO) pulp oil at different concentrations (50, 100, 200, 400, and 800 μg/mL); negative control (NC): vehicle (DMSO, 0.6%) and positive control (PC): Triton X100 (1%). (***) indicates statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to NC. (<b>B</b>) In vivo systemic toxicity in <span class="html-italic">G. mellonella</span> model treated with different concentrations (50, 100, 400, and 800 μg/mL) of AAPO. The percentage of survival was evaluated for 72 h; negative control (NC): saline and positive control: DMSO (100%). Difference estimates in survival were compared using a <span class="html-italic">p</span> &lt; 0.05 log-rank test.</p>
Full article ">Figure 2
<p>Effect of AAPO on platelet aggregation induced by ADP and epinephrine. Percentage (%) of platelet aggregation at different concentrations of AAPO (50, 100, 200, 400, and 800 μg/mL), induced by ADP (30 μM) (<b>A</b>,<b>B</b>) and epinephrine (5 μg/mL) (<b>C</b>,<b>D</b>) for 5 and 10 min, respectively; negative control—NC (DMSO 0.6%) and positive control—PC (Ticlopidine 10 μM). (***) The statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to negative control (NC). (#) The statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to positive control (PC).</p>
Full article ">Figure 3
<p>Expression of platelet surface P-selectin after exposure of platelets to AAPO. (<b>A</b>) Representative dot plots generated by FlowJo software. The gate shows the platelet population. (<b>B</b>) Representative dot plot showing a platelet-positive population (CD42b-FITC). (<b>C</b>) Representative histograms showing the activation of platelets incubated with AAPO and stimulated by ADP (30 μM) for 5 min. Activated platelets were labeled with CD42b-FITC (5 μL) and CD62P (CD62P-PE, 5 μL) and kept in the dark for 15 min. (<b>D</b>) Mean fluorescence intensity (MFI) of CD62P-PE expressed on the membrane of activated platelets treated with different concentrations of AAPO (50, 100, 200, 400, and 800 μg/mL). DMSO (0.6%): negative control; ADP (30 μM): positive control. Three independent experiments were performed. (***) The statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to negative control. (##) The statistical difference with <span class="html-italic">p</span> &lt; 0.01 and (###) statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to ADP group.</p>
Full article ">Figure 4
<p>Intraplatelet content of ROS in AAPO-treated platelets. Intraplatelet ROS content after 30 min of incubation with DCFH-DA (10 µM), in platelets treated with 50, 100, 200, 400, and 800 μg/mL of <span class="html-italic">A. aculeata</span> pulp oil (AAPO) or controls; negative control—NC (DMSO 0.6%) and positive control—PC (hydrogen peroxide, H<sub>2</sub>O<sub>2</sub>). (***) The statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to negative control (NC). (#) The statistical difference with <span class="html-italic">p</span> &lt; 0.001 compared to positive control (PC).</p>
Full article ">
17 pages, 1173 KiB  
Systematic Review
Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis
by Miguel Á. Huerta, Miguel Á. Tejada and Francisco R. Nieto
Mar. Drugs 2024, 22(7), 290; https://doi.org/10.3390/md22070290 - 24 Jun 2024
Viewed by 3941
Abstract
Fucoidan is a polymer of L-fucose and L-fucose-4-sulphate naturally found in marine sources that inhibits p-selectin, preventing neutrophil recruitment to the site of injury. Fucoidan is employed in many studies as a tool to investigate the contribution of neutrophils to pain, showing analgesic [...] Read more.
Fucoidan is a polymer of L-fucose and L-fucose-4-sulphate naturally found in marine sources that inhibits p-selectin, preventing neutrophil recruitment to the site of injury. Fucoidan is employed in many studies as a tool to investigate the contribution of neutrophils to pain, showing analgesic effects. We performed a systematic review and meta-analysis to quantify the analgesic effects of pretreatment with fucoidan reported in the available preclinical studies. In addition, we summarized the articles which have studied the therapeutic effects of fucoidan in pathological pain at preclinical and clinical levels. The results of this systematic review reveal that pretreatment with fucoidan is a powerful tool which reduces neutrophil infiltration by 70–90% at early time points. This meta-analysis showed that preventative treatment with fucoidan produced a significant pain reduction. In addition, several preclinical studies have observed that fucoidan treatment reduces the pain that is associated with various pathologies. Finally, fucoidan has also been tested in several clinical trials, with some degree of analgesic efficacy, but they were mostly small pilot studies. Considering all the above information, it can be concluded that fucoidan is not only a preclinical tool for studying the role of neutrophils in pain but also a promising therapeutic strategy for pain treatment. Full article
(This article belongs to the Section Marine Pharmacology)
Show Figures

Figure 1

Figure 1
<p>Study selection PRISMA flow diagram.</p>
Full article ">Figure 2
<p>Forest plot of the pain reduction efficacy of pretreatment with fucoidan. CI, confidence interval; RE, random effects [<a href="#B62-marinedrugs-22-00290" class="html-bibr">62</a>,<a href="#B63-marinedrugs-22-00290" class="html-bibr">63</a>,<a href="#B64-marinedrugs-22-00290" class="html-bibr">64</a>,<a href="#B65-marinedrugs-22-00290" class="html-bibr">65</a>,<a href="#B66-marinedrugs-22-00290" class="html-bibr">66</a>,<a href="#B68-marinedrugs-22-00290" class="html-bibr">68</a>,<a href="#B69-marinedrugs-22-00290" class="html-bibr">69</a>,<a href="#B71-marinedrugs-22-00290" class="html-bibr">71</a>,<a href="#B72-marinedrugs-22-00290" class="html-bibr">72</a>,<a href="#B73-marinedrugs-22-00290" class="html-bibr">73</a>,<a href="#B74-marinedrugs-22-00290" class="html-bibr">74</a>,<a href="#B75-marinedrugs-22-00290" class="html-bibr">75</a>,<a href="#B76-marinedrugs-22-00290" class="html-bibr">76</a>,<a href="#B77-marinedrugs-22-00290" class="html-bibr">77</a>,<a href="#B78-marinedrugs-22-00290" class="html-bibr">78</a>,<a href="#B79-marinedrugs-22-00290" class="html-bibr">79</a>,<a href="#B80-marinedrugs-22-00290" class="html-bibr">80</a>,<a href="#B82-marinedrugs-22-00290" class="html-bibr">82</a>,<a href="#B85-marinedrugs-22-00290" class="html-bibr">85</a>,<a href="#B86-marinedrugs-22-00290" class="html-bibr">86</a>,<a href="#B87-marinedrugs-22-00290" class="html-bibr">87</a>,<a href="#B90-marinedrugs-22-00290" class="html-bibr">90</a>,<a href="#B91-marinedrugs-22-00290" class="html-bibr">91</a>,<a href="#B92-marinedrugs-22-00290" class="html-bibr">92</a>,<a href="#B94-marinedrugs-22-00290" class="html-bibr">94</a>].</p>
Full article ">
16 pages, 1038 KiB  
Article
Intestinal Microbiota and Derived Metabolites in Myocardial Fibrosis and Postoperative Atrial Fibrillation
by Antonio Nenna, Alice Laudisio, Chiara Taffon, Marta Fogolari, Cristiano Spadaccio, Chiara Ferrisi, Francesco Loreni, Omar Giacinto, Ciro Mastroianni, Raffaele Barbato, David Rose, Antonio Salsano, Francesco Santini, Silvia Angeletti, Anna Crescenzi, Raffaele Antonelli Incalzi, Massimo Chello and Mario Lusini
Int. J. Mol. Sci. 2024, 25(11), 6037; https://doi.org/10.3390/ijms25116037 - 30 May 2024
Viewed by 1079
Abstract
The high incidence of atrial fibrillation (AFib) following cardiac surgery (postoperative atrial fibrillation, POAF) relies on specific surgical features. However, in the setting of POAF, the role of the microbiome in the modulation of cardiac fibrosis is still not clear. This study aimed [...] Read more.
The high incidence of atrial fibrillation (AFib) following cardiac surgery (postoperative atrial fibrillation, POAF) relies on specific surgical features. However, in the setting of POAF, the role of the microbiome in the modulation of cardiac fibrosis is still not clear. This study aimed to analyze the effect of the microbiome and its main metabolic product (trimethylamine-N-oxide, TMAO) in the fibrosis of myocardial tissue, to investigate its role in POAF. Patients undergoing elective cardiac surgery with cardiopulmonary bypass, central atrio-caval cannulation and no history of AFib, were included. A fragment of the right atrium was analyzed for qualitative and mRNA-quantitative evaluation. A preoperative blood sample was analyzed with enzyme-linked immunosorbent assay (ELISA). A total of 100 patients have been included, with POAF occurring in 38%. Histologically, a higher degree of fibrosis, angiogenesis and inflammation has been observed in POAF. Quantitative evaluation showed increased mRNA expression of collagen-1, collagen-3, fibronectin, and transforming growth factor beta (TGFb) in the POAF group. ELISA analysis showed higher levels of TMAO, lipopolysaccharide and TGFb in POAF, with similar levels of sP-selectin and zonulin. TMAO ≥ 61.8 ng/mL (odds ratio, OR 2.88 [1.35–6.16], p = 0.006), preoperative hemoglobin < 13.1 g/dL (OR 2.37 [1.07–5.24], p = 0.033) and impaired right ventricular function (OR 2.38 [1.17–4.83], p = 0.017) were independent predictors of POAF. Also, TMAO was significantly associated with POAF by means of increased fibrosis. Gut microbiome product TMAO is crucial for myocardial fibrosis, which is a key factor for POAF. Patients in preoperative sinus rhythm who will develop POAF have increased genetic expression of pro-fibrotic genes and enhanced fibrosis in histological staining. Elevated TMAO level (≥61.8 ng/mL) is an independent risk factor for POAF. Full article
(This article belongs to the Special Issue New Cardiovascular Risk Factors)
Show Figures

Figure 1

Figure 1
<p>Relative gene expression (graphical visualization of data shown in <a href="#ijms-25-06037-t004" class="html-table">Table 4</a>). Statistically significant inter-group differences (POAF vs. No POAF) are marked as ** (if <span class="html-italic">p</span> &lt; 0.01) or * (if <span class="html-italic">p</span> &lt; 0.05) on top of the relevant column.</p>
Full article ">Figure 2
<p>Box plot for ELISA analysis (graphical visualization of data shown in <a href="#ijms-25-06037-t005" class="html-table">Table 5</a>). Statistically significant inter-group differences (POAF vs. No POAF) are marked as ** (if <span class="html-italic">p</span> &lt; 0.01) on top of the relevant box.</p>
Full article ">Figure 3
<p>Dot plot of the odds ratio relative to the final model in <a href="#ijms-25-06037-t009" class="html-table">Table 9</a>.</p>
Full article ">
14 pages, 1385 KiB  
Article
Effect of GLP-1RA Treatment on Adhesion Molecules and Monocyte Chemoattractant Protein-1 in Diabetic Patients with Atherosclerosis
by Marcin Hachuła, Marcin Basiak, Michał Kosowski and Bogusław Okopień
Life 2024, 14(6), 690; https://doi.org/10.3390/life14060690 - 28 May 2024
Cited by 1 | Viewed by 1172
Abstract
Cardiovascular disease (CVD) remains a prominent cause of global mortality, primarily driven by atherosclerosis. Diabetes mellitus, as a modifiable risk factor, significantly contributes to atherogenesis. Monocyte recruitment to the intima is a critical step in atherosclerotic plaque formation, involving chemokines and adhesion molecules [...] Read more.
Cardiovascular disease (CVD) remains a prominent cause of global mortality, primarily driven by atherosclerosis. Diabetes mellitus, as a modifiable risk factor, significantly contributes to atherogenesis. Monocyte recruitment to the intima is a critical step in atherosclerotic plaque formation, involving chemokines and adhesion molecules such as selectins, ICAM-1, and MCP-1. Glucagon-like peptide 1 receptor agonists (GLP-1RAs) are a promising group of drugs for reducing cardiovascular risk in diabetic patients, prompting investigation into their mechanisms of action. This interventional study enrolled 50 diabetes patients with atherosclerotic plaque, administering GLP-1RA for 180 days. Serum concentrations of MCP-1, ICAM-1, and L-selectin were measured before and after treatment. Anthropometric and biochemical parameters were also assessed. GLP-1RA treatment resulted in significant improvements in anthropometric parameters, glycemic control, blood pressure, and biochemical markers of liver steatosis. Biomarker laboratory analysis revealed higher baseline levels of MCP-1, ICAM-1, and L-selectin in diabetic patients with atherosclerotic plaque compared to healthy controls. Following treatment, MCP-1 and L-selectin levels decreased significantly (p < 0.001), while ICAM-1 levels increased (p < 0.001). GLP-1RA treatment in diabetic patients with atherosclerotic plaque leads to favorable changes in serum molecule levels associated with monocyte recruitment to the endothelium. The observed reduction in MCP-1 and L-selectin suggests a potential mechanism underlying GLP-1RA-mediated cardiovascular risk reduction. Further research is warranted to elucidate the precise mechanisms and clinical implications of these findings in diabetic patients with atherosclerosis. Full article
(This article belongs to the Section Physiology and Pathology)
Show Figures

Figure 1

Figure 1
<p>Concentration of ICAM-1 in study group before and after intervention.</p>
Full article ">Figure 2
<p>Concentration of L-selectin in study group before and after intervention.</p>
Full article ">Figure 3
<p>Concentration of MCP-1 in study group before and after intervention.</p>
Full article ">
12 pages, 547 KiB  
Article
Effects of Water-Based Exercise on Patients Older than 60 Years Undergoing Cardiac Rehabilitation after Coronary Intervention
by Jus Ksela, Jan Kafol, Danijela Vasic and Borut Jug
J. Cardiovasc. Dev. Dis. 2024, 11(5), 151; https://doi.org/10.3390/jcdd11050151 - 15 May 2024
Viewed by 1874
Abstract
Cardiac rehabilitation (CR) plays a crucial role in managing patients who have undergone coronary intervention (CI) following acute myocardial infarction. While water-based exercise is gaining recognition as an exercise modality in this patient population, its impact on the subgroup of older adults remains [...] Read more.
Cardiac rehabilitation (CR) plays a crucial role in managing patients who have undergone coronary intervention (CI) following acute myocardial infarction. While water-based exercise is gaining recognition as an exercise modality in this patient population, its impact on the subgroup of older adults remains unexplored. In this post hoc analysis, we investigated the effects of water-based exercise on adults older than 60 years undergoing CR after CI, comparing it to land-based exercise and a control group. In total, 45 patients aged over 60 participated in 14-day exercise programs, featuring two daily 30-min sessions. We assessed exercise capacity (VO2peak), vascular function (flow-mediated vasodilation (FMD)), heart rate variability (HRV), and blood markers (Interleukins 6, 8, and 10, P-Selectin, ICAM, and High-sensitivity CRP) before and after CR. VO2peak in the water-based group improved significantly after CR in comparison with the land-based group: 1.35 kg/mL/min (95% CI [0.20–2.50], p = 0.022). The significant difference between water-based and land-based groups was observed in several HRV parameters: Total power −1129.20 ms2 (95% CI [−1951.92–−306.49], p = 0.008); peak LF 0.04 Hz (95% CI [0.00–0.08], p = 0.036); SD1 −9.02 millisecond (95% CI [−16.86–−1.18], p = 0.025); and SD2 −19.71 ms (95% CI [−35.08–−4.34], p = 0.013). FMD and blood markers did not vary significantly based on the exercise group. These findings suggest that short-term water-based CR may have potential as an alternative to traditional land-based CR, improving VO2peak and cardiorespiratory fitness among adults over 60 years undergoing CR after CI. Full article
(This article belongs to the Special Issue Exercise and Cardiovascular Disease in Older Adults)
Show Figures

Figure 1

Figure 1
<p>VO<sub>2peak</sub> change before and after the intervention based on the exercise group.</p>
Full article ">
22 pages, 6199 KiB  
Article
Rapamycin Induces Phenotypic Alterations in Oral Cancer Cells That May Facilitate Antitumor T Cell Responses
by Amirmoezz Yonesi, Kei Tomihara, Danki Takatsuka, Hidetake Tachinami, Manabu Yamazaki, Amir Reza Younesi Jadidi, Mayu Takaichi, Shuichi Imaue, Kumiko Fujiwara, Shin-Ichi Yamada, Jun-Ichi Tanuma and Makoto Noguchi
Biomedicines 2024, 12(5), 1078; https://doi.org/10.3390/biomedicines12051078 - 13 May 2024
Cited by 3 | Viewed by 1616
Abstract
Objectives: In this study, we investigated the antitumor immunomodulatory effects of rapamycin in oral cancer. Study Design: We examined the proliferation, apoptosis, and migration of cancer cells and investigated the cell surface expression levels of immune accessory molecules and T cell immune responses [...] Read more.
Objectives: In this study, we investigated the antitumor immunomodulatory effects of rapamycin in oral cancer. Study Design: We examined the proliferation, apoptosis, and migration of cancer cells and investigated the cell surface expression levels of immune accessory molecules and T cell immune responses in vitro. We investigated the effect of in vivo administration of rapamycin on immune cell distribution and T cell immune responses in oral tumor-bearing mice. Results: Rapamycin treatment significantly inhibited OSCC cell proliferation and migration, increased apoptotic cell death, and upregulated cell surface expression of several immune accessory and adhesion molecules, including CD40, CD83, PD-L1, PD-L2, MHC class I, P-selectin, and VCAM-1. These cancer cells augmented T cell proliferation. In vivo rapamycin administration significantly attenuated mouse tumor growth with an increased proportion of immune cells, including CD4+ T cells, CD8+ T cells, and dendritic cells (DCs); decreased the proportion of immune suppressive cells, such as myeloid-derived suppressor cells and regulatory T cells; enhanced DC maturation and upregulated the surface expression of CD40, CD86, and ICAM-1. Conclusions: Our results suggest that the therapeutic effect of mTOR inhibition in oral cancer can cause direct antitumor and immunomodulatory effects. Full article
(This article belongs to the Special Issue Cellular and Pathogenesis Mechanisms in Oral Cancer)
Show Figures

Figure 1

Figure 1
<p>Immunohistochemical expression of mTOR in oral squamous cell carcinoma (OSCC) cases. Immunohistochemical mTOR expression profile in normal oral epithelia and OSCC tissues; normal oral epithelia (<b>A</b>,<b>B</b>), well-differentiated OSCC (<b>C</b>,<b>D</b>), and poorly differentiated OSCC (<b>E</b>,<b>F</b>). Hematoxylin and eosin (H&amp;E) staining (<b>A</b>,<b>C</b>,<b>E</b>) and immunostaining for mTOR (<b>B</b>,<b>D</b>,<b>E</b>). Scale bars: 100 μm (<b>A</b>–<b>F</b>).</p>
Full article ">Figure 2
<p>mTOR signal contributed to the proliferative and migratory activities of oral squamous cell carcinoma (OSCC) cells. (<b>A</b>) OSCC cells were cultured in the presence or absence of various concentrations of rapamycin for 72 h, and proliferative activity of cells was measured by the WST-1 cell proliferation assay. The results are presented as mean ± standard deviation from sextuplicate determinations. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (<b>B</b>) OSCC cells were cultured in the presence or absence of rapamycin for 48 h, and the migratory activity of cells treated with or without rapamycin was compared. The results are presented as mean ± standard deviation from quadruplicate determinations. An asterisk indicates a significant difference between two groups. (<span class="html-italic">p</span> &lt; 0.02). (<b>C</b>) OSCC cells were cultured in the presence or absence of rapamycin for 48 h and had been stained with propidium iodide (PI) and annexin V for quantification.</p>
Full article ">Figure 2 Cont.
<p>mTOR signal contributed to the proliferative and migratory activities of oral squamous cell carcinoma (OSCC) cells. (<b>A</b>) OSCC cells were cultured in the presence or absence of various concentrations of rapamycin for 72 h, and proliferative activity of cells was measured by the WST-1 cell proliferation assay. The results are presented as mean ± standard deviation from sextuplicate determinations. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (<b>B</b>) OSCC cells were cultured in the presence or absence of rapamycin for 48 h, and the migratory activity of cells treated with or without rapamycin was compared. The results are presented as mean ± standard deviation from quadruplicate determinations. An asterisk indicates a significant difference between two groups. (<span class="html-italic">p</span> &lt; 0.02). (<b>C</b>) OSCC cells were cultured in the presence or absence of rapamycin for 48 h and had been stained with propidium iodide (PI) and annexin V for quantification.</p>
Full article ">Figure 3
<p>Rapamycin induced phenotypic alteration in oral squamous cell carcinoma (OSCC) cells. OSCC cells were cultured in the presence or absence of rapamycin for 72 h, and cell surface expression of various molecules was analyzed using flow cytometry. Experiments were performed in triplicate and similar results were obtained. Representative histograms from one experiment are shown. Dashed lines, non-filled solid lines, and filled solid lines indicate results for non-stained cells, non-treated cells, and rapamycin-treated cells, respectively. Numbers in each panel indicate the mean fluorescence intensity of each molecule in non-treated cells (<b>upper</b>) and rapamycin-treated cells (<b>lower</b>).</p>
Full article ">Figure 4
<p>In vivo rapamycin administration altered the proportion of immune cells in oral cancer-bearing mice. Mice were challenged with NR-S1K cells. After 14 days, mice were treated with either 2 mg/kg of rapamycin or saline every 2 days. (<b>A</b>) Tumor sizes in mice were measured at the indicated time points (<span class="html-italic">n</span> = 5/group); <span class="html-italic">p</span> &lt; 0.05, control (non-treated) vs. rapamycin. After 12 days of treatment, peripheral blood, cervical lymph nodes, peripheral lymph nodes, and spleen were harvested and the percentages of different immune cell types in each organ in control mice or rapamycin-treated mice were determined using flow cytometry. (<b>B</b>) Representative scatter plots and (<b>C</b>) summary of these results are shown (<span class="html-italic">n</span> = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (PB: peripheral blood, PLN: peripheral lymph nodes, CLN: cervical lymph nodes, Spl: spleen).</p>
Full article ">Figure 4 Cont.
<p>In vivo rapamycin administration altered the proportion of immune cells in oral cancer-bearing mice. Mice were challenged with NR-S1K cells. After 14 days, mice were treated with either 2 mg/kg of rapamycin or saline every 2 days. (<b>A</b>) Tumor sizes in mice were measured at the indicated time points (<span class="html-italic">n</span> = 5/group); <span class="html-italic">p</span> &lt; 0.05, control (non-treated) vs. rapamycin. After 12 days of treatment, peripheral blood, cervical lymph nodes, peripheral lymph nodes, and spleen were harvested and the percentages of different immune cell types in each organ in control mice or rapamycin-treated mice were determined using flow cytometry. (<b>B</b>) Representative scatter plots and (<b>C</b>) summary of these results are shown (<span class="html-italic">n</span> = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (PB: peripheral blood, PLN: peripheral lymph nodes, CLN: cervical lymph nodes, Spl: spleen).</p>
Full article ">Figure 4 Cont.
<p>In vivo rapamycin administration altered the proportion of immune cells in oral cancer-bearing mice. Mice were challenged with NR-S1K cells. After 14 days, mice were treated with either 2 mg/kg of rapamycin or saline every 2 days. (<b>A</b>) Tumor sizes in mice were measured at the indicated time points (<span class="html-italic">n</span> = 5/group); <span class="html-italic">p</span> &lt; 0.05, control (non-treated) vs. rapamycin. After 12 days of treatment, peripheral blood, cervical lymph nodes, peripheral lymph nodes, and spleen were harvested and the percentages of different immune cell types in each organ in control mice or rapamycin-treated mice were determined using flow cytometry. (<b>B</b>) Representative scatter plots and (<b>C</b>) summary of these results are shown (<span class="html-italic">n</span> = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (PB: peripheral blood, PLN: peripheral lymph nodes, CLN: cervical lymph nodes, Spl: spleen).</p>
Full article ">Figure 4 Cont.
<p>In vivo rapamycin administration altered the proportion of immune cells in oral cancer-bearing mice. Mice were challenged with NR-S1K cells. After 14 days, mice were treated with either 2 mg/kg of rapamycin or saline every 2 days. (<b>A</b>) Tumor sizes in mice were measured at the indicated time points (<span class="html-italic">n</span> = 5/group); <span class="html-italic">p</span> &lt; 0.05, control (non-treated) vs. rapamycin. After 12 days of treatment, peripheral blood, cervical lymph nodes, peripheral lymph nodes, and spleen were harvested and the percentages of different immune cell types in each organ in control mice or rapamycin-treated mice were determined using flow cytometry. (<b>B</b>) Representative scatter plots and (<b>C</b>) summary of these results are shown (<span class="html-italic">n</span> = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (PB: peripheral blood, PLN: peripheral lymph nodes, CLN: cervical lymph nodes, Spl: spleen).</p>
Full article ">Figure 4 Cont.
<p>In vivo rapamycin administration altered the proportion of immune cells in oral cancer-bearing mice. Mice were challenged with NR-S1K cells. After 14 days, mice were treated with either 2 mg/kg of rapamycin or saline every 2 days. (<b>A</b>) Tumor sizes in mice were measured at the indicated time points (<span class="html-italic">n</span> = 5/group); <span class="html-italic">p</span> &lt; 0.05, control (non-treated) vs. rapamycin. After 12 days of treatment, peripheral blood, cervical lymph nodes, peripheral lymph nodes, and spleen were harvested and the percentages of different immune cell types in each organ in control mice or rapamycin-treated mice were determined using flow cytometry. (<b>B</b>) Representative scatter plots and (<b>C</b>) summary of these results are shown (<span class="html-italic">n</span> = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. An asterisk indicates a significant difference between two groups (<span class="html-italic">p</span> &lt; 0.05). (PB: peripheral blood, PLN: peripheral lymph nodes, CLN: cervical lymph nodes, Spl: spleen).</p>
Full article ">Figure 5
<p>In vivo rapamycin administration induced phenotypic alterations in dendritic cells (DCs) in tumors, spleens, peripheral lymph nodes, submandibular lymph nodes, and peripheral blood cells in oral cancer-bearing mice. The phenotypic profiles of dendritic cells in NR-S1K tumor-bearing mice after rapamycin administration were analyzed. Cells were harvested from each organ of rapamycin-treated or non-treated (control) OSCC-bearing mice, and the cell surface expression of various immune accessory molecules on CD11c<sup>+</sup> cells was determined using flow cytometry. Representative histograms are shown (n = 4/group). Dashed lines, unfilled solid lines, and filled solid lines indicate the results for non-stained cells, control mice, and rapamycin-treated mice, respectively. Numbers in each panel indicate the mean fluorescence intensity of each molecule in the control mice (<b>upper panel</b>) and rapamycin-treated mice (<b>lower panel</b>).</p>
Full article ">Figure 5 Cont.
<p>In vivo rapamycin administration induced phenotypic alterations in dendritic cells (DCs) in tumors, spleens, peripheral lymph nodes, submandibular lymph nodes, and peripheral blood cells in oral cancer-bearing mice. The phenotypic profiles of dendritic cells in NR-S1K tumor-bearing mice after rapamycin administration were analyzed. Cells were harvested from each organ of rapamycin-treated or non-treated (control) OSCC-bearing mice, and the cell surface expression of various immune accessory molecules on CD11c<sup>+</sup> cells was determined using flow cytometry. Representative histograms are shown (n = 4/group). Dashed lines, unfilled solid lines, and filled solid lines indicate the results for non-stained cells, control mice, and rapamycin-treated mice, respectively. Numbers in each panel indicate the mean fluorescence intensity of each molecule in the control mice (<b>upper panel</b>) and rapamycin-treated mice (<b>lower panel</b>).</p>
Full article ">Figure 5 Cont.
<p>In vivo rapamycin administration induced phenotypic alterations in dendritic cells (DCs) in tumors, spleens, peripheral lymph nodes, submandibular lymph nodes, and peripheral blood cells in oral cancer-bearing mice. The phenotypic profiles of dendritic cells in NR-S1K tumor-bearing mice after rapamycin administration were analyzed. Cells were harvested from each organ of rapamycin-treated or non-treated (control) OSCC-bearing mice, and the cell surface expression of various immune accessory molecules on CD11c<sup>+</sup> cells was determined using flow cytometry. Representative histograms are shown (n = 4/group). Dashed lines, unfilled solid lines, and filled solid lines indicate the results for non-stained cells, control mice, and rapamycin-treated mice, respectively. Numbers in each panel indicate the mean fluorescence intensity of each molecule in the control mice (<b>upper panel</b>) and rapamycin-treated mice (<b>lower panel</b>).</p>
Full article ">Figure 6
<p>In vivo rapamycin administration induced phenotypic alterations in tumor cells that facilitate T cell responses. (<b>A</b>) Phenotypic alterations of tumor cells in oral cancer-bearing mice after rapamycin administration were analyzed. Tumor cells were harvested from rapamycin-treated or non-treated (control) OSCC-bearing mice and cell surface expression of various immune accessory molecules were determined using flow cytometry. Representative histograms are shown (n = 4/group); <span class="html-italic">p</span> &lt; 0.05, control vs. rapamycin. Dashed lines, non-filled solid lines, and filled solid lines indicate results for non-stained cells, cells from control mice, and cells from rapamycin-treated mice, respectively. Numbers in each panel indicate the mean fluorescence intensity of each molecule in control mice (<b>upper</b>) and rapamycin-treated mice (<b>lower</b>). (<b>B</b>) T cell responses from OSCC cells from in vivo rapamycin-treated mice were measured in vitro. Spleen cells (1 × 10<sup>5</sup>) from naïve mice and tumor cells (1 × 10<sup>4</sup>) from rapamycin-treated or control mice were cocultured in a 96-well U-bottom culture plate in the presence of 0.5 μg/mL anti-CD3 for 72 h. Cells were restimulated with 50 ng/mL PMA, 500 ng/mL ionomycin, and 4 μM monensin 4 h before the end of culture. Interferon (IFN)-γ-producing T cells were determined using flow cytometry. Experiments were performed in triplicate and similar results were obtained. Representative scatter plots from one experiment are shown.</p>
Full article ">
Back to TopTop