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

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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (520)

Search Parameters:
Keywords = HMGB1

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 765 KiB  
Systematic Review
Translating Biomarker Research into Clinical Practice in Orthopaedic Trauma: A Systematic Review
by Alexander Baur and Augustine Mark Saiz
J. Clin. Med. 2025, 14(4), 1329; https://doi.org/10.3390/jcm14041329 - 17 Feb 2025
Viewed by 171
Abstract
Background/Objectives: Orthopaedic trauma management in polytrauma patients presents challenges, particularly in selecting between damage control orthopaedics (DCO) and early appropriate care (EAC). This systematic review evaluates these approaches and explores the role of biomarkers in optimising surgical timing. The primary objective of this [...] Read more.
Background/Objectives: Orthopaedic trauma management in polytrauma patients presents challenges, particularly in selecting between damage control orthopaedics (DCO) and early appropriate care (EAC). This systematic review evaluates these approaches and explores the role of biomarkers in optimising surgical timing. The primary objective of this review was to evaluate the potential clinical utility of biomarkers in guiding surgical timing and predicting perioperative complications. The secondary objective was to compare the effectiveness of DCO and EAC approaches, focusing on their impact on patient outcomes when controlled for Injury Severity Scores (ISSs). Methods: A systematic search of PubMed, MEDLINE, and Google Scholar identified studies focusing on fracture management (DCO versus EAC), timing protocols, and biomarkers in polytrauma patients. Twenty-seven studies met inclusion criteria. Results: Among the 27 studies, 12 evaluated biomarkers and 15 compared DCO and EAC. Point-of-care (POC) biomarkers, including lactate (p < 0.001; OR 1.305), monocyte L-selectin (p = 0.001; OR 1.5), and neutrophil L-selectin (p = 0.005; OR 1.56), demonstrated predictive value for sepsis, infection, and morbidity. CD16bright/CD62Ldim neutrophils were significant predictors of infection (p = 0.002). Advanced biomarkers, such as IL-6, IL-10, RNA IL-7R, HMGB1, and leptin offered prognostic insights but required longer processing times. No clear superiority was identified between DCO and EAC, with comparable outcomes when injury severity scores (ISS) were controlled. Conclusions: This systematic review highlights the challenge of translating biomarker research into clinical practice, identifying several point-of-care and advanced laboratory biomarkers with significant potential to predict complications like sepsis, infection, and MODS. Future efforts should focus on refining biomarker thresholds, advancing point-of-care technologies, and validating their role in improving surgical timing and trauma care outcomes. Full article
(This article belongs to the Special Issue Acute Trauma and Trauma Care in Orthopedics)
Show Figures

Figure 1

Figure 1
<p>PRISMA flow diagram.</p>
Full article ">
20 pages, 3572 KiB  
Article
Paeoniflorin Attenuates APAP-Induced Liver Injury via Intervening the Crosstalk Between Hepatocyte Pyroptosis and NETs
by Yu-Ru Zhu, Ya-Qin Yang, Dan-Dan Ruan, Yue-Mei Que, Hang Gao, Yan-Zi Yang and Hua-Jun Zhao
Int. J. Mol. Sci. 2025, 26(4), 1493; https://doi.org/10.3390/ijms26041493 - 11 Feb 2025
Viewed by 286
Abstract
(1) Liver injury caused by an overdose of acetaminophen (APAP) represents a major public health concern. Paeoniflorin (PF) has been reported to have anti-inflammatory and liver-protective effects, but the underlying mechanisms remain unclear. This study aimed to investigate the effect of PF on [...] Read more.
(1) Liver injury caused by an overdose of acetaminophen (APAP) represents a major public health concern. Paeoniflorin (PF) has been reported to have anti-inflammatory and liver-protective effects, but the underlying mechanisms remain unclear. This study aimed to investigate the effect of PF on the crosstalk between pyroptosis and NETs in AILI. (2) APAP-treated C57BL/6J mice were used to demonstrate the protective effect of PF on liver injury. HepG2 and dHL-60 cells were cultured to study the effects of PF on hepatocyte pyroptosis and neutrophil extracellular traps (NETs) in vitro. Moreover, cell co-culture experiments were performed, and mice were treated with a NETs-depleting agent and hepatocyte pyroptosis inhibitor to investigate the improvement of AILI induced by PF through regulating the crosstalk between hepatocyte pyroptosis and NETs. (3) PF significantly alleviated AILI. Additionally, PF inhibited the expression of pyroptosis-related proteins, high-mobility group box 1 (HMGB1), and NETs-associated proteins in vitro and in vivo. The co-culture experiments demonstrated that PF not only inhibited the NETs triggered by hepatocyte pyroptosis, but also suppressed the hepatocyte pyroptosis induced by NETs. In mice with depleted neutrophils, the level of hepatocyte pyroptosis notably decreased, indicating a diminished impact of PF. Similarly, NETs formation was reduced in mice receiving a pyroptosis inhibitor compared to the APAP group. Compared with DNase I alone, the reduction effect of PF combined with DNase I on serum ALT and AST levels decreased from 46.857% and 39.927% to 44.347% and 33.419%, respectively. Compared with DSF alone, PF combined with DSF reduced the ALT and AST levels from 46.857% and 39.927% to 45.347% and 36.419%, respectively. (4) PF demonstrated therapeutic effects on AILI. Its mechanism involves the regulation of the crosstalk between hepatocyte pyroptosis and NETs. This research substantiates the pharmacological promise of PF as a therapeutic intervention for acute AILI. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>PF attenuates AILI in mice. (<b>A</b>) Schematic diagram of drug administration to animals (n = 6); (<b>B</b>) body weights of mice during administration; (<b>C</b>) liver index measurements; (<b>D</b>–<b>G</b>) serum concentrations of ALT, AST, LDH, and MDA; (<b>H</b>) MPO levels in liver tissues of mice; (<b>I</b>) H&amp;E staining images. The magnification used for the H&amp;E staining images was ×100, with the scale bars representing 100 μm. All experimental data are presented as the mean ± SD. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p &lt;</span> 0.001 vs. the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. the APAP group.</p>
Full article ">Figure 2
<p>PF attenuates APAP-induced hepatocyte inflammation and pyroptosis. (<b>A</b>) The mRNA expression levels of <span class="html-italic">IL-1β</span>, <span class="html-italic">TNF-α</span>, <span class="html-italic">IL-6</span>, and <span class="html-italic">IL-18</span> in the liver of the mice were quantified using the quantitative real-time polymerase chain reaction (qRT-PCR). (<b>B</b>) The mRNA expression levels of <span class="html-italic">NLRP3</span>, <span class="html-italic">caspase-1</span>, <span class="html-italic">GSDMD</span>, and <span class="html-italic">HMGB1</span> in the livers of mice were measured via qRT-PCR. (<b>C</b>,<b>D</b>) The protein levels of NLRP3, caspase-1, GSDMD, and HMGB1 in the livers of mice were assessed using Western blotting (WB) analysis and are reported relative to β-actin levels. (<b>E</b>) The levels of HMGB1 in the serum of mice were quantified using enzyme-linked immunosorbent assays (ELISAs). (<b>F</b>) HepG2 cells were treated with various concentrations of PF (0, 10, 20, and 40 μM) for 24 h, and then the cell viability was assessed using the thiazolyl blue tetrazolium bromide (MTT) assay (n = 3). (<b>G</b>) HepG2 cells were pretreated with various concentrations of PF (0, 10, 20, and 40 μM) for 24 h and subsequently treated with APAP (10 mM) for 6 h, and then the cell viability was assessed using the MTT assay (n = 3). (<b>H</b>) The mRNA expression levels of <span class="html-italic">IL-1β</span>, <span class="html-italic">TNF-α</span>, <span class="html-italic">IL-6</span>, and <span class="html-italic">IL-18</span> in HepG2 cells were quantified using qRT-PCR. (<b>I</b>) The mRNA expression levels of <span class="html-italic">NLRP3</span>, <span class="html-italic">caspase-1</span>, <span class="html-italic">GSDMD</span>, and <span class="html-italic">HMGB1</span> in the HepG2 cells were quantified using qRT-PCR. (<b>J</b>,<b>K</b>) The protein levels of NLRP3, caspase-1, GSDMD, and HMGB1 in the HepG2 cells were assessed using WB analysis, and are reported relative to β-actin levels. (<b>L</b>) The concentration of HMGB1 in the culture medium of the HepG2 cells was quantified using ELISA. All experimental data are presented as the mean ± SD. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. the NC group; * <span class="html-italic">p</span>&lt; 0.05, ** <span class="html-italic">p</span>&lt; 0.01, and *** <span class="html-italic">p</span>&lt; 0.001 vs. the APAP group.</p>
Full article ">Figure 3
<p>PF inhibits the APAP-induced formation of NETs. (<b>A</b>) NETs in the serum were quantified using an ELISA for NE–DNA complexes. (<b>B</b>,<b>C</b>) The protein levels of MPO, PADI4, NE, and CitH3 in the mouse liver were assessed using WB analysis and are reported relative to β-actin levels. (<b>D</b>) To evaluate the distribution of MPO and CitH3, the positive areas for MPO and CitH3 in liver sections were assessed using IHC staining. The magnification in the images is ×200. (<b>E</b>) The surface characterization of the dHL-60 cells was performed using FESEM. The magnification used was ×2500, with the scale bars representing 20 μm. (<b>F</b>,<b>G</b>) A cellular immunofluorescence analysis was conducted to assess the protein expression of MPO and CitH3 in the dHL-60 cells (n = 3). The magnification used was ×400, with the scale bars representing 20 μm. (<b>H</b>,<b>I</b>) The protein levels of MPO, PADI4, NE, and CitH3 in the dHL-60 cells were assessed using WB analysis, and are reported relative to β-actin levels. All experimental data are presented as the mean ± SD. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. the APAP group or PMA group.</p>
Full article ">Figure 4
<p>PF intervenes the crosstalk between hepatocyte pyroptosis and NETs. (<b>A</b>) HepG2 cells in the co-culture were treated with PMA (100 nM) for 4 h, followed by treatment with various concentrations of PF (0, 10, 20, and 40 μM) for 24 h; subsequently, cell viability was assessed using the MTT assay (n = 3). (<b>B</b>) The mRNA expression level of GSDMD in the HepG2 cells in the co-culture was quantified using qRT-PCR. (<b>C</b>,<b>D</b>) The protein level of GSDMD in the HepG2 cells in the co-culture was assessed by WB analysis and reported relative to β-actin levels. (<b>E</b>,<b>F</b>) The protein levels of CitH3 in the dHL-60 cells in the co-culture were assessed using WB analysis and are reported relative to β-actin levels. (<b>G</b>) A cellular immunofluorescence analysis was performed to evaluate the expression of MPO and CitH3 in the dHL-60 cells in the co-culture (n = 3). The magnification used was ×400. All experimental data are presented as the mean ± SD. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. the NC group; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. the APAP group or PMA group.</p>
Full article ">Figure 5
<p>PF attenuates AILI in mice by intervening the crosstalk between hepatocyte pyroptosis and NETs. (<b>A</b>) The levels of NETs in the serum of the neutropenic mice were quantified using ELISAs. (<b>B</b>,<b>C</b>) The ALT and AST levels in the serum of the neutropenic mice. (<b>D</b>,<b>E</b>) The protein levels of GSDMD in the liver of the neutropenic mice were determined using WB analysis and are reported relative to β-actin levels. (<b>F</b>) The mRNA expression level of GSDMD in the liver of the neutropenic mice was quantified via qRT-PCR. (<b>G</b>,<b>H</b>) The protein levels of GSDMD, HMGB1, and CitH3 in the liver of the pyroptosis inhibitor-treated mice were assessed using WB analysis and are reported relative to β-actin levels. (<b>I</b>) The mRNA expression level of GSDMD in the liver of the pyroptosis inhibitor-treated mice was quantified via qRT-PCR. (<b>J</b>,<b>K</b>) The serum levels of ALT and AST in the pyroptosis inhibitor-treated mice. (<b>L</b>,<b>M</b>) The levels of NETs and HMGB1 in the serum of the pyroptosis inhibitor-treated mice were measured using ELISA. All experimental data are presented as the mean ± SD. ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. the NC group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 vs. the APAP or PMA group; <sup>Δ</sup> <span class="html-italic">p</span> &lt; 0.05, ns <span class="html-italic">p</span> &gt; 0.05 vs. the APAP + DNase I or APAP + DSF group.</p>
Full article ">
22 pages, 6338 KiB  
Article
Oxidative High Mobility Group Box-1 Accelerates Mitochondrial Transfer from Mesenchymal Stem Cells to Colorectal Cancer Cells Providing Cancer Cell Stemness
by Rika Sasaki, Yi Luo, Shingo Kishi, Ruiko Ogata, Yukiko Nishiguchi, Takamitsu Sasaki, Hitoshi Ohmori, Rina Fujiwara-Tani and Hiroki Kuniyasu
Int. J. Mol. Sci. 2025, 26(3), 1192; https://doi.org/10.3390/ijms26031192 - 30 Jan 2025
Viewed by 455
Abstract
Mitochondria are important organelles for cell metabolism and tissue survival. Their cell-to-cell transfer is important for the fate of recipient cells. Recently, bone marrow mesenchymal stem cells (BM-MSCs) have been reported to provide mitochondria to cancer cells and rescue mitochondrial dysfunction in cancer [...] Read more.
Mitochondria are important organelles for cell metabolism and tissue survival. Their cell-to-cell transfer is important for the fate of recipient cells. Recently, bone marrow mesenchymal stem cells (BM-MSCs) have been reported to provide mitochondria to cancer cells and rescue mitochondrial dysfunction in cancer cells. However, the details of the mechanism have not yet been fully elucidated. In this study, we investigated the humoral factors inducing mitochondrial transfer (MT) and the mechanisms. BM-MSCs produced MT in colorectal cancer (CRC) cells damaged by 5-fluorouracil (5-FU), but were suppressed by the anti-high mobility group box-1 (HMGB1) antibody. BM-MSCs treated with oxidized HMGB1 had increased expression of MT-associated genes, whereas reduced HMGB1 did not. Inhibition of nuclear factor–κB, a downstream factor of HMGB1 signaling, significantly decreased MT-associated gene expression. CRC cells showed increased stemness and decreased 5-FU sensitivity in correlation with MT levels. In a mouse subcutaneous tumor model of CRC, 5-FU sensitivity decreased and stemness increased by the MT from host mouse BM-MSCs. These results suggest that oxidized HMGB1 induces MTs from MSCs to CRC cells and promotes cancer cell stemness. Targeting of oxidized HMGB1 may attenuate stemness of CRCs. Full article
(This article belongs to the Special Issue Mitochondrial Function in Human Health and Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>TNT formation and MT between MSCs and CRC cells. (<b>A</b>) Effect of coculture of HT29 and hBM-MSCs with attached or unattached conditions. (<b>B</b>) Effect of coculture of CT26 and mBM-MSCs with attached or unattached conditions. (<b>C</b>) HMGB1 concentration in medium of 5-FU-trated CRC cells. (<b>D</b>,<b>E</b>) MSCs were cocultured with 5-FU (1.5 μg/mL)-treated CRC cells with attached condition. Green color, CT26 cells; red color, mitochondria of BM-MSCs. (<b>D</b>) Timelapse analysis of TNT formation of mMSC to 5FU treated-mouse CT26 CRC cell. The 10 s to 120 s panels expand the range indicated by the white square in the 0 s panel. White arrow, TNT; yellow arrow, MSC’s mitochondria in TNT; red arrow, MSC’s mitochondria in CT2t cell. (<b>E</b>) Timelapse analysis of MT between hMSCs and HT29 human CRC cell treated with 5FU. Right panel, temporal change in HT29 cell number containing hMSC mitochondria; i.e., mitochondrial transferred HT29 cell number. Scale bar, 10 μm. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. TNT: tunneling nanotube; MT: mitochondrial transfer; CRC: colorectal cancer; hMSCs: human bone marrow-derived mesenchymal stem cells; mBM-MSCs: mouse bone marrow-derived mesenchymal stem cells; 5-FU: 5-fluorouracil.</p>
Full article ">Figure 2
<p>Effect of HMGB1 on mitochondrial transfer from BM-MSCs to CRC cells. (<b>A</b>) Fluorescence image of mitochondrial transfer from hBM-MSCs to HT29 cells and mBM-MSCs to CT26 cells. Scale bar, 50 μm. (<b>Left</b>) Mito Deep Red-labeled hBM-MSCs were cocultured with PKH67-labeled HT29 cells. (<b>Right</b>) hBM-MSC mitochondria were transferred from hBM-MSCs to PKH67-labeled HT29 cells. (<b>B</b>,<b>C</b>) The percentage of Mito Deep Red-positive CRC cells relative to all CRC cells. (<b>D</b>,<b>E</b>) TNT number in 1000 CRC cells. CRC cells were pretreated with 5-FU (1.5 μg/mL) or co-treated with 5-FU (1.5 μg/mL) and anti-HMGB1 antibody (αHMGB1, 10 μg/mL) for 48 h. CRC cells were cocultured with Mito Deep Red-labeled BM-MSCs for 6, 12, and 24 h. (<b>F</b>,<b>G</b>) Effects of coculture with BM-MSCs with 5-FU (1.5 μg/mL) and/or αHMGB1 (10 μg/mL) for 48 h on mitochondrial hydroxyradical (mtSOX) (<b>G</b>) and mitochondrial membrane potential (TMRE). Scale bar, 50 μm. Right panels, semi-quantification of fluorescent intensities of mtSOX and TMRE. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. hBM-MSCs: human bone marrow-derived mesenchymal stem cells; mBM-MSCs: mouse bone marrow-derived mesenchymal stem cells; HMGB1: high mobility group B-1; 5-FU: 5-fluorouracil; MMP: mitochondrial membrane potential; TNT: tunneling nanotube.</p>
Full article ">Figure 3
<p>Effect of culture supernatant on mitochondrial-<span class="html-italic">transfer</span> associated factors in BM-MSCs. (<b>A</b>) Expression of genes associated with mitochondria transfer (<span class="html-italic">Miro1</span> and <span class="html-italic">Cx43</span>) in BM-MSCs. BM-MSCs were exposed to culture medium of CRC cells with or without αHMGB1 (10 μg/mL) for 48 h. CRC cells were pretreated with 5-FU (1.5 μg/mL). <span class="html-italic">ACTB</span> was used as a loading control. Right panels, semi-quantification of the signal densities in RT–PCR standardized with <span class="html-italic">ACTB</span> signal intensity. (<b>B</b>) Protein levels of Miro1 and Cx43 in BM-MSCs treated with same protocols. ACTB was used as a loading control. Right panels, semi-quantification of the signal densities in western blot standardized with ACTB signal intensity. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. hBM-MSCs: human bone marrow-derived mesenchymal stem cells; mBM-MSCs: mouse bone marrow-derived mesenchymal stem cells; 5-FU: 5-fluorouracil; CM: culture medium; HMGB1: high mobility group B-1; αHMGB1: anti-HMGB1 antibody; Miro1: mitochondrial Rho GTPase 1; Cx43: connexin43; ACTB: β-actin.</p>
Full article ">Figure 4
<p>Effect of oxidized HMGB1 on mitochondrial transfer associated factors of BM-MSCs. (<b>A</b>) Western blotting of H<sub>2</sub>O<sub>2</sub>-treated rHMGB1 (oxHMGB1) and 2-merucaptoethanol-treated rHMGB1 (redHMGB1). (<b>B</b>) Effect of redox modification of HMGB1 (100 ng/mL) on MT and TNT formation in coculture of MSCs and CRC cells. (<b>C</b>) Expression of mitochondria transfer-associated genes (<span class="html-italic">Miro1</span> and <span class="html-italic">Cx43</span>) in BM-MSCs treated with or without oxHMGB1 (100 ng/mL) for 48 h. ACTB was used as a loading control. Right panels, semi-quantification of the signal intensities in RT–PCR standardized with <span class="html-italic">ACTB</span> signal intensity. (<b>D</b>) Protein levels of Miro1 and Cx43. ACTB was used as a loading control. Right panels, semi-quantification of the signal intensities in western blot standardized with ACTB signal intensity. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. MSC: mesenchymal stem cell; MT: mitochondrial transfer; TNT: tunneling nanotube; hBM-MSCs: human bone marrow-derived mesenchymal stem cells; mBM-MSCs: mouse bone marrow-derived mesenchymal stem cells; HMGB1: high mobility group B-1; rHMGB1: recombinant HMGB1; oxHMGB1: oxidized HMGB1; redHMGB1: reduced HMGB1; Miro1: mitochondrial Rho GTPase 1; Cx43: connexin43; ACTB: β-actin.</p>
Full article ">Figure 5
<p>Effects of NF–κB inhibitor on mitochondrial function in CRC cells and mitochondrial transfer associated factors in BM-MSCs. (<b>A</b>) RT–PCR of RAGE and TLR4 as receptors for HMGB1 in BM-MSCs. (<b>B</b>,<b>C</b>) MtROS (<b>B</b>) and MMP (<b>C</b>) of CRC cells were examined. CRC cells were pretreated with or without 5-FU (1.5 μg/mL) for 48 h and cocultured with hBM-MSCs and then treated with or without a NF–κB inhibitor, JSH-23 (20 μM) for 24 h. (<b>D</b>,<b>E</b>) Expression of mitochondria transfer-associated genes (<span class="html-italic">Miro1</span> and <span class="html-italic">Cx43</span>) in BM-MSCs. hBM-MSCs (<b>D</b>) and mBM-MSCs (<b>E</b>). BM-MSCs were exposed to culture medium of 5-FU (1.5 μg/mL)-pretreated CRC cells and treated with or without JSH-23 (20 μM) for 48 h. GAPDH was used as a loading control. Right panels, Semi-quantification of the signal densities in RT–PCR standardized with <span class="html-italic">GAPDH</span> signal intensity. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. hBM-MSCs: human bone marrow-derived mesenchymal stem cells; mBM-MSCs: mouse bone marrow-derived mesenchymal stem cells; RAGE: receptor for advanced glycation end products; TLR4: toll-like receptor 4; mtROS: mitochondrial reactive oxidative species; mtSOX: mitochondrial hydroxyradical; MMP: mitochondrial membrane potential; TMRE: tetramethyl rhodamine; 5-FU: 5-fluorouracil; GAPDH: glyceraldehyde-3-phosphate dehydrogenase.</p>
Full article ">Figure 6
<p>Effect of artificial mitochondria transfer (mitoception) on metabolism of CRC cells. (<b>A</b>) FACS analysis of HT29 cells mitocepted with increasing amounts of hBM-MSCs-derived mitochondria. hBM-MSCs mitochondria were labeled with MitoBright LT Green. Right panel, relationship between loaded mitochondrial amounts and transferred mitochondrial fluorescent intensities. (<b>B</b>) Effect of hBM-MSCs coculture or mitoception on 5-FU sensitivity of HT29 cells. (<b>C</b>) Sensitivities to 5FU and CDDP of HT29 cells cocultured with hMSCs. Cell viability was determined by counting the number of cells after treatment with different concentrations of 5-FU or CDDP for 48 h (<b>B</b>,<b>C</b>). Insert, MT-positive cell (%). (<b>D</b>) Effect of mitoception on mtROS and MMP. Right panel, semi-quantification of fluorescent intensities. (<b>E</b>) Effect of mitoception on OXPHOS. Error bar, standard deviation from three independent trials. Statistical significance was calculated by an ordinary ANOVA test. FACS: fluorescence-activated cell sorting; MFI: mean fluorescence intensity; hBM-MSCs: human bone marrow-derived mesenchymal stem cells; 5-FU: 5-fluorouracil; ROS: reactive oxidative species; mtSOX: mitochondrial hydroxyradical; MMP: mitochondrial membrane potential; TMRE: tetramethyl rhodamine; OXPHOS: oxidative stress; OCR: oxygen consumption rate; Max: maximum; CDDP: cisplatin; MT: mitotransfer; hMSC: human bone marrow mesenchymal stem cells.</p>
Full article ">Figure 7
<p>Effect of artificial mitochondria transfer (mitoception) on stemness of CRC cells. (<b>A</b>) Effect of mitoception on sphere formation in HT29 cells. Sphere formation was examined in 10,000 cells for 7 days. Pictures were images of phase-contrast microscopy. Scale bar, 300 μm. Right panel, number of spheres was counted with microscopy at 7 days. (<b>B</b>) Expression of stemness-associated genes (<span class="html-italic">Sox2</span>, <span class="html-italic">CD44</span>, <span class="html-italic">LGR5</span>, and <span class="html-italic">KLF4</span>). <span class="html-italic">GAPDH</span> expression was used as a loading control. Right panel, semi-quantification of the signal densities in RT–PCR standardized with <span class="html-italic">GAPDH</span> signal intensity. (<b>C</b>) Tumorigenicity of HT29 cells with mitoception. Cells were inoculated subcutaneously in mice. Error bar, standard deviation from three independent trials. Statistical differences were calculated by an ordinary ANOVA test with Bonferroni correlation from five mice. hBM-MSCs: human bone marrow-derived mesenchymal stem cells; 5-FU: 5-fluorouracil; GAPDH: glyceraldehyde-3-phosphate dehydrogenase, SOX2: sex determining region Y box 2; LGR5: leucine-rich repeat-containing G-protein coupled receptor 5; KLF4: Krüppel-like factor 4.</p>
Full article ">Figure 8
<p>Effect of inhibition of MT in mouse subcutaneous tumor model. (<b>A</b>) Experimental protocol. 5-FU (30 mg/kg BW), and αHMGB1 (5 μg/mouse) [<a href="#B22-ijms-26-01192" class="html-bibr">22</a>] or NF–κB-I (JSH-23, 20 mg/kg BW) [<a href="#B23-ijms-26-01192" class="html-bibr">23</a>] were administered intraperitoneally twice a week. (<b>B</b>,<b>C</b>) Effect of MT inhibition on tumor growth; time course (<b>B</b>) and tumor volume at 4 weeks after inoculation (<b>C</b>) Right panel, loupe images of the maximum cut surface of representative tumors stained with hematoxylin and eosin. Scale bar, 1 cm. (<b>D</b>) Immunohistochemistry of CD73 and SOX2 in tumors. Scale bar, 50 μm. (<b>E</b>,<b>F</b>) Contents of mBM-MSC-related proteins, CD73 (<b>E</b>) and SOX (<b>F</b>) in tumor tissues. (<b>G</b>) Contents of ROS levels (4HNE). s. Error bar, standard deviation from five mice. Statistical differences were calculated by an ordinary ANOVA test with Bonferroni correlation. Miro1: mitochondrial Rho GTPase 1; SOX2: sex determining region Y box 2; MT: mitochondrial transfer; 4HNE: 4-hydroxynonenal; αHMGB1: anti-high mobility group box-1 antibody; 5-FU: 5-fluorouracil; NF–κB-I: nuclear factor–κB inhibitor, JSH-23.</p>
Full article ">Figure 9
<p>Effect of in vivo MT on CT26 cells in mice with <span class="html-italic">Miro1</span>-knockdown. (<b>A</b>) Experimental protocol. BALB/c mice were transplanted with <span class="html-italic">Miro1</span>-KD and mitochondria labeled BM cells (KD-mice) or mitochondria labeled BM cells (C-mice). CT26 cells were inoculated subcutaneously into KD- or C-mice. From the tumors, CT26 cells were isolated at 4 weeks after inoculation. (<b>B</b>) Effect of <span class="html-italic">Miro1</span>-KD on tumor growth. Left panel, loupe images of the maximum cut surface of representative tumors in each group (hematoxylin and eosin staining). Scale bar, 1 cm. (<b>C</b>) Fluorescent intensity in transplanted BM cells, CT26 cells isolated from KD- or c-mice in comparison with those in parental CT26 cells. Left panels, representative fluorescence images of CT26 tumors. Scale bar, 50 μm. (<b>D</b>–<b>H</b>) Effect of <span class="html-italic">Miro1</span>-KD on stem cell phenotypes in CT26 cells isolated from KD- or c-mice. Expression of stemness-associated genes, <span class="html-italic">NS</span> and <span class="html-italic">Lgr5</span> (<b>D</b>), apoptosis; Right panel, PARP cleavage by western blot analysis, (<b>E</b>), sphere formation (<b>F</b>), 5-FU sensitivity (<b>G</b>), and tumorigenicity (<b>H</b>). Right panel, loupe images of the maximum cut surface of representative tumors in 1 × 10<sup>4</sup>-inoculated group stained with hematoxylin and eosin. Scale bar, 1 cm. Error bar, standard deviation from five mice. Statistical differences were calculated by an ordinary ANOVA test with Bonferroni correlation. BM: bone marrow; C-mice: mice whose BM replaced with mitochondria labeled BM cells; KD-mice: mice whose BM replaced with <span class="html-italic">Miro1</span>-KD and mitochondria labeled BM cells; MSC: mesenchymal stem cells; Miro1: mitochondrial Rho GTPase 1; NS: nucleostemin; Lgr5: leucine-rich repeat-containing G-protein coupled receptor 5; KD: knockdown by siRNA; parent: parental CT26 cells without inoculation.</p>
Full article ">
29 pages, 6516 KiB  
Article
Investigating the Inhibitory Effects of Paliperidone on RAGEs: Docking, DFT, MD Simulations, MMPBSA, MTT, Apoptosis, and Immunoblotting Studies
by Akash Pratap Singh, Shaban Ahmad, Ahona Roy, Khalid Raza and Hemant K. Gautam
Int. J. Mol. Sci. 2025, 26(3), 1060; https://doi.org/10.3390/ijms26031060 - 26 Jan 2025
Viewed by 545
Abstract
Chronic diseases such as diabetes and cancer are the leading causes of mortality worldwide. Receptors for Advanced Glycation End products (RAGEs) are ubiquitous factors that catalyse Advanced Glycation End products (AGEs), proteins, and lipids that become glycated from sugar ingestion. RAGEs are cell [...] Read more.
Chronic diseases such as diabetes and cancer are the leading causes of mortality worldwide. Receptors for Advanced Glycation End products (RAGEs) are ubiquitous factors that catalyse Advanced Glycation End products (AGEs), proteins, and lipids that become glycated from sugar ingestion. RAGEs are cell surface receptor proteins and play a broad role in mediating the effects of AGEs on cells, contributing to modifying biological macromolecules like proteins and lipids, which can cause Reactive Oxygen Species (ROS) generation, inflammation, and cancer. We targeted RAGE inhibition analysis and screening of United States Food and Drug Administration (FDA) libraries through molecular docking studies that identified the four most suitable FDA compounds: Zytiga, Paliperidone, Targretin, and Irinotecan. We compared them with the control substrate, Carboxymethyllysine, which showed good binding interaction through hydrogen bonding, hydrophobic interactions, and π-stacking at active site residues of the target protein. Following a 100 ns simulation run, the docked complex revealed that the Root Mean Square Deviation (RMSD) values of two drugs, Irinotecan (1.3 ± 0.2 nm) and Paliperidone (1.2 ± 0.3 nm), were relatively stable. Subsequently, the Molecular Mechanics Poisson–Boltzmann Surface Area (MMPBSA) determined that the Paliperidone molecule had a high negative energy of −13.49 kcal/mol, and the Absorption, Distribution, Metabolism, and Excretion (ADME) properties were in control for use in the mentioned cases. We extended this with many in vitro studies, including an immunoblotting assay, which revealed that RAGEs with High Mobility Group Box 1 (HMGB1) showed higher expression, while RAGEs with Paliperidone showed lower expressions. Furthermore, cell proliferation assay and Apoptosis assay (Annexin-V/PI staining) results revealed that Paliperidone was an effective anti-glycation and anti-apoptotic drug—however, more extensive in vivo studies are needed before its use. Full article
(This article belongs to the Section Molecular Pharmacology)
Show Figures

Figure 1

Figure 1
<p>The (<b>A</b>) cartoon view of RAGEs and their active site (predicted with FTSite server), indicated by meshwork in red colour with domains highlighted differently to make it clearer, and (<b>B</b>) 1030 FDA drug binding energy with RAGEs plotted to make it clear against the number of the compounds and docking energy in kcal/mol.</p>
Full article ">Figure 2
<p>Interaction analysis of five binding dock poses in ribbon and 2D of (<b>A</b>) Zytiga (red), (<b>B</b>) Targretin (cyan), (<b>C</b>) Paliperidone (Green), (<b>D</b>) Irinotecan (blue), and (<b>E</b>) Carboxymethyllysine (green). The 2D analysis shows a black stick H-bond, and a green line indicates hydrophobic.</p>
Full article ">Figure 2 Cont.
<p>Interaction analysis of five binding dock poses in ribbon and 2D of (<b>A</b>) Zytiga (red), (<b>B</b>) Targretin (cyan), (<b>C</b>) Paliperidone (Green), (<b>D</b>) Irinotecan (blue), and (<b>E</b>) Carboxymethyllysine (green). The 2D analysis shows a black stick H-bond, and a green line indicates hydrophobic.</p>
Full article ">Figure 3
<p>Results of 100 ns MD simulation. (<b>A</b>) RMSD trajectory for Zytiga, Paliperidone, Targretin, Irinotecan, and Carboxymethyllysine. (<b>B</b>) The gyration radius (Rg) is the same colour as the RMSD.</p>
Full article ">Figure 4
<p>Showing the H-bond analysis (100 ns run) for the receptors for (<b>A</b>) Carboxymethyllysine, (<b>B</b>) Paliperidone, (<b>C</b>) Zytiga, (<b>D</b>) Irinotecan, and (<b>E</b>) Targretin. The colour-highlighted sticks represent the H-bond donor and acceptor atom pairs within a 0.35 nm range.</p>
Full article ">Figure 5
<p>DFT analysis of (<b>A</b>) Paliperidone (selected based on MD simulation and MMPBSA analysis) compared with (<b>B</b>) Carboxymethyllysine. HOMO and LUMO sites indicate electron donating and accepting affinity.</p>
Full article ">Figure 6
<p>The (<b>A</b>) representative dot blot showing the effect of Paliperidone on RAGE protein expression with/without HMGB1 stimulation in MCF7 cells; NC-negative control/untreated, VC-vehicle control (DMSO). (<b>B</b>) Corresponding bar graph showing the quantitative integrated density of the bands—* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 versus control, n = 3, data are presented as the means +/− SE of triplicate experiments. (<b>C</b>) Representative bar graph showing the effect of Paliperidone on the proliferation of MCF7 cells with/without HMGB1 stimulation where ‘0’ signifies untreated cells; * <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 versus control, n = 3, data are presented as the means +/− SE of triplicate experiments. (<b>D</b>) Corresponding histogram denoting IC50 (conc. which causes 50% reduction in cell viability) values for Paliperidone in presence and absence of HMGB1; * <span class="html-italic">p</span> &lt; 0.05, n = 3.</p>
Full article ">Figure 7
<p>The microscopic images show the effect of Paliperidone on MCF7 cell morphology in the absence/presence of HMGB1 (scale-200 μm, 10× magnification under Nikon E200 microscope, Nikon Instrumental Inc., Melville, NY, USA).</p>
Full article ">Figure 8
<p>The (<b>A</b>,<b>B</b>) representative scatter plots of PI (<span class="html-italic">y</span>-axis) versus Annexin V (<span class="html-italic">x</span>-axis), showing the effect of PALI (Paliperidone) on apoptosis of MCF7 cells with/without HMGB1 stimulation; DOX-doxorubicin, a well-known pro-apoptotic compound is taken as a positive control here. (<b>C</b>) Corresponding bar graph denoting the percentage of apoptotic cells due to Paliperidone in the presence/absence of HMGB1 (* <span class="html-italic">p</span> &lt; 0.05, n = 3, data are presented as the means +/− SE of triplicate experiments).</p>
Full article ">Figure 8 Cont.
<p>The (<b>A</b>,<b>B</b>) representative scatter plots of PI (<span class="html-italic">y</span>-axis) versus Annexin V (<span class="html-italic">x</span>-axis), showing the effect of PALI (Paliperidone) on apoptosis of MCF7 cells with/without HMGB1 stimulation; DOX-doxorubicin, a well-known pro-apoptotic compound is taken as a positive control here. (<b>C</b>) Corresponding bar graph denoting the percentage of apoptotic cells due to Paliperidone in the presence/absence of HMGB1 (* <span class="html-italic">p</span> &lt; 0.05, n = 3, data are presented as the means +/− SE of triplicate experiments).</p>
Full article ">Figure 8 Cont.
<p>The (<b>A</b>,<b>B</b>) representative scatter plots of PI (<span class="html-italic">y</span>-axis) versus Annexin V (<span class="html-italic">x</span>-axis), showing the effect of PALI (Paliperidone) on apoptosis of MCF7 cells with/without HMGB1 stimulation; DOX-doxorubicin, a well-known pro-apoptotic compound is taken as a positive control here. (<b>C</b>) Corresponding bar graph denoting the percentage of apoptotic cells due to Paliperidone in the presence/absence of HMGB1 (* <span class="html-italic">p</span> &lt; 0.05, n = 3, data are presented as the means +/− SE of triplicate experiments).</p>
Full article ">
24 pages, 18910 KiB  
Article
ADAMTS13 Improves Endothelial Function and Reduces Inflammation in Diabetic Retinopathy
by Ahmed M. Abu El-Asrar, Mohd I. Nawaz, Ajmal Ahmad, Mairaj Siddiquei, Eef Allegaert, Lowie Adyns, Lotte Vanbrabant, Priscilla W. Gikandi, Gert De Hertogh, Sofie Struyf and Ghislain Opdenakker
Cells 2025, 14(2), 85; https://doi.org/10.3390/cells14020085 - 9 Jan 2025
Viewed by 771
Abstract
The protease, a disintegrin and metalloproteinase with thrombospondin type 1 motif member 13 (ADAMTS13), known to cleave only the von Willebrand factor (VWF), has powerful regulatory effects on microvascular platelet adhesion, thrombosis, inflammation, and endothelial dysfunction. We study the protection against diabetes-induced retinal [...] Read more.
The protease, a disintegrin and metalloproteinase with thrombospondin type 1 motif member 13 (ADAMTS13), known to cleave only the von Willebrand factor (VWF), has powerful regulatory effects on microvascular platelet adhesion, thrombosis, inflammation, and endothelial dysfunction. We study the protection against diabetes-induced retinal injury in experimental rats by supplementation with recombinant ADAMTS13. We compare human epiretinal membranes and vitreous samples from nondiabetic subjects and patients with proliferative diabetic retinopathy (PDR) and extend in vitro analyses with the use of various immunodetection and spectrofluorimetric methods on rat retina and human retinal glial and endothelial cell cultures. Functional studies include the assessment of the blood–retinal barrier (BRB), cell adhesion, and in vitro angiogenesis. In epiretinal membranes, endothelial cells and monocytes/macrophages express ADAMTS13. The levels of VWF, the platelet marker CD41, ADAMTS13, and the biomarkers of endothelial cell injury soluble VE-cadherin and soluble syndecan-1 are increased in PDR vitreous. ADAMTS13 is downregulated in diabetic rat retinas. The intravitreal administration of ADAMTS13 attenuates diabetes-induced BRB breakdown, the downregulation of VE-cadherin and β-catenin, and the upregulation of VWF, CD41, phospho-ERK1/2, HMGB1, VCAM-1, and ICAM-1. In Müller cells, ADAMTS13 attenuates MCP-1, MMP-9, and ROS upregulation induced by diabetic mimetic conditions. In HRMECs, ADAMTS13 attenuates the shedding of the soluble VE-cadherin and soluble syndecan-1 and the levels of phospho-ERK1/2, MCP-1, fractalkine, and ROS induced by diabetic mimetic conditions, the upregulation of ICAM-1 and VCAM-1 elicited by TNF-α, the adherence of monocytes induced by TNF-α, and VEGF-induced migration of human retinal microvascular endothelial cells. Our findings suggest that enhancing ADAMTS13 levels in situ ameliorates diabetes-induced retinal inflammation and vascular dysfunction. Full article
Show Figures

Figure 1

Figure 1
<p>Epiretinal membranes from PDR patients contain endothelial and stromal cells expressing ADAMTS13. (<b>A</b>) Negative control slide showing no staining. (<b>B</b>) Staining for the endothelial cell marker CD31 showing new blood vessels (arrows). (<b>C</b>) Staining for CD68 identifying monocytes/macrophages in the stroma (arrows). (<b>D</b>) Staining for ADAMTS13 showing immunoreactivity in vascular endothelial cells (arrows) and in stromal cells (arrowheads). (<b>E</b>) Double immunohistochemical staining for ADAMTS13 (red) and CD68 (brown) showing co-expression in stromal cells (arrows). No counterstain to visualize the cell nuclei was applied (black scale bar, 10 µM).</p>
Full article ">Figure 2
<p>Hemostasis biomarkers in vitreous fluid of diabetes patients. Detection of thrombotic markers in vitreous fluid of patients with proliferative diabetic retinopathy (PDR). Determination of the von Willebrand factor (VWF) (<b>A</b>), the platelet marker CD41 (<b>B</b>), and ADAMTS13 (<b>C</b>) levels in vitreous fluid samples. A total of 15 µL of vitreous fluid samples from 12 patients with PDR and from 12 nondiabetic patients with rhegmatogenous retinal detachment (RD) was subjected to gel electrophoresis and the presence of VWF, CD41, and ADAMTS13 (5C11 monoclonal antibody) was illustrated by representative western blots and the levels of the antigens compared between the RD and PDR cohorts. Results are expressed as medians (interquartile range). (* <span class="html-italic">p</span> &lt; 0.05; Mann-Whitney test).</p>
Full article ">Figure 3
<p>Markers of endothelial injury and dysfunction are enhanced in the vitreous fluid of patients with proliferative diabetic retinopathy (PDR). Soluble vascular endothelial (VE)-cadherin (<b>A</b>) and soluble syndecan-1 (<b>B</b>) levels were quantified by ELISA and expressed as medians (interquartile ranges) in equal aliquots of vitreous fluid derived from 39 PDR patients and 36 nondiabetic patients with rhegmatogenous retinal detachment (RD) (* <span class="html-italic">p</span> &lt; 0.05; Mann–Whitney U test).</p>
Full article ">Figure 4
<p>ADAMTS13 expression levels in diabetic rat retinas and effects of exogenously administered ADAMTS13. ADAMTS13 expression levels in the retinal lysates of diabetic rats (<b>D</b>) (<span class="html-italic">n</span> = 12) and nondiabetic control animals (<span class="html-italic">n</span> = 12) were determined by western blot analysis. After the measurement of the intensities of ADAMTS13 proteoform bands, the immunoblots were stripped and reprobed to evaluate ß-tubulin intensities in all sample panels (<b>A</b>). Results are expressed as means ± standard deviation of the ratios between ADAMTS13 and ß-tubulin (* <span class="html-italic">p</span> &lt; 0.05; independent t-test). The effects of intravitreal ADAMTS13 injection on vascular permeability and markers of hemostasis and inflammation in rat retinas after streptozotocin-induced diabetes were evaluated by quantifications of the BRB breakdown by detection of FITC dextran seeped into the retina after the systemic injection (<b>B</b>). Retinal protein expression levels of the von Willebrand factor (VWF) (<b>C</b>), the platelet marker CD41 (<b>D</b>), vascular endothelial (VE)-cadherin (<b>E</b>), and ß-catenin (<b>F</b>) were determined by immunoblot analysis. Statistical comparisons (mean ± standard deviation of 8–10 rats) were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from nondiabetic controls. # <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from diabetic rats.</p>
Full article ">Figure 5
<p>Intravitreal administration of ADAMTS13 reduces retinal inflammation in diabetic rats. The relative protein amounts of phospho-ERK1/2 (<b>A</b>), high-mobility group box-1 (HMGB1) (<b>B</b>), vascular cell adhesion molecule-1 (VCAM-1) (<b>C</b>), and intercellular adhesion molecule-1 (ICAM-1) (<b>D</b>) were determined in rat retinas with the use of western blots. The animals were made diabetic with the use of a single streptozotocin bolus, ADAMTS13 was injected in the vitreous, and its effects on inflammation markers were evaluated by comparison of ADAMTS13-injected with the contralateral PBS-injected eyes in single animals. Statistical comparisons (mean standard deviation of 8–10 rats in each group) were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.05 compared with nondiabetic controls. # <span class="html-italic">p</span> &lt; 0.05 compared with diabetic rats.</p>
Full article ">Figure 6
<p>Regulation of proangiogenic and inflammatory molecule expression by ADAMTS13 in human retinal Müller glial cells. Human glial cells were left untreated or treated with high glucose (HG) (25 mM), tumor necrosis factor-α (TNF-α) (1 ng/mL), or cobalt chloride (CoCl<sub>2</sub>) (300 µM) for 24 h or ADAMTS13 (100 ng/mL) for 1h followed by HG, CoCl<sub>2</sub>, or TNF-α. A total of 25 mM of mannitol was used as an inert control for osmotic effects by HG treatment. Levels of monocyte chemotactic protein-1 (MCP-1), matrix metalloproteinase-9 (MMP-9), and vascular endothelial growth factor (VEGF) were quantified in the culture media by ELISA. The present data were generated from three different experiments, each performed in triplicates, and the results are provided as means ± standard deviation; statistical comparisons were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.05 indicates the comparisons with values obtained from control cells. # <span class="html-italic">p</span> &lt; 0.05 documents the differences with values obtained from cells treated with HG, TNF-α, or CoCl<sub>2</sub>.</p>
Full article ">Figure 7
<p>ADAMTS13 promotes the integrity of adherens junctions in human retinal microvascular endothelial cells. Retinal endothelial cells were left untreated (control) or were cultured in the presence of 25 mM of high glucose (HG) concentrations (<b>A</b>), 300 µM of cobalt chloride (CoCl<sub>2</sub>) (<b>B</b>), or 1 ng/mL of TNF-α (<b>C</b>) for 24 h. A third type of treatment consisted of the pretreatment with 100 ng/mL of ADAMTS13 for 1 h followed by HG, CoCl<sub>2</sub>, or TNF-α. A total of 25 mM of mannitol was used as a control for the treatment with high glucose. Levels of soluble VE-cadherin (left histograms) and soluble syndecan-1 (right histograms) in cell culture media were quantified with the use of specific ELISAs. The data represent means ± standard deviations from different (<span class="html-italic">n</span> = 3) experiments performed in triplicates, and statistical comparisons were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.05 indicates comparisons with values obtained from control cells. # <span class="html-italic">p</span> &lt; 0.05 provides the comparisons with values obtained from cells treated with HG, CoCl<sub>2</sub>, or TNF-α.</p>
Full article ">Figure 8
<p>Regulation of inflammatory cytokine expression by ADAMTS13 in human retinal microvascular endothelial cells. Endothelial cultures were left untreated (control) or were treated with 25 mM of high glucose (HG), 300 µM of cobalt chloride (CoCl<sub>2</sub>), or 1 ng/mL of tumor necrosis factor-α (TNF-α) for 24 h with or without a 1 h preincubation with 100 ng/mL of ADAMTS13. A total of 25 mM of mannitol was used as control for cell damage induced by high glucose levels. In panel (<b>A</b>), signaling events were probed by measurement of phospho-ERK1/2 levels in cell lysates. In panel (<b>B</b>), cell culture medium levels of monocyte chemotactic protein-1 (MCP-1) and, in panel (<b>C</b>), cell culture medium levels of fractalkine were quantified with the use of specific ELISAs. Data are expressed as means ± standard deviation from independent (<span class="html-italic">n</span> = 3) experiments with triplicates per experiment. Statistical comparisons were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.05 indicates comparisons with values obtained from control cells. # <span class="html-italic">p</span> &lt; 0.05 indicates comparisons with values obtained from stimulated cells.</p>
Full article ">Figure 9
<p>ADAMTS13 affects leukocyte adhesion to human retinal microvascular endothelial cells (HRMECs). HRMECs were left untreated or were stimulated with 1 ng/mL of tumor necrosis factor-α (TNF-α) for 24 h with or without a 1 h preincubation with ADAMTS13 (100 ng/mL). Monocyte adhesion to HRMEC monolayers was assessed with the use of fluorescently labeled THP-1 monocytic cells (<b>A</b>). The effects of exogenous ADAMTS13 on protein expression levels of intercellular adhesion molecule-1 (ICAM-1) (<b>B</b>) and vascular cell adhesion molecule-1 (VCAM-1) (<b>C</b>) were determined with the use of western blots. Results are expressed as means ± standard deviation from three different experiments performed in triplicates. One-way ANOVA and independent t-tests were used for comparisons among three groups and between two groups, respectively. * <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from untreated cells. # <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from cells treated with TNF-α (RFU = relative fluorescence units).</p>
Full article ">Figure 10
<p>ADAMTS13 reduces cellular oxidative stress. Oxidative stress was induced in HRMECs and human retinal Müller glial cells and monitored with the use of 2′-7′-dichlorofluorescein (DCF) fluorescence intensity analysis. The in vitro effects of cell pretreatment with 100 ng/mL of ADAMTS13 for 1 h were quantified. HRMECs were left untreated (control) or were treated with 25 mM of high glucose (HG) for 24 h. A total of 25 mM of mannitol was used as a control condition for the HG treatment (<b>A</b>). Human retinal Müller glial cells (<b>B</b>) and HRMECs (<b>C</b>) were left untreated or were treated with 10 mM of hydrogen peroxide (H<sub>2</sub>O<sub>2</sub>) for 1 h. Data are provided as medians (interquartile range) from independent (<span class="html-italic">n</span> = 3) experiments, each performed in triplicates. Statistical comparisons were performed as described in <a href="#sec2dot12-cells-14-00085" class="html-sec">Section 2.12</a>. * <span class="html-italic">p</span> &lt; 0.5 indicates comparisons with values obtained from control cells. # <span class="html-italic">p</span> &lt; 0.05 indicates comparisons with values obtained from cells treated with HG or H<sub>2</sub>O<sub>2</sub>.</p>
Full article ">Figure 11
<p>Inhibition of endothelial cell migration by ADAMTS13. (<b>A</b>,<b>B</b>) Confluent monolayers of overnight starved HRMECs were scratched with sterile micropipette tips and monolayer regeneration was microscopically monitored, subject to various treatments. In one set of experiments, the cultures were pretreated with a dilution medium or ADAMTS13 (60, 200, or 600 ng/mL) for 1 h, followed by stimulation with vascular endothelial growth factor (VEGF) (10 ng/mL) for 16 h. Two independent experiments were performed in duplicates and two-to-three independent field images were taken for the migration analysis with the Image J software (summarized in (<b>B</b>)). In panel (<b>A</b>), representative images illustrate the effect of ADAMTS13, at a dose of 600 ng/mL, on VEGF-induced cell migration. (<b>C</b>) In a second set of experiments, endothelial cell migration through 8 µm pores of polyethylene terephthalate (PET) membranes in response to VEGF (10 ng/mL) with or without pretreatment with ADAMTS13 (6 to 600 ng/mL) was analyzed with the xCELLigence instrument (three or four independent experiments in duplicates). Results are expressed as means ± standard deviation. One-way ANOVA and independent t-tests were used for comparisons among five groups and between two groups, respectively. * <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from untreated cells. # <span class="html-italic">p</span> &lt; 0.05 compared with values obtained from cells treated with VEGF only.</p>
Full article ">
35 pages, 3838 KiB  
Review
Roles of the Receptor for Advanced Glycation End Products and Its Ligands in the Pathogenesis of Alzheimer’s Disease
by Wen Li, Qiuping Chen, Chengjie Peng, Dan Yang, Si Liu, Yanwen Lv, Langqi Jiang, Shijun Xu and Lihua Huang
Int. J. Mol. Sci. 2025, 26(1), 403; https://doi.org/10.3390/ijms26010403 - 5 Jan 2025
Viewed by 1070
Abstract
The Receptor for Advanced Glycation End Products (RAGE), part of the immunoglobulin superfamily, plays a significant role in various essential functions under both normal and pathological conditions, especially in the progression of Alzheimer’s disease (AD). RAGE engages with several damage-associated molecular patterns (DAMPs), [...] Read more.
The Receptor for Advanced Glycation End Products (RAGE), part of the immunoglobulin superfamily, plays a significant role in various essential functions under both normal and pathological conditions, especially in the progression of Alzheimer’s disease (AD). RAGE engages with several damage-associated molecular patterns (DAMPs), including advanced glycation end products (AGEs), beta-amyloid peptide (Aβ), high mobility group box 1 (HMGB1), and S100 calcium-binding proteins. This interaction impairs the brain’s ability to clear Aβ, resulting in increased Aβ accumulation, neuronal injury, and mitochondrial dysfunction. This further promotes inflammatory responses and oxidative stress, ultimately leading to a range of age-related diseases. Given RAGE’s significant role in AD, inhibitors that target RAGE and its ligands hold promise as new strategies for treating AD, offering new possibilities for alleviating and treating this serious neurodegenerative disease. This article reviews the various pathogenic mechanisms of AD and summarizes the literature on the interaction between RAGE and its ligands in various AD-related pathological processes, with a particular focus on the evidence and mechanisms by which RAGE interactions with AGEs, HMGB1, Aβ, and S100 proteins induce cognitive impairment in AD. Furthermore, the article discusses the principles of action of RAGE inhibitors and inhibitors targeting RAGE-ligand interactions, along with relevant clinical trials. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>String representation of RAGE–ligand interactions. To ensure understanding, this representation includes exclusively the most significant and extensively researched protein interactions. The highlighted interactions consist of AGER (RAGE), HMGB1, the S100 family of calcium-binding proteins (S100B, S100P, S100A1, S100A4, S100A12), DIAPH1, RhoA, TLR9, MyD88, TIRP4, IRAK4, EGFR, GR-B2, FGFR1, BDNF, FGF2, NRP1, NCK1, Aβ, FPR1, and FPR2. Abbreviations: RAGE, the receptor for advanced glycation end products; HMGB1, high mobility group box 1; DIAPH1, protein diaphanous homolog 1; RhoA, ras homolog family member A; TLR9, toll-like receptor 9; MyD88, myeloid differentiation primary response 88; TIRP4, toll/interleukin-1 receptor domain-containing protein 4; IRAK4, interleukin-1 receptor-associated kinase 4; EGFR, epidermal growth factor receptor; GR-B2, glucocorticoid receptor B2; FGFR1, fibroblast growth factor receptor 1; BDNF, brain-derived neurotrophic factor; FGF2, fibroblast growth factor 2; NRP1, neuropilin-1; NCK1, non-catalytic region of tyrosine kinase adaptor protein 1; Aβ, beta-amyloid peptide; FPR1, fMet-Leu-Phe receptor; FPR2, n-formyl peptide receptor 2. Source: <a href="https://string-db.org/" target="_blank">https://string-db.org/</a> (Accessed on 30 August 2024).</p>
Full article ">Figure 2
<p>Structure and isoforms of RAGE. As part of the immunoglobulin superfamily, RAGE is categorized among cell surface proteins. It is a type I membrane protein that spans the membrane once and features one variable (V) sector along with two constant (C1 and C2) immunoglobulin-like domains in its external cellular environment. Following this is a transmembrane region, along with a cytoplasmic domain that has a high charge density. The primary isoforms of RAGE comprise full-length RAGE, cleaved RAGE (cRAGE), endogenous secretory RAGE (esRAGE), N-terminal truncated RAGE (N-RAGE), and dominant-negative RAGE (DN-RAGE) (Created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a>) (Accessed 27 September 2024).</p>
Full article ">Figure 3
<p>RAGE and its ligands interaction signaling pathways. Following the activation of the AGE-RAGE signaling cascade, the stimulation of the ERK1 and PI3K pathways can activate the nuclear factor NF-κB. This factor then translocates to the nucleus, promoting ROS production, which ultimately leads to cellular damage and mitochondrial dysfunction. NF-κB is capable of translocating to the nucleus, where it facilitates the transcription of pro-inflammatory cytokines and chemokines, including TNF-α, IL-6, MCP-1, VCAM-1, and ICAM-1. This process promotes inflammation and activates immune cells. AGEs can also activate p38 MAPK and RhoA kinase, which modifies the shape of the endothelial F-actin cytoskeleton and enhances the permeability of the endothelial cell monolayer. HMGB1 binding to RAGE acts as a key initiator of neuroinflammation and can also bind to TLR4, participating in neuroinflammation and neurotoxicity. The interaction between HMGB1 and RAGE initiates the activation of MAPK, which facilitates the translocation of TLR4 to the cell surface. Moreover, TLR4 at the cell membrane can interact with HMGB1, leading to the transcription and translation of RAGE, which in turn promotes the translocation of RAGE to the cell surface. HMGB1 inhibits microglial activation and phagocytosis through RAGE, promoting the accumulation of neuronal Aβ. HMGB1 activates astrocytes, increasing iNOS expression in cortical astrocytes via TLR4 signaling. S100A4 binds to RAGE and initiates a signaling cascade that ultimately causes MMP-13 release through the NF-κB pathway in chondrocytes. Two neurotrophic motifs on S100A4 activate the JAK/STAT pathway, preventing neurodegeneration. S100A6 promotes neuronal apoptosis through its interaction with RAGE, which triggers activation of JNK dependent on ROS as well as caspases-3 and -7. S100A12 binding to RAGE increases the activation and discharge of NF-κB, IL-1β, IL-6, and TNF-α. S100B accumulates at the RAGE receptor sites, resulting in the initiation of downstream MAPK pathways, which subsequently leads to the phosphorylation and activation of NF-κB. Stimulation of S100B/RAGE signaling can promote the shedding of the endothelial glycocalyx by enhancing the expression, translocation, and activity of the sheddase ADAM17 in endothelial cells. Abbreviations: RAGE, the receptor for advanced glycation end products; ERK, extracellular signal-regulated kinase; PI3K, phosphatidylinositol-3 kinase; ROS, reactive oxygen species; NF-κB, nuclear factor-kappa B; TNF-α, tumor necrosis factor-alpha; IL-6, interleukin-6; MCP-1, monocyte chemoattractant protein-1; VCAM-1, vascular cell adhesion molecule-1; ICAM-1, intercellular adhesion molecule-1; AGEs, advanced glycation end products; p38 MAPK, p38 mitogen-activated protein kinase; RhoA, Rho family GTPase A; HMGB1, high mobility group box 1; TLR4, toll-like receptor 4; MAPK, mitogen-activated protein kinase; Aβ, beta-amyloid peptide; iNOS, inducible nitric oxide synthase; MMP-13, matrix metalloproteinase-13; JAK/STAT, Janus kinase/signal transducer and activator of transcription; JNK, c-Jun N-terminal kinase; ADAM17, a disintegrin and metalloproteinase 17 (Created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a>). The arrow colors correspond to the colors of the ligands in the figure, each activating different signaling pathways (Accessed on 2 January 2025).</p>
Full article ">Figure 4
<p>Aβ(Beta-amyloid peptide) transport across the normal and disrupted blood–brain barrier (BBB). RAGE mediates the transport of circulating Aβ from plasma to the brain across the BBB. Disruption of the BBB impairs the vascular clearance of brain Aβ and may increase the influx of peripheral Aβ into the brain. (Created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a>). (Accessed on 2 January 2025).</p>
Full article ">
17 pages, 2166 KiB  
Article
Immunogenic Cell Death Traits Emitted from Chronic Lymphocytic Leukemia Cells Following Treatment with a Novel Anti-Cancer Agent, SpiD3
by Elizabeth Schmitz, Abigail Ridout, Audrey L. Smith, Alexandria P. Eiken, Sydney A. Skupa, Erin M. Drengler, Sarbjit Singh, Sandeep Rana, Amarnath Natarajan and Dalia El-Gamal
Biomedicines 2024, 12(12), 2857; https://doi.org/10.3390/biomedicines12122857 - 16 Dec 2024
Viewed by 1105
Abstract
Background: Targeted therapies (e.g., ibrutinib) have markedly improved chronic lymphocytic leukemia (CLL) management; however, ~20% of patients experience disease relapse, suggesting the inadequate depth and durability of these front-line strategies. Moreover, immunotherapeutic success in CLL has been stifled by its pro-tumor microenvironment milieu [...] Read more.
Background: Targeted therapies (e.g., ibrutinib) have markedly improved chronic lymphocytic leukemia (CLL) management; however, ~20% of patients experience disease relapse, suggesting the inadequate depth and durability of these front-line strategies. Moreover, immunotherapeutic success in CLL has been stifled by its pro-tumor microenvironment milieu and low mutational burden, cultivating poor antigenicity and limited ability to generate anti-tumor immunity through adaptive immune cell engagement. Previously, we have demonstrated how a three-carbon-linker spirocyclic dimer (SpiD3) promotes futile activation of the unfolded protein response (UPR) in CLL cells through immense misfolded-protein mimicry, culminating in insurmountable ER stress and programmed CLL cell death. Method: Herein, we used flow cytometry and cell-based assays to capture the kinetics and magnitude of SpiD3-induced damage-associated molecular patterns (DAMPs) in CLL cell lines and primary samples. Result: SpiD3 treatment, in vitro and in vivo, demonstrated the capacity to propagate immunogenic cell death through emissions of classically immunogenic DAMPs (CALR, ATP, HMGB1) and establish a chemotactic gradient for bone marrow-derived dendritic cells. Conclusions: Thus, this study supports future investigation into the relationship between novel therapeutics, manners of cancer cell death, and their contributions to adaptive immune cell engagement as a means for improving anti-cancer therapy in CLL. Full article
Show Figures

Figure 1

Figure 1
<p>CLL cells display ecto-CALR following SpiD3 treatment. HG-3 ((<b>A</b>,<b>B</b>); n = 3); OSU-CLL ((<b>C</b>,<b>D</b>); n = 3); or patient-derived CLL ((<b>E</b>,<b>F</b>); n = 5) cells were treated with vehicle (Veh), SpiD3 (0.25–2 µM), FeCl<sub>2</sub> (160 μM), or the positive control, etoposide (Etop; 20 µM) for the indicated durations. Viable cells were analyzed by flow cytometry for changes in surface CALR expression (ecto-CALR). Primary patient-derived CLL cells were additionally designated as CD19+/CD5+ by flow cytometry. Data are presented as mean ± SEM. Comparisons across treatment groups were analyzed with respect to the vehicle by one-way ANOVA. Asterisks denote magnitude of significance: * <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 2
<p>SpiD3 treatment evokes extracellular ATP release. HG-3 (<b>A</b>); and OSU-CLL (<b>B</b>) cells were treated over 24 h (n = 3) with vehicle (Veh), SpiD3 (0.5–2 µM), or the positive control, etoposide (Etop; 20 µM). Extracellular ATP measurements at 8, 16, and 24 h were parsed out to evaluate the average extracellular ATP measured at these timepoints in comparison to the matched timepoint vehicle. Data are presented as mean ± SEM. Comparisons across treatment groups were analyzed with respect to the matched timepoint average vehicle by one-way ANOVA. Asterisks denote magnitude of significance: * <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; 0.0001.</p>
Full article ">Figure 3
<p>SpiD3-treated cells release extracellular HMGB1. Supernatant from HG-3 ((<b>A</b>,<b>B</b>); n = 3); OSU-CLL ((<b>C</b>,<b>D</b>); n = 3); and primary CLL ((<b>E</b>); n = 10) cells were evaluated for extracellular HMGB1 after 24 h or 48 h of treatment with the vehicle (Veh), SpiD3 (0.5–2 µM), ibrutinib (1 µM), or positive control, etoposide (Etop; 20 µM). Data are presented as mean ± SEM. Comparisons across treatment groups were analyzed with respect to the vehicle by one-way ANOVA. Asterisks denote magnitude of significance: * <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 4
<p>Chemotactic potential of SpiD3-treated cell supernatants. Bone marrow dendritic cells (BMDCs) were allowed to migrate for 6 h toward supernatant collected from HG-3 (<b>A</b>); and OSU-CLL (<b>B</b>) cells after 24 h treatment with the vehicle (Veh), SpiD3 (0.5–2 µM), or the positive control, etoposide (Etop; 20 µM). GM-CSF (20 ng/mL) stimulated media, and supernatant derived from heat-shocked CLL cells (HS) served as positive chemotactic controls. The number of migrated BMDCs were counted via flow cytometry analysis (n = 3). The chemotactic index is a comparison of the migrated events observed from treatment conditions to that of the vehicle condition. Data are represented as mean ± SEM. Comparisons across treatment groups were analyzed with respect to the vehicle by one-way ANOVA. Asterisks denote magnitude of significance: * <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 5
<p><span class="html-italic">In vivo</span> SpiD3 treatment yields an immunostimulatory response. (<b>A</b>) Schematic of experiment design: Eµ-TCL1 mice with comparable leukemia burden were treated intravenously with SpiD3 prodrug (SpiD3_AP, 10 mg/kg; n = 6) or equivalent vehicle (Veh; 50% PEG400, 10% DMSO, 40% water; n = 5) once daily for 3 days, as previously reported [<a href="#B20-biomedicines-12-02857" class="html-bibr">20</a>]. Following treatment, spleen cells were collected for flow cytometry analysis and plasma was isolated from murine blood; (<b>B</b>) leukemic (CD19+/CD5+) cells from murine spleens were analyzed by flow cytometry for changes in surface CALR expression (ecto-CALR) and compared to the percentage of leukemic cells detected in spleens of the same mice (as reported in Eiken, et al. [<a href="#B20-biomedicines-12-02857" class="html-bibr">20</a>]). The concentrations of plasma inflammatory cytokines and chemokines were assessed using Mouse Anti-Virus Response (<b>C</b>,<b>E</b>); and Mouse Pro-Inflammatory Chemokine (<b>D</b>,<b>F</b>) LEGENDplex™ flow cytometry-based multiplex immunoassays. (<b>C</b>,<b>D</b>) Heatmaps display fold change in the plasma analyte concentration compared to the average of vehicle-treated mice. Columns represent individual mice per treatment group. (<b>E</b>,<b>F</b>) Raw plasma analyte concentration and correlation with the percentage of CD19+/CD5+ spleen-derived cells are shown for select analytes. Individual data points (Veh = black circles; SpiD3_AP = blue triangles) in addition to summary statistics (mean ± SEM) are shown. Comparisons between treatment groups were analyzed by unpaired <span class="html-italic">t</span>-test. Asterisks denote magnitude of significance: * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Illustrative summary of SpiD3 anti-leukemic activity. CLL cell cytotoxicity via SpiD3 is demonstrated by: (i) inhibition of NF-κB signaling; and (ii) accumulation of unfolded proteins, promoting ER stress, activating a futile UPR and, subsequently, the associated programmed cell death pathways. ER stress is a proposed prerequisite for immunogenic DAMP emissions; we hypothesize it is this facet of SpiD3-associated effects that result in detectable hallmarks of immunogenic cell death from CLL cells. This diagram is adapted from Eiken, et al. CLL, chronic lymphocytic leukemia; DC, dendritic cell; iDAMP, immunogenic damage-associated molecular pattern; ER, endoplasmic reticulum; UPR, unfolded protein response.</p>
Full article ">
16 pages, 1348 KiB  
Article
Genome-Wide Analysis Reveals Key Genes and MicroRNAs Related to Pathogenic Mechanism in Wuchereria bancrofti
by Caoli Zhu, Yicheng Yan, Yaning Feng, Jiawei Sun, Mingdao Mu and Zhiyuan Yang
Pathogens 2024, 13(12), 1088; https://doi.org/10.3390/pathogens13121088 - 10 Dec 2024
Viewed by 841
Abstract
Wuchereria bancrofti is a parasite transmitted by mosquitoes and can cause a neglected tropical disease called Lymphatic filariasis. However, the genome of W. bancrofti was not well studied, making novel drug development difficult. This study aims to identify microRNA, annotate protein function, and [...] Read more.
Wuchereria bancrofti is a parasite transmitted by mosquitoes and can cause a neglected tropical disease called Lymphatic filariasis. However, the genome of W. bancrofti was not well studied, making novel drug development difficult. This study aims to identify microRNA, annotate protein function, and explore the pathogenic mechanism of W. bancrofti by genome-wide analysis. Novel miRNAs were identified by analysis of expressed sequence tags (ESTs) from this parasite. Protein homology was obtained by a bidirectional best-hit strategy using BLAST. By an EST-based method, we identified 20 novel miRNAs in the genome. The AU content of these miRNAs ranged from 39.7% to 80.0%, with a mean of 52.9%. Among them, 14 miRNA homologs were present in mammal genomes, while six miRNA homologs were present in non-mammal genomes. By conducting a detailed sequence alignment using BLAST, we have successfully annotated the functions of 75 previously unannotated proteins, enhancing our understanding of the proteome and potentially revealing new targets for therapy. Homology distribution analysis indicated that a set of critical proteins were present in parasites and mosquitoes, but not present in mammals. By searching the literature, ten proteins were found to be involved in the pathogenic infection process of W. bancrofti. In addition, the miRNA–gene network analysis indicated that two pathogenic genes (CALR and HMGB2) are regulated by newly identified miRNAs. These genes were supposed to play key roles in the infection mechanism of W. bancrofti. In conclusion, our genome-wide analysis provided new clues for the prevention and treatment of W. bancrofti infection. Full article
(This article belongs to the Section Parasitic Pathogens)
Show Figures

Figure 1

Figure 1
<p>Five identified miRNA hairpin structures of <span class="html-italic">W. bancrofti</span>. The sequence highlighted in blue is the mature sequence of the miRNA.</p>
Full article ">Figure 2
<p>Venn diagrams of <span class="html-italic">W. bancrofti</span> homologous proteins in different groups. (<b>A</b>) Venn diagram of homologs in two parasites (<span class="html-italic">Brugia malayi</span> and <span class="html-italic">Brugia timori</span>); (<b>B</b>) Venn diagram of homologs in four mosquito vectors; (<b>C</b>) Venn diagram of homologs in three mammals; (<b>D</b>) Venn diagram of parasite, mosquito, and mammal.</p>
Full article ">Figure 3
<p>The miRNA–gene interaction network diagram. The miRNAs are shown by light orange nodes, pathogenic genes are shown by light yellow nodes and other genes are shown by light blue nodes.</p>
Full article ">
12 pages, 1866 KiB  
Article
Potential Serum HMGB1, HSP90, and S100A9 as Metastasis Predictive Biomarkers for Cancer Patients and Relevant Cytokines: A Pilot Study
by Worawat Songjang, Chatchai Nensat, Wittawat Jitpewngarm and Arunya Jiraviriyakul
Int. J. Mol. Sci. 2024, 25(24), 13232; https://doi.org/10.3390/ijms252413232 - 10 Dec 2024
Viewed by 923
Abstract
Metastatic cancer is still one of the leading causes of death worldwide despite significant advancements in diagnosis and treatment. Biomarkers are one of the most promising diagnostic tools that are used alongside traditional diagnostic tools in cancer patients. DAMPs are intracellular molecules released [...] Read more.
Metastatic cancer is still one of the leading causes of death worldwide despite significant advancements in diagnosis and treatment. Biomarkers are one of the most promising diagnostic tools that are used alongside traditional diagnostic tools in cancer patients. DAMPs are intracellular molecules released in response to cellular stress, tissue injury, and cell death. There have been shown to be associated with worsening prognosis among such patients, and some DAMPs could potentially be used as predictive biomarkers of metastatic status. The goal of this study is to investigate DAMP expression and the probability that certain DAMPs could be predictive biomarkers of the metastatic stage in various cancer types. Forty cancer patients at Naresuan University Hospital, Thailand, were enrolled. Then, an investigation of HSP90, HMGB1, S100A9, and ATP expression and cytokine/chemokine profiling in serum was performed using an immunological-based assay. We assessed the predictive biomarker candidates and the association between DAMP expression and cytokines/chemokines using an ROC curve analysis and a correlation regression analysis. The results showed that HSP90 has strong potential as a metastatic predictive biomarker, with a cutoff value of 25.46 ng/mL (AUC 0.8207, sensitivity 82.61%, specificity 75.00%, 95% CI 0.6860–0.9553). This was followed by HMGB1 and S100A9, which exhibited sensitivity of 82.61 and 65.22%, and specificity of 68.75 and 56.25%, respectively. Interestingly, the candidate DAMPs negatively correlate with various serum cytokines, for example, HMGB1 vs. IL-15 (slope 88.05, R 0.3297, p-value 0.005), HMGB1 vs. IFN-γ (slope 2.235, R 0.3052, p-value 0.0013) and HSP90 vs. IFN-γ (slope 0.0614, R 0.2187, p-value 0.008), suggesting that they are highly elevated in advanced metastatic tumors, which is possibly associated with the immunomodulation effect. We postulated that HSP90, HMGB1, and S100A9 have the potential to be predictive biomarkers for supporting tumor metastasis categorization using histopathology. Full article
Show Figures

Figure 1

Figure 1
<p>Serum candidate DAMP profiling according to TNM staging. Candidate DAMPs’ concentration levels in serum were determined with ELISA kit. All data are shown as individual mean values, M (mean values) ± SEM (standard error deviation). A significance threshold of <span class="html-italic">p</span>-value &lt; 0.05 was considered as statistical significance. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001; ns: <span class="html-italic">p</span> &gt; 0.05.</p>
Full article ">Figure 2
<p>ROC analysis of candidate DAMPs for biomarkers of tumor metastatic status. ROC curve analysis of candidate DAMPs in predictive discrimination of M0 and M1 patients.</p>
Full article ">Figure 3
<p>Cytokine and chemokine profiling of cancer patients’ serum according to tumor metastatic status. (<b>A</b>) Cytokines and chemokines were clustered according to their main deduced functions, including Th1/2 cytokines. (<b>B</b>) Analysis of selected cytokines, including IL-15 and IFN-γ in different stages of tumor metastasis. A significance threshold of <span class="html-italic">p</span>-value &lt; 0.05 was considered as statistical significance. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01.</p>
Full article ">
18 pages, 3482 KiB  
Article
A High-Throughput Immune-Oncology Screen Identifies Immunostimulatory Properties of Cytotoxic Chemotherapy Agents in TNBC
by Kennady K. Bullock, Thomas Hasaka, Emily Days, Joshua A. Bauer, Patricia A. Ward and Ann Richmond
Cancers 2024, 16(23), 4075; https://doi.org/10.3390/cancers16234075 - 5 Dec 2024
Viewed by 1038
Abstract
Background: Triple-negative breast cancers (TNBCs) typically have a greater immune cell infiltrate and are more likely to respond to immune checkpoint inhibition (ICI) than ER+ or HER2+ breast cancers. However, there is a crucial need to optimize combining chemotherapy strategies with ICI to [...] Read more.
Background: Triple-negative breast cancers (TNBCs) typically have a greater immune cell infiltrate and are more likely to respond to immune checkpoint inhibition (ICI) than ER+ or HER2+ breast cancers. However, there is a crucial need to optimize combining chemotherapy strategies with ICI to enhance overall survival in TNBC. Methods: Therefore, we developed a high-throughput co-culture screening assay to identify compounds that enhance CD8+ T-cell-mediated tumor cell cytotoxicity. Over 400 FDA-approved compounds or agents under investigation for oncology indications were included in the screening library. Results: Four chemotherapy agents were chosen as priority hits for mechanistic follow-up due to their ability to enhance T-cell-mediated cytotoxicity at multiple doses and multiple time points: paclitaxel, bleomycin sulfate, ispinesib, and etoposide. Lead compounds affected the expression of MHCI, MHCII, and PD-L1 and induced markers of immunogenic cell death (extracellular ATP or HMGB1). Conclusions: Based on the ability to increase tumor cell susceptibility to T-cell-mediated cytotoxicity while minimizing T-cell toxicity, bleomycin was identified as the most promising lead candidate. Overall, the results of these studies provide mechanistic insight into potential new chemotherapy partners to enhance anti-PD-1 efficacy in TNBC patients. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Figure 1

Figure 1
<p>Schematic of experimental workflow from initial high-throughput screening strategy to validation of compounds of interest.</p>
Full article ">Figure 2
<p>Schematic of high-throughput screening experimental timeline.</p>
Full article ">Figure 3
<p>Selection of high-priority compounds (<b>a</b>) Compounds with a Tf &gt; 3 at more than one concentration for each time point (<b>b</b>) Average AUC of Tf v concentration across three time points plotted v standard deviation. Colored compounds chosen for detailed mechanistic follow-up due to clinical relevance.</p>
Full article ">Figure 4
<p>Confirmation of increased T-cell-mediated cytotoxicity. CellTiterGlo viability assay of PyMT-OVA cells after 48 h of treatment of (<b>a</b>) PTX (<b>b</b>) Isp (<b>c</b>) Bleo (<b>d</b>) Etop with or without CD8+ T-cells co-cultured at a 1:5 (T-cell:tumor cell) ratio. T-cell groups were compared to DMSO+T using a one-way ANOVA with Tukey’s post hoc analysis. For each treatment concentration, T-cell versus no T-cell conditions were compared using an unpaired <span class="html-italic">t</span>-test. ns: no significance, * = <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 (<b>e</b>). Apoptosis assessed via FACS analysis of staining for Annexin V and 7-AAD. CD8+ T-cells stained with CellTrace blue and excluded by gating (<b>f</b>). representative flow plots of Annexin V and 7-AAD staining of PyMT-OVA tumor cells.</p>
Full article ">Figure 5
<p>Effect of lead compounds on MHCI, MHCII, and PD-L1 expression by tumor cells. (<b>a</b>) PyMT-OVA tumor cells were treated with indicated compounds at 50 nM for 48 h, and MHCI (H2kb) expression was assessed via flow cytometry. (<b>b</b>) PTX and Bleo tested at a range of doses for 48 h. MHCI induction assessed via flow cytometry. (<b>c</b>) CellTiterBlue viability assay of OT-1 CD8+ T-cells treated with indicated compounds for 48 h. IC50 values were calculated using a non-linear regression of the data plotted at log10[drug] versus fluorescence using Prism version 10.1.0. (<b>d</b>) MHCII expression assessed via flow cytometry after 48 h of treatment with indicated compounds at 50 nM. (<b>e</b>) PD-L1 expression assessed via flow cytometry after 48 h of treatment of indicated compounds at 50 nM. Treatment groups were compared to the DMSO control using a one-way ANOVA and Tukey’s post hoc analysis. * = <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 6
<p>Induction of markers of ICD. (<b>a</b>) Extracellular ATP and (<b>b</b>) HMGB1 measured after 24 h of treatment with indicated compounds. Treatment conditions were compared to the DMSO control using a one-way ANOVA and Tukey’s post hoc analysis. * = <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 7
<p>Effect of lead compounds on CD8+ T-cells. (<b>a</b>) Luminex cytokine array analysis of supernatants from OT-1 CD8+ T-cells treated with the indicated compounds at 50 nM for 48 h. n = 3 biological replicates. Data from IFNy and TNF are shown in expanded view. (<b>b</b>) CellTiterGlo viability assay of OT-1 CD8+ T-cells treated with indicated compounds for 48 h. IC50 values were calculated using a non-linear regression of the data plotted at log10[drug] versus luminescence using Prism version 10.1.0. Values presented as mean plus SD of three independent experiments. (<b>c</b>) CellTracker Assessment of OT-1 CD8+ T-cell proliferation via CellTracker proliferation assay. Mean fluorescent intensity (MFI) measured from samples at 24 h, 48 h, and 72 h. For each time point, treatment conditions were compared to the corresponding DMSO control using a one-way ANOVA and Tukey’s post hoc analysis. ** = <span class="html-italic">p</span> &lt; 0.01. Representative plot of fluorescent intensity peak shown.</p>
Full article ">Figure 8
<p>Validation of PTX and Bleo in 3D culture models. (<b>a</b>) PyMT-OVA cells grown as spheroids in 5% Matrigel in 24-well low-attachment plates and treated with indicated compounds for 48 h. Representative brightfield 20× images are shown. Scale bar = 150 µm. Enlarged insert showing small tumor spheroid. (<b>b</b>) PyMT-OVA cells grown as spheroids in 5% Matrigel in 96-well low-attachment plates. OT-1 CD8+ T-cells labeled with CellTracker Deep Red (red) and tumor cells labeled with DAPI (blue). Representative 10× fluorescent images are shown. Scale bar = 275 µm. For (<b>a</b>,<b>b</b>) PTX + T and Bleo + T compared to DMSO+T using a one-way ANOVA and Tukey’s post hoc analysis. For each treatment condition, T-cell versus no T-cell conditions were compared using an unpaired <span class="html-italic">t</span>-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, **** = <span class="html-italic">p</span> &lt; 0.0001. (<b>c</b>) CellTiterGlo viability assay of E0771-OVA cells after 48 h of treatment of PTX or (<b>d</b>) Bleo with or without CD8+ T-cells co-cultured at a 1:5 (T-cell:tumor cell) ratio. T-cell groups were compared to DMSO+T using a one-way ANOVA with Tukey’s post hoc analysis. For each treatment concentration, T-cell versus no T-cell condition was compared using an unpaired <span class="html-italic">t</span>-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, **** = <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
19 pages, 2115 KiB  
Article
Class B Scavenger Receptor CD36 as a Potential Therapeutic Target in Inflammation Induced by Danger-Associated Molecular Patterns
by Irina N. Baranova, Alexander V. Bocharov, Tatyana G. Vishnyakova, Zhigang Chen, Yunbo Ke, Anna A. Birukova, Peter S. T. Yuen, Takayuki Tsuji, Robert A. Star, Konstantin G. Birukov, Amy P. Patterson and Thomas L. Eggerman
Cells 2024, 13(23), 1992; https://doi.org/10.3390/cells13231992 - 3 Dec 2024
Viewed by 913
Abstract
The class B scavenger receptor CD36 is known to bind and mediate the transport of lipid-related ligands and it functions as a pattern recognition receptor (PRR) for a variety of pathogens, including bacteria and viruses. In this study, we assessed CD36’s role as [...] Read more.
The class B scavenger receptor CD36 is known to bind and mediate the transport of lipid-related ligands and it functions as a pattern recognition receptor (PRR) for a variety of pathogens, including bacteria and viruses. In this study, we assessed CD36’s role as a PRR mediating pro-inflammatory effects of several known Danger-Associated Molecular Patterns (DAMPs) used either as a single preparation or as a combination of DAMPs in the form of total cell/skeletal muscle tissue lysates. Our data demonstrated that multiple DAMPs, including HMGB1, HSPs, histone H3, SAA, and oxPAPC, as well as cell/tissue lysate preparations, induced substantially higher (~7–10-fold) IL-8 cytokine responses in HEK293 cells overexpressing CD36 compared to control WT cells. At the same time, DAMP-induced secretion of IL-6 in bone marrow-derived macrophages (BMDM) from CD36−/− mice was markedly (~2–3 times) reduced, as compared to macrophages from normal mice. Synthetic amphipathic helical peptides (SAHPs), known CD36 ligands, efficiently blocked CD36-dependent inflammatory responses induced by both cell and tissue lysates, HMGB1 and histone H3 in CD36+ cells. IP injection of total cellular lysate preparation induced inflammatory responses that were assessed by the expression of liver and lung pro-inflammatory markers, including IL-6, TNF-α, CD68, and CXCL1, and was reduced by ~50% in CD36-deficient mice compared to normal mice. Our findings demonstrate that CD36 is a PRR contributing to the innate immune response via mediating DAMP-induced inflammatory signaling and highlight the importance of this receptor as a potential therapeutic target in DAMP-associated inflammatory conditions. Full article
Show Figures

Figure 1

Figure 1
<p>Dose-dependent IL-8 secretion induced by HeLa cell and skeletal muscle CF in WT and CD36-overexpressing HEK293 cells. WT and CD36-overexpressing HEK293 cells were incubated with increasing concentrations of the CF preparations (see <a href="#sec2-cells-13-01992" class="html-sec">Section 2</a>) from HeLa cells, c-CF (<b>A</b>) or murine skeletal muscle, SM-CF (<b>B</b>) for 20 h. IL-8 levels were quantified in duplicate samples of cell culture supernatants by ELISA. Data represent one of three separate experiments that yielded similar results.</p>
Full article ">Figure 2
<p>Dose-dependent IL-8 secretion induced by the HMGB1 and LPS in WT and CD36-overexpressing HEK293 cells. Effect of heat treatment on the cytokine-inducing activity of HMGB1 and LPS. WT and CD36-overexpressing HEK293 cells were incubated with increasing concentrations of recombinant HMGB1 (<b>A</b>) or LPS (<b>B</b>) for 20 h. IL-8 levels were quantified in duplicate samples of cell culture supernatants by ELISA. Data represent one of three separate experiments that yielded similar results. The IL-8 levels were determined using duplicate samples of cell culture supernatants collected after a 20 h incubation of cells with either intact or heat-treated (100 °C for 45 min) HMGB1 (<b>C</b>) and LPS (<b>D</b>). The data presented are from one of two separate representative experiments.</p>
Full article ">Figure 3
<p>Effects of SAHPs on IL-8 secretion induced by the CF preparations and HMGB1 in CD36-overexpressing HEK293 cells. Cells were preincubated with or without increasing doses of L-37pA, ELK-B, or L3D-37pA for 1 h before a 20 h treatment with 1% c-CF (<b>A</b>) or 0.3% SM-CF (<b>B</b>). Cells were preincubated for 1 h with or without 10 µg/mL of L-37pA, ELK-B, or L3D-37pA before a 20 h treatment with increasing doses of HMGB1 (0.25, 1, and 5 µg/mL). IL-8 levels were determined in cell culture supernatants in duplicate (<b>C</b>). Data are from one of at least two representative experiments. * <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, and ns—nonsignificant, versus no peptide.</p>
Full article ">Figure 4
<p>Dose-dependent IL-8 secretion induced by the histone H3 in WT and CD36-overexpressing HEK293 cells. Effect of L-37pA on hH3-induced IL-8 secretion in CD36-HEK293 cells. WT and CD36-overexpressing HEK293 cells were incubated with increasing concentrations of histone H3 for 20 h. IL-8 levels were quantified in duplicate samples of cell culture supernatants by ELISA (<b>A</b>). Data represent one of three separate experiments that yielded similar results. CD36-HEK293 cells were preincubated for 1 h with 0, 10 µg/mL, and 25 µg/mL of L-37pA or L3D peptides before a 20 h treatment with 25 µg/mL of histone H3. IL-8 levels were determined in cell culture supernatants in duplicate (<b>B</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and ns—nonsignificant, versus histone H3 alone. Data are from one of at least two representative experiments.</p>
Full article ">Figure 5
<p>DAMPs-induced dose-dependent pro-inflammatory responses in WT and CD36-overexpressing HEK293 cells. WT and CD36-overexpressing HEK293 cells were incubated with increasing concentrations of HSP60 (<b>A</b>), HSP70 (<b>B</b>), oxPAPC (<b>C</b>), and SAA (<b>D</b>) for 20 h. IL-8 levels were quantified in duplicate samples of cell culture supernatants by ELISA. Data represent one of two separate experiments that yielded similar results.</p>
Full article ">Figure 6
<p>Pro-inflammatory responses induced by the various DAMPs in BMDM from WT and CD36-knockout mice. BMDM isolated from WT and CD36−/− mice were incubated with increasing doses of cCF (<b>A</b>), HMGB1 (<b>B</b>), histone H3 (<b>C</b>), and LPS (<b>D</b>) for 20 h. IL-6 levels were quantified in duplicate samples of cell culture supernatants by ELISA. Data represent one of two separate experiments that yielded similar results.</p>
Full article ">Figure 7
<p>Effects of signaling pathways Inhibitors on CF-induced IL-6 secretion in BMDM from WT and CD36-knockout mice. BMDM isolated from WT and CD36−/− were pre-incubated for 1 h with increasing doses of PD98059 (<b>A</b>), SP600125 (<b>B</b>), SB202190 (<b>C</b>), or PP2 (<b>D</b>). Following pre-incubation, cells were incubated with CF preparation for the next 20 h. IL-6 levels were quantified in duplicate samples of cell culture supernatants by ELISA. Data represent one of two separate experiments that yielded similar results.</p>
Full article ">Figure 8
<p>Hepatic gene expression of inflammatory markers in WT and CD36-knockout mice IP injected with a TCL preparation. Livers were collected for mRNA extraction and qRT-PCR assay as described in Materials and Methods. Expression levels of IL-6 (<b>A</b>), TNF-α (<b>B</b>), CD68 (<b>C</b>), and CCL2 (<b>D</b>) were normalized by GAPDH expression and are presented as the fold change relative to the PBS-treated WT control mice. Values shown are the means ± STD (n = 12–15 for the WT group, n = 20 for the CD36-KO group). * <span class="html-italic">p</span> &lt; 0.05, versus WT TCL-treated mice.</p>
Full article ">Figure 9
<p>Pulmonary gene expression of inflammatory markers in WT and CD36-knockout mice IP injected with a TCL preparation. Lungs were collected for mRNA extraction and qRT-PCR assay as described in Materials and Methods. Expression levels of IL-6 (<b>A</b>), TNF-α (<b>B</b>), and CXCL1 (<b>C</b>) were normalized by GAPDH expression and are presented as the fold change relative to PBS-treated WT control mice. Values shown are the means ± STD (n = 10 for the WT group, n = 14 for the CD36-KO group). * <span class="html-italic">p</span> &lt; 0.05, versus WT TCL-treated mice.</p>
Full article ">
12 pages, 9431 KiB  
Article
HOXA11-As Promotes Lymph Node Metastasis Through Regulation of IFNL and HMGB Family Genes in Pancreatic Cancer
by Hayato Nishiyama, Takeshi Niinuma, Hiroshi Kitajima, Kazuya Ishiguro, Eiichiro Yamamoto, Gota Sudo, Hajime Sasaki, Akira Yorozu, Hironori Aoki, Mutsumi Toyota, Masahiro Kai and Hiromu Suzuki
Int. J. Mol. Sci. 2024, 25(23), 12920; https://doi.org/10.3390/ijms252312920 - 30 Nov 2024
Viewed by 1035
Abstract
Recent studies have shown that long noncoding RNAs (lncRNAs) play pivotal roles in the development and progression of cancer. In the present study, we aimed to identify lncRNAs associated with lymph node metastasis in pancreatic ductal adenocarcinoma (PDAC). We analyzed data from The [...] Read more.
Recent studies have shown that long noncoding RNAs (lncRNAs) play pivotal roles in the development and progression of cancer. In the present study, we aimed to identify lncRNAs associated with lymph node metastasis in pancreatic ductal adenocarcinoma (PDAC). We analyzed data from The Cancer Genome Atlas (TCGA) database to screen for genes overexpressed in primary PDAC tumors with lymph node metastasis. Our screen revealed 740 genes potentially associated with lymph node metastasis, among which were multiple lncRNA genes located in the HOXA locus, including HOXA11-AS. Elevated expression of HOXA11-AS was associated with more advanced tumor stages and shorter overall survival in PDAC patients. HOXA11-AS knockdown suppressed proliferation and migration of PDAC cells. RNA-sequencing analysis revealed that HOXA11-AS knockdown upregulated interferon lambda (IFNL) family genes and downregulated high-mobility group box (HMGB) family genes in PDAC cells. Moreover, HMGB3 knockdown suppressed proliferation and migration by PDAC cells. These results suggest that HOXA11-AS contributes to PDAC progression, at least in part, through regulation of IFNL and HMGB family genes and that HOXA11 AS is a potential therapeutic target in PDAC. Full article
(This article belongs to the Section Molecular Oncology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Identification of HOXA11-AS upregulation in PDAC with lymph node metastasis. (<b>A</b>) Heatmap showing expression of 740 genes upregulated in PDAC with lymph node metastasis in TCGA dataset. (<b>B</b>) GO analysis of genes upregulated in PDAC with lymph node metastasis. (<b>C</b>) Circular plot showing the chromosome positions of upregulated genes in PDAC with lymph node metastasis. (<b>D</b>) Heatmap showing expression of HOXA cluster genes upregulated in PDAC with lymph node metastasis. (<b>E</b>) Heatmap showing expression of HOXA cluster genes in PDAC cell lines in the CCLE dataset. (<b>F</b>) qRT-PCR analysis of HOXA11-AS in normal pancreatic tissue and PDAC cell lines (<span class="html-italic">n</span> = 3). Error bars represent SDs.</p>
Full article ">Figure 2
<p>Association between HOXA11-AS expression and clinical and molecular features in primary PDAC. (<b>A</b>) Levels of HOXA11-AS expression in primary PDAC tumors with the indicated N factors (left) or T factors (right) in TCGA dataset. (<b>B</b>) Kaplan–Meier curves showing the effect of HOXA11-AS expression on survival of PDAC patients (<span class="html-italic">n</span> = 176). (<b>C</b>) HOXA11-AS expression in normal pancreatic tissue (<span class="html-italic">n</span> = 5) and primary PDAC tumors (<span class="html-italic">n</span> = 42) in the GSE55643 dataset. (<b>D</b>) Summarized results of GSEA of the indicated gene sets using genes upregulated in PDAC with high HOXA11-AS expression in TCGA dataset. NES, normalized enrichment score; FDR, false discovery rate. (<b>E</b>) Results of GSEA of the indicated gene sets in PDAC with high HOXA11-AS expression. (<b>F</b>) Summarized results of GO analysis using genes upregulated in PDAC with high HOXA11-AS expression in TCGA dataset. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Functional analysis of HOXA11-AS in PDAC cells. (<b>A</b>) qRT-PCR analysis of HOXA11-AS in PDAC cells transfected with a control siRNA or siRNAs targeting HOXA11-AS (<span class="html-italic">n</span> = 3). (<b>B</b>) Cell viability assays with PDAC cells transfected with the indicated siRNAs (<span class="html-italic">n</span> = 6). (<b>C</b>,<b>D</b>) Cell migration (<b>C</b>) and invasion (<b>D</b>) assays with PDAC cells transfected with the indicated siRNAs. Representative results are shown on the left, summarized results on the right (<span class="html-italic">n</span> = 3). Error bars represent SDs. * <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 4
<p>HOXA11-AS regulates IFNL and HMGB family genes in PDAC cells. (<b>A</b>) Results of RNA-seq in KP1-NL cells transfected with the indicated siRNAs. Shown is a heatmap of genes whose expression was altered (&gt;1.5-fold) by HOXA11-AS knockdown. Representative genes are indicated on the right. (<b>B</b>) Summarized results of GSEA of the indicated gene sets using genes upregulated by HOXA11 AS knockdown. (<b>C</b>) GO analysis of genes upregulated by HOXA11-AS knockdown. (<b>D</b>–<b>F</b>) qRT-PCR analysis of IFNL1 (<b>D</b>), IFNL2 (<b>E</b>) and HMGB3 (<b>F</b>) in the indicated PDAC cell lines transfected with the indicated siRNAs (<span class="html-italic">n</span> = 3). Error bars represent SDs. * <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>Functional analysis of HMGB3 in PDAC cells. (<b>A</b>) qRT-PCR analysis of HMGB3 in KP1-NL cells transfected with a control siRNA or siRNA targeting HMGB3 (<span class="html-italic">n</span> = 3). (<b>B</b>) Cell viability assays with KP1-NL cells transfected with the indicated siRNAs (<span class="html-italic">n</span> = 6). (<b>C</b>) Cell migration assays with KP1-NL cells transfected with the indicated siRNAs. Representative results are shown on the left, summarized results on the right (<span class="html-italic">n</span> = 3). (<b>D</b>) Summarized results of GSEA of indicated gene sets using genes upregulated in PDAC with high HMGB3 expression in TCGA dataset. (<b>E</b>) Results of GSEA of the indicated gene sets in PDAC with high HMGB3 expression. Error bars represent SDs. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
27 pages, 2401 KiB  
Review
High Mobility Group Box 1 (HMGB1): Molecular Signaling and Potential Therapeutic Strategies
by Sayantap Datta, Mohammad Atiqur Rahman, Saisudha Koka and Krishna M. Boini
Cells 2024, 13(23), 1946; https://doi.org/10.3390/cells13231946 - 23 Nov 2024
Cited by 1 | Viewed by 1688
Abstract
High Mobility Group Box 1 (HMGB1) is a highly conserved non-histone chromatin-associated protein across species, primarily recognized for its regulatory impact on vital cellular processes, like autophagy, cell survival, and apoptosis. HMGB1 exhibits dual functionality based on its localization: both as a non-histone [...] Read more.
High Mobility Group Box 1 (HMGB1) is a highly conserved non-histone chromatin-associated protein across species, primarily recognized for its regulatory impact on vital cellular processes, like autophagy, cell survival, and apoptosis. HMGB1 exhibits dual functionality based on its localization: both as a non-histone protein in the nucleus and as an inducer of inflammatory cytokines upon extracellular release. Pathophysiological insights reveal that HMGB1 plays a significant role in the onset and progression of a vast array of diseases, viz., atherosclerosis, kidney damage, cancer, and neurodegeneration. However, a clear mechanistic understanding of HMGB1 release, translocation, and associated signaling cascades in mediating such physiological dysfunctions remains obscure. This review presents a detailed outline of HMGB1 structure–function relationship and its regulatory role in disease onset and progression from a signaling perspective. This review also presents an insight into the status of HMGB1 druggability, potential limitations in understanding HMGB1 pathophysiology, and future perspective of studies that can be undertaken to address the existing scientific gap. Based on existing paradigm of various studies, HMGB1 is a critical regulator of inflammatory cascades and drives the onset and progression of a broad spectrum of dysfunctions. Studies focusing on HMGB1 druggability have enabled the development of biologics with potential clinical benefits. However, deeper understanding of post-translational modifications, redox states, translocation mechanisms, and mitochondrial interactions can potentially enable the development of better courses of therapy against HMGB1-mediated physiological dysfunctions. Full article
Show Figures

Figure 1

Figure 1
<p>Molecular structure and functional correlation of HMGB1 domains. The Box-A chiefly exhibits anti-HMGB1 effects through specific intradomain regions, regulating heparin binding and proteolytic cleavage. The Box-B chiefly mediates pro-inflammatory functions. The acidic C-terminal regulates DNA-bending capabilities, chromosomal derotation, and the interactive potential of HMGB1 with core and linker histones [<a href="#B1-cells-13-01946" class="html-bibr">1</a>,<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B52-cells-13-01946" class="html-bibr">52</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>].</p>
Full article ">Figure 2
<p>Schematic representation of HMGB1-induced signaling cascades culminating to atherosclerosis. Extracellularly released HMGB1 augments expression of cytokines (TNF-α), cell adhesion molecules (ICAM-1 and VCAM-1), and other signaling receptors (RAGE) to induce TNF-α pro-inflammatory signaling, monocyte, macrophage aggregation, NF-κB signaling. HMGB1-induced inflammation and concomitant decrease in anti-coagulant proteins like thrombomodulin lead to atherosclerotic plaque formation [<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>].</p>
Full article ">Figure 3
<p>HMGB1-associated NF-κB signaling activation, G1 cell cycle arrest, and the augmentation of EMT (via RAGE signaling) culminates to kidney damage, attributing to subsequent renal dysfunctions [<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>].</p>
Full article ">Figure 4
<p>HMGB1 binds to α-synuclein aggregates in Lewy bodies, inhibits microglial phagocytosis, and upregulates NADPH oxidase levels (chiefly via NF-κB signaling) to mediate neurodegeneration [<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>].</p>
Full article ">Figure 5
<p>Polyclonal- and monoclonal-antibody-mediated HMGB1 targeting attenuates the onset and progression of varied dysfunctions, viz., arthritis, drug-induced pulmonary fibrosis, hepatic injury, and BBB defects [<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>].</p>
Full article ">Figure 6
<p>Synthetically derived SMIs, viz., nafamostat mesylate, gabexate mesylate, and silvestat prevent extracellular HMGB1 release, downregulate NF-κB and TNF-α pro-inflammatory signaling, and attenuate vascular inflammation and atherosclerosis progression [<a href="#B51-cells-13-01946" class="html-bibr">51</a>,<a href="#B53-cells-13-01946" class="html-bibr">53</a>,<a href="#B225-cells-13-01946" class="html-bibr">225</a>].</p>
Full article ">
19 pages, 3389 KiB  
Article
Anti-Inflammatory Effects of Extracellular Vesicles from Ecklonia cava on 12-O-Tetradecanoylphorbol-13-Acetate-Induced Skin Inflammation in Mice
by Geebum Kim, So Young Lee, Seyeon Oh, Jong-Won Jang, Jehyuk Lee, Hyun-Seok Kim, Kuk Hui Son and Kyunghee Byun
Int. J. Mol. Sci. 2024, 25(23), 12522; https://doi.org/10.3390/ijms252312522 - 21 Nov 2024
Viewed by 799
Abstract
Steroids, which are often used to treat the inflammation associated with various skin diseases, have several negative side effects. As Ecklonia cava extract has anti-inflammatory effects in various diseases, we evaluated the efficacy of Ecklonia cava-derived extracellular vesicles (EVEs) in decreasing 12-O-tetradecanoylphorbol-13-acetate [...] Read more.
Steroids, which are often used to treat the inflammation associated with various skin diseases, have several negative side effects. As Ecklonia cava extract has anti-inflammatory effects in various diseases, we evaluated the efficacy of Ecklonia cava-derived extracellular vesicles (EVEs) in decreasing 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation. We determined the effect of the EVEs on the TLR4/NF-κB/NLRP3 inflammasome in human keratinocytes and mouse ear skin. TPA-treated human keratinocytes showed an increased expression of TLR4 and its ligands HMGB1 and S100A8. TPA also increased the expression of (1) NF-κB; (2) the NLRP3 inflammasome components NLRP3, ASC, and caspase 1; and (3) the pyroptosis-related factors GSDMD-NT, IL-18, and IL-1β. However, the expression of these molecules decreased in the TPA-treated human keratinocytes after EVE treatment. Similar to the in vitro results, TPA increased the expression of these molecules in mouse ear skin, and EVE treatment decreased their expression. The TPA treatment of skin increased edema, redness, neutrophil infiltration, and epidermal thickness, and EVE reduced these symptoms of inflammation. In conclusion, the EVEs decreased TPA-induced skin inflammation, which was associated with a decrease in the TLR4/NF-κB/NLRP3 inflammasome. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Regulation of HMGB1, S100A8, and TLR4 by EVEs in TPA-treated human keratinocytes. (<b>A</b>) Western blot detection of HMGB1 and S100A8 expressions in TPA-treated human keratinocytes subjected to EVEs or DXA. (<b>B</b>,<b>C</b>) Quantification analysis of (<b>B</b>) HMGB1 and (<b>C</b>) S100A8 with (<b>A</b>) Western blot images using Image J software version 1.53. (<b>D</b>,<b>E</b>) TLR4 protein expression in TPA-treated human keratinocytes subjected to EVEs or DXA after TLR4 silencing. β-actin was determined as loading control. Human keratinocytes were treated with 100 nM TPA for 4 h, followed by 48 h incubation with PBS, EVEs (0.05 mg/mL), or DXA (0.001 mM). TLR4 knockdown was achieved by transfecting 500 ng of TLR4 shRNA plasmid for 24 h prior to treatment. Data are presented as mean ± SD of three independent experiments. <span class="html-italic">p</span> &lt; 0.05; a–d; same letters indicate nonsignificant differences between groups, as determined by multiple comparisons (Mann–Whitney U test). DXA, dexamethasone; EVE, extracellular vesicle from <span class="html-italic">E. cava</span>; HMGB1, high mobility-group box-1 protein; ICC, immunocytochemistry; PBS, phosphate-buffered saline; SD, standard deviation; TLR4, Toll-like receptor 4; TPA, 12-O-tetradecanoylphorbol-13-acetate.</p>
Full article ">Figure 2
<p>Regulation of NF-κB, NLRP3 inflammasome, and pyroptosis by EVEs in TPA-treated human keratinocytes. (<b>A</b>) Representative images of NF-κB (green) immunofluorescence staining in TPA-treated human keratinocytes subjected to EVEs or DXA after TLR4 silencing. Nuclei were stained with DAPI (blue). Scale bar = 50 μm. (<b>B</b>) Western blot detection of NLRP3, ASC, pro-caspase 1, and cleaved-caspase expressions in TPA-treated human keratinocytes subjected to EVEs or DXA after TLR4 silencing. (<b>C</b>) Western blot detection of GSDMD and GSDMD-NT expressions in TPA-treated human keratinocytes subjected to EVEs or DXA after TLR4 silencing. β-actin was determined as loading control. (<b>D</b>,<b>E</b>) Quantification analysis of IL-18 and IL-1β secretions in TPA-treated human keratinocytes subjected to EVEs or DXA after TLR4 silencing using ELISA. Human keratinocytes were treated with 100 nM TPA for 4 h, followed by 48 h incubation with PBS, EVEs (0.05 mg/mL), or DXA (0.001 mM). TLR4 knockdown was performed by transfecting 500 ng of TLR4 shRNA plasmid for 24 h before treatment. Data are presented as mean ± SD of three independent experiments. <span class="html-italic">p</span> &lt; 0.05; a–f; same letters indicate nonsignificant differences between groups, as determined by multiple comparisons (Mann–Whitney U test). ASC, apoptosis-associated speck-like protein; DXA, dexamethasone; ELISA, enzyme-linked immunosorbent assay; EVE, extracellular vesicle from <span class="html-italic">E. cava</span>; GSDMD, gasdermin D; GSDMD-NT, gasdermin D N-terminal domain; IL, interleukin; NLRP3, NOD-like receptor protein 3; SD, standard deviation; TPA, 12-O-tetradecanoylphorbol-13-acetate.</p>
Full article ">Figure 3
<p>Regulation of HMGB1, S100A8, TLR4, and NF-κB by EVEs in TPA-treated mouse ears. (<b>A</b>) Western blot detection of HMGB1, S100A8, and TLR4 expressions in TPA-treated mouse ears subjected to EVEs or DXA. β-actin was determined as loading control. (<b>B</b>–<b>E</b>) Quantification analysis of (<b>B</b>) HMGB1, (<b>C</b>) S100A8, and (<b>D</b>) TLR4 with (<b>A</b>) Western blot images using Image J software. (<b>F</b>) Representative images of NF-κB immunohistochemistry staining in TPA-treated mouse ears subjected to EVEs or DXA. Nuclei were stained with hematoxylin (blue). Red arrows indicate positive signals. Scale bar = 100 μm. For each mouse, 50 μM TPA was applied topically to one ear five times at 3-day intervals over 15 days, followed by weekly application of EVEs (0.5, 1.0, or 2.0 mg/mL) or DXA (0.4 mg/kg). Data are presented as mean ± SD of three independent experiments. <span class="html-italic">p</span> &lt; 0.05; a–e; same letters indicate nonsignificant differences between groups, as determined by multiple comparisons (Mann–Whitney U test). DW, distilled water; DXA, dexamethasone; EVE, extracellular vesicle from <span class="html-italic">E. cava</span>; HMGB1, high mobility group box-1 protein; IHC, immunohistochemistry; NF-κB, nuclear factor-κB; SD, standard deviation; TLR4, Toll-like receptor 4; TPA, 12-O-tetradecanoylphorbol-13-acetate.</p>
Full article ">Figure 4
<p>Regulation of NLRP3 inflammasome and pyroptosis by EVEs in TPA-treated mouse ears. (<b>A</b>) Western blot detection of NLRP3, ASC, pro-caspase 1, and cleaved-caspase expressions in TPA-treated mouse ears subjected to EVEs or DXA. (<b>B</b>–<b>E</b>) Quantification analysis of (<b>B</b>) NLRP3, (<b>C</b>) ASC, (<b>D</b>) pro-caspase 1, and (<b>E</b>) cleaved-caspase 1 with (<b>A</b>) Western blot images using Image J software. (<b>F</b>) Western blot detection of GSDMD and GSDMD-NT expressions in TPA-treated mouse ears subjected to EVEs or DXA. β-actin was determined as loading control. (<b>G</b>) Quantification analysis of GSDMD-NT with (<b>F</b>) Western blot images using Image J software. (<b>H</b>,<b>I</b>) Quantification analysis of IL-18 and IL-1β in TPA-treated mouse ears subjected to EVEs or DXA using ELISA. For each mouse, 50 μM TPA was applied topically to one ear five times at 3-day intervals over 15 days, with weekly applications of EVEs (0.5, 1.0, or 2.0 mg/mL) or DXA (0.4 mg/kg). Data are presented as mean ± SD of three independent experiments. <span class="html-italic">p</span> &lt; 0.05; a–e; same letters indicate nonsignificant differences between groups, as determined by multiple comparisons (Mann–Whitney U test). ASC, apoptosis-associated speck-like protein; DW, distilled water; DXA, dexamethasone; ELISA, enzyme-linked immunosorbent assay; EVE, extracellular vesicle from <span class="html-italic">E. cava</span>; GSDMD, gasdermin D; GSDMD-NT, gasdermin D N-terminal domain; IL, interleukin; NLRP3, NOD-like receptor protein 3; SD, standard deviation; TPA, 12-O-tetradecanoylphorbol-13-acetate.</p>
Full article ">Figure 5
<p>Effect of EVEs on TPA-induced inflammation in TPA-treated mouse ears. (<b>A</b>) Representative images of TPA-treated mouse ears subjected to EVEs or DXA at 15 days. (<b>B</b>,<b>C</b>) (<b>B</b>) Ear redness and (<b>C</b>) thickness of TPA-treated mouse ears subjected to EVEs or DXA. (<b>D</b>) Representative images of hematoxylin and eosin staining in TPA-treated epidermis (blue box) and dermis (green box) of mouse ears subjected to EVEs or DXA. (<b>E</b>,<b>F</b>) (<b>E</b>) Number of neutrophils and (<b>F</b>) thickness of epidermis in TPA-treated mouse ears subjected to EVEs or DXA. Red circles indicate positive neutrophil infiltration. TPA (50 μM) was applied topically to each ear five times at 3-day intervals over 15 days. EVEs (0.5, 1.0, or 2.0 mg/mL) or DXA (0.4 mg/kg) were applied weekly. Data are presented as mean ± SD of three independent experiments. <span class="html-italic">p</span> &lt; 0.05; a–e; same letters indicate nonsignificant differences between groups, as determined by multiple comparisons (Mann–Whitney U test). DW, distilled water; DXA, dexamethasone; EVE, extracellular vesicle from <span class="html-italic">E. cava</span>; SD, standard deviation; TPA, 12-O-tetradecanoylphorbol-13-acetate.</p>
Full article ">
16 pages, 2746 KiB  
Article
Novel Inhibitory Actions of Neuroactive Steroid [3α,5α]-3-Hydroxypregnan-20-One on Toll-like Receptor 4-Dependent Neuroimmune Signaling
by Alejandro G. Lopez, Venkat R. Chirasani, Irina Balan, Todd K. O’Buckley, Makayla R. Adelman and A. Leslie Morrow
Biomolecules 2024, 14(11), 1441; https://doi.org/10.3390/biom14111441 - 13 Nov 2024
Viewed by 1169
Abstract
The endogenous neurosteroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP) modulates inflammatory and neuroinflammatory signaling through toll-like receptors (TLRs) in human and mouse macrophages, human blood cells and alcohol-preferring (P) rat brains. Although it is recognized that 3α,5α-THP inhibits TLR4 activation by blocking interactions with MD2 and MyD88, [...] Read more.
The endogenous neurosteroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP) modulates inflammatory and neuroinflammatory signaling through toll-like receptors (TLRs) in human and mouse macrophages, human blood cells and alcohol-preferring (P) rat brains. Although it is recognized that 3α,5α-THP inhibits TLR4 activation by blocking interactions with MD2 and MyD88, the comprehensive molecular mechanisms remain to be elucidated. This study explores additional TLR4 activation sites, including TIRAP binding to MyD88, which is pivotal for MyD88 myddosome formation, as well as LPS interactions with the TLR4:MD2 complex. Both male and female P rats (n = 8/group) received intraperitoneal administration of 3α,5α-THP (15 mg/kg; 30 min) or a vehicle control, and their hippocampi were analyzed using immunoprecipitation and immunoblotting techniques. 3α,5α-THP significantly reduces the levels of inflammatory mediators IL-1β and HMGB1, confirming its anti-inflammatory actions. We found that MyD88 binds to TLR4, IRAK4, IRAK1, and TIRAP. Notably, 3α,5α-THP significantly reduces MyD88-TIRAP binding (Males: −31 ± 9%, t-test, p < 0.005; Females: −53 ± 15%, t-test, p < 0.005), without altering MyD88 interactions with IRAK4 or IRAK1, or the baseline expression of these proteins. Additionally, molecular docking and molecular dynamic analysis revealed 3α,5α-THP binding sites on the TLR4:MD2 complex, targeting a hydrophobic pocket of MD2 usually occupied by Lipid A of LPS. Surface plasmon resonance (SPR) assays validated that 3α,5α-THP disrupts MD2 binding of Lipid A (Kd = 4.36 ± 5.7 μM) with an inhibition constant (Ki) of 4.5 ± 1.65 nM. These findings indicate that 3α,5α-THP inhibition of inflammatory mediator production involves blocking critical protein-lipid and protein-protein interactions at key sites of TLR4 activation, shedding light on its mechanisms of action and underscoring its therapeutic potential against TLR4-driven inflammation. Full article
(This article belongs to the Special Issue Role of Neuroactive Steroids in Health and Disease: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>3α,5α-THP reduces HMGB1 and IL-1β protein levels in P rat hippocampus. Male and female alcohol-preferring (P) rats (n = 8/group) were treated with 3α,5α-THP (15 mg/kg; 30 min) or the vehicle (45% <span class="html-italic">w</span>/<span class="html-italic">v</span> 2-hydroxypropyl-β-cyclodextrin; 30 min) and hippocampus lysate was examined for IL-1β and HMGB1 protein expression. Data were collected from two separate P rat cohorts and protein expression was analyzed via immunoblotting with data expressed as the percent change from the control (vehicle) in each sex. (<b>A</b>) IL-1β expression was decreased in both male P rats by 46.95% ± 11.37 (2-way ANOVA: F(1,69) = 23, <span class="html-italic">p</span> &lt; 0.0001 ****, n = 18/vehicle and n = 18/3α,5α-THP) and female P rats by 24.68% ± 9.23 (2-way ANOVA: F(1,69) = 23, <span class="html-italic">p</span> &lt; 0.0001 ****, n = 18/vehicle and n = 18/3α,5α-THP). (<b>B</b>) The expression of hippocampal HMGB1 was also tested in male and female P rats. HMGB1 was decreased in males by 38.60% ± 10.12 (2-way ANOVA: F(1,66) = 25.85, <span class="html-italic">p</span> &lt; 0.0001 ****, n = 18/vehicle and n = 18/3α,5α-THP) and 42.36% ± 12.58 (2-way ANOVA: F(1,66) = 25.85, <span class="html-italic">p</span> &lt; 0.0001 ****, n = 18/vehicle and n = 17/3α,5α-THP) in females. Western blot original images are in the <a href="#app1-biomolecules-14-01441" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 2
<p>3α,5α-THP inhibits TIRAP binding to MyD88 in P rat male (<b>A</b>) and female (<b>B</b>) hippocampi. MyD88 immunoprecipitation of various components of the myddosome complex was conducted in male and female, vehicle- (45% <span class="html-italic">w</span>/<span class="html-italic">v</span> 2-hydroxypropyl-β-cyclodextrin; 30 min) and 3α,5α-THP-treated (15 mg/kg; 30 min) P rats (<b>A</b>,<b>B</b>). Densiometric comparison of the effect of 3α,5α-THP on MyD88 immunoprecipitation of TIRAP (<b>A</b>,<b>B</b>), IRAK4 (<b>C</b>,<b>D</b>) and IRAK1 (<b>E</b>,<b>F</b>) in female and male hippocampi. Immunoblots and densiometric measurements of extracted hippocampal TIRAP (<b>G</b>) and MyD88 (<b>H</b>) in vehicle- and 3α,5α-THP-treated animals. Western blot original images are in the <a href="#app1-biomolecules-14-01441" class="html-app">Supplementary Materials</a>. *** <span class="html-italic">p</span> &lt; 0.005, **** <span class="html-italic">p</span> &lt; 0.001, ns: no significant difference.</p>
Full article ">Figure 3
<p>3α,5α-THP has no effect on TLR4 binding to MD-2 in the hippocampus of male and female P rats. (<b>A</b>,<b>B</b>) Hippocampus whole lysate was immunoblotted for the presence of MD-2 in vehicle- (45% <span class="html-italic">w</span>/<span class="html-italic">v</span> 2-hydroxypropyl-β-cyclodextrin; 30 min) and 3α,5α-THP-treated (15 mg/kg; 30 min) P rats and densiometric comparison of the effect of 3α,5α-THP on MD-2 expression. (<b>C</b>,<b>E</b>) Immunoprecipitation of TLR4/MD-2 and densiometric comparison of MD2 between vehicle and 3α,5α-THP-treated P rats (male: <span class="html-italic">t</span>-test, <span class="html-italic">p</span> = 0.56, n = 4/group female: (<span class="html-italic">t</span>-test, <span class="html-italic">p</span> = 0.46, n = 4/group) (<b>D</b>,<b>F</b>). Western blot original images are in the <a href="#app1-biomolecules-14-01441" class="html-app">Supplementary Materials</a>. ns: no significant difference.</p>
Full article ">Figure 4
<p>3α,5α-THP docks TLR4-bound MD-2 and inhibits Lipid A binding to MD-2 in SPR studies. (<b>A</b>) Molecular docking shows that 3α,5α-THP favors binding to MD-2, with multiple docking poses of 3α,5α-THP found within the MD-2 binding pocket. (<b>B</b>) The top pose of 3α,5α-THP in MD-2 shows multiple π-alkyl bonds formed with key MD-2 residues. (<b>C</b>) Two-dimensional representation of 3α,5α-THP binding MD-2, depicting the amino acid interactions. (<b>D</b>) SPR studies show Lipid A binds immobilized MD-2 with a K<sub>D</sub> = 4.3 ± 0.5 µM (k<sub>a</sub> = 1.5 × 10<sup>2</sup> M<sup>−1</sup>S<sup>−1</sup>, k<sub>d</sub> = 6.78 × 10<sup>−4</sup> S<sup>−1</sup>). (<b>E</b>) SPR competition assay showing that 3α,5α-THP competitively binds MD2, as increasing concentrations of 3α,5α-THP decrease Lipid A binding (<b>D</b>).</p>
Full article ">Figure 5
<p>Structural and interaction analysis of MD-2 with 3α,5α-THP binding. Panels a–c present the key findings from MD simulations and trajectory analysis of the MD-2 complexed with the neuroactive steroid (3α,5α)-3-hydroxypregnan-20-one (3α,5α-THP), performed using GROMACS with the CHARMM36 force field. (<b>A</b>) The root-mean-square deviation (RMSD) of the backbone atoms of MD-2 relative to its initial structure is plotted over the 1000 ns simulation trajectory for both the 3α,5α-THP-bound (red) and apo (black) states. (<b>B</b>) The root-mean-square fluctuation (RMSF) of individual Cα atoms of MD-2 is plotted over the 1000 ns simulation trajectory for both the 3α,5α-THP-bound (red) and apo (black) states. (<b>C</b>) The average number of non-bonded contacts between 3α,5α-THP and MD-2, calculated across the simulation. To calculate the non-bonded contacts between 3α,5α-THP and MD-2, the gmx mindist tool was used. A cutoff distance of 5 Å was applied, which defines contacts as any non-bonded interactions where the distance between atoms of 3α,5α-THP and MD-2 is 5 Å or less. On average, approximately 1200 contacts were observed between 3α,5α-THP and MD-2, indicative of stable interactions within the binding pocket.</p>
Full article ">
Back to TopTop