[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

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
remove_circle_outline

Search Results (1,058)

Search Parameters:
Keywords = NLRP3 inflammasome

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 3235 KiB  
Article
Dysregulation of NLRP3 Inflammasome and Promotion of Disease by IL-1β in a Murine Model of Sandhoff Disease
by Nick Platt, Dawn Shepherd, David A. Smith, Claire Smith, Kerri-Lee Wallom, Raashid Luqmani, Grant C. Churchill, Antony Galione and Frances M. Platt
Cells 2025, 14(1), 35; https://doi.org/10.3390/cells14010035 (registering DOI) - 1 Jan 2025
Viewed by 62
Abstract
Sandhoff disease (SD) is a progressive neurodegenerative lysosomal storage disorder characterized by GM2 ganglioside accumulation as a result of mutations in the HEXB gene, which encodes the β-subunit of the enzyme β-hexosaminidase. Lysosomal storage of GM2 triggers inflammation in the CNS and periphery. [...] Read more.
Sandhoff disease (SD) is a progressive neurodegenerative lysosomal storage disorder characterized by GM2 ganglioside accumulation as a result of mutations in the HEXB gene, which encodes the β-subunit of the enzyme β-hexosaminidase. Lysosomal storage of GM2 triggers inflammation in the CNS and periphery. The NLRP3 inflammasome is an important coordinator of pro-inflammatory responses, and we have investigated its regulation in murine SD. The NLRP3 inflammasome requires two signals, lipopolysaccharide (LPS) and ATP, to prime and activate the complex, respectively, leading to IL-1β secretion. Peritoneal, but not bone-marrow-derived, macrophages from symptomatic SD mice, but not those from pre-symptomatic animals, secrete the cytokine following priming with LPS with no requirement for activation with ATP, suggesting that such NLRP3 deregulation is related to the extent of glycosphingolipid storage. Dysregulated production of IL-1β was dependent upon caspase activity but not cathepsin B. We investigated the role of IL-1β in SD pathology using two approaches: the creation of hexb−/−Il1r1−/− double knockout mice or by treating hexb−/− animals with anakinra, a recombinant form of the IL-1 receptor antagonist, IL-1Ra. Both resulted in modest but significant extensions in lifespan and improvement of neurological function. These data demonstrate that IL-1β actively participates in the disease process and provides proof-of-principle that blockade of the pro-inflammatory cytokine IL-1β may provide benefits in patients. Full article
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">hexb</span><sup>−/−</sup> resident peritoneal macrophages (RPMϕ) isolated from 14-week-old mice display significantly greater LysoTracker<sup>TM</sup> staining intensity in comparison with age-matched <span class="html-italic">hexb</span><sup>+/+</sup> RPMϕ. Panel (<b>a</b>), cartoon of the 2-signal regulation of IL-1β production by NLRP3 inflammasome. Priming and activation steps are highlighted in yellow. (<b>b</b>) Histogram of relative intensity of LysoTracker<sup>TM</sup> staining of <span class="html-italic">hexb</span><sup>+/+</sup> RPMϕ (open columns) and <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ cyan-filled columns). Values, mean ± SEM, n = 5. Statistical analysis, Student’s <span class="html-italic">t</span>-test. ** <span class="html-italic">p</span> &lt; 0.0029. Data are representative of a minimum of 3 independent experiments.</p>
Full article ">Figure 2
<p><span class="html-italic">hexb</span><sup>−/−</sup> resident peritoneal macrophages (RPMϕ) but not <span class="html-italic">hexb</span><sup>+/+</sup> RPMϕ isolated from 14-week-old mice secrete significant quantities of IL-1β and caspase-1 in response to priming of NLRP3 inflammasome. (<b>a</b>) Upper panel: Western blot of culture supernatants of <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ primed with either LPS or PGN or primed and activated with ATP and probed with anti-IL-1β antisera. Lower panel: Western blot of cell lysates of <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ primed with either LPS or PGN or primed and activated with ATP and probed with anti-β-actin antisera. (<b>b</b>) Upper panel: Western blot of culture supernatants of <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ primed with either LPS or PGN or primed and activated with ATP and probed with anti-caspase-1 antisera. Lower panel. Western blot of cell lysates of <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ primed with either LPS or PGN or primed and activated with ATP and probed with anti-β-actin antisera. Migration of ProIL-1β and mature IL-1β or Procaspase-1 and caspase-1 are indicated with arrows. Migration of molecular weight markers as indicated. Data are representative of two independent experiments.</p>
Full article ">Figure 3
<p><span class="html-italic">hexb</span><sup>−/−</sup> resident peritoneal macrophages (RPMϕ) from 14-week-old mice but not <span class="html-italic">hexb</span><sup>+/+</sup> RPMϕ secrete significant levels of IL-1β following LPS priming. Histogram of cytokine concentrations (pg/mL) determined by specific ELISA in culture supernatants of <span class="html-italic">hexb</span><sup>−/−</sup> and <span class="html-italic">hexb</span><sup>+/+</sup> RPMϕ either untreated (unfilled circles), primed with LPS (cyan filled columns), primed with LPS and activated with ATP (red filled columns) or activated only (unfilled diamonds). Data are mean ± SEM, n = 8. Statistical analysis, One-way ANOVA. **** <span class="html-italic">p</span> &lt; 0.0001 or *** <span class="html-italic">p</span> = 0.0056. Results are representative of 2 independent experiments.</p>
Full article ">Figure 4
<p>BMMϕ derived from 14-week-old <span class="html-italic">hexb</span><sup>−/−</sup> mice do not display enhanced LysoTracker<sup>TM</sup> staining or aberrant production of IL-1β. (<b>a</b>) Histogram of relative intensity of LysoTracker<sup>TM</sup> staining of <span class="html-italic">hexb</span><sup>+/+</sup> BMMϕ (open columns) and <span class="html-italic">hexb</span><sup>−/−</sup> BMMϕ (magenta-filled columns). Values, mean ± SEM, n = 7. Statistical analysis, Student’s <span class="html-italic">t</span>-test. ns, not significant. Data are representative of a minimum of 3 independent experiments. (<b>b</b>) Histogram of ELISA measurements of IL-1β concentrations in culture supernatants of BMMϕ derived from 14-week-old <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> mice, either untreated (open columns), LPS treated (cyan filled columns) or LPS + ATP (red filled columns). Values, mean ± SEM, n = 4. Statistical analysis, one-way ANOVA. **** <span class="html-italic">p</span> &lt; 0.0001, ns, not significant. Data are representative of a minimum of 3 independent experiments.</p>
Full article ">Figure 5
<p><span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ isolated from 12-week-old and 14-week-old symptomatic but not 8-week-old non-symptomatic mice display aberrant IL-1β production after priming. Histograms of ELISA measurements of IL-1β concentrations in supernatants of RPMϕ isolated from 8-week-old (<b>a</b>), 12-week-old (<b>b</b>), and 14-week-old (<b>c</b>) <span class="html-italic">hexb</span><sup>+/+</sup> and <span class="html-italic">hexb</span><sup>−/−</sup> mice either untreated (open columns), primed with LPS (blue columns), or primed with LPS and activated with ATP (red columns). Data, mean ± SEM. n = 5 replicates for each sample. Statistical analysis, Student’s <span class="html-italic">t</span>-test **** <span class="html-italic">p</span> &lt; 0.0001, ** <span class="html-italic">p</span> &lt; 0.0.01, * <span class="html-italic">p</span> &lt; 0.05. ns, not significant. Data are representative of three independent experiments.</p>
Full article ">Figure 6
<p>Inhibition of caspase-1 but not cathepsin B activity significantly reduces IL-1β production by LPS-primed <span class="html-italic">hexb</span><sup>−/−</sup> RPMϕ isolated from 14-week-old mice. Histogram of ELISA determinations of IL-1β concentrations in supernatants of RPMϕ from 14-week-old <span class="html-italic">hexb</span><sup>+/+</sup> (open columns) or <span class="html-italic">hexb</span><sup>−/−</sup> (magenta columns) mice either untreated, primed with LPS, primed with LPS and activated with ATP, or primed with LPS and incubated with monosodium urate crystals (MSU) in the absence or presence of the caspase-1 inhibitor zVAD-fmk or cathepsin B inhibitor, CA-074. Data are mean ± SEM, n = 5 replicates per treatment. Statistical analysis, one-way ANOVA. **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> = 0.001, ns, not significant. Data are representative of three independent experiments.</p>
Full article ">Figure 7
<p><span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>−/−</sup> <span class="html-italic">mice</span> have a significantly extended lifespan and display improved tremors: (<b>a</b>) Kaplan–Meier survival plot of <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>−/−</sup> mice as compared with <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>+/<span class="html-italic">−</span></sup> animals. (<b>b</b>) Both male and female <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>−/−</sup>mice have significantly extended lifespans in comparison with male and female <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>+/<span class="html-italic">−</span></sup> animals. Data are mean ± SEM, n = 5–24. Statistical analysis, Student’s <span class="html-italic">t</span>-test; *** <span class="html-italic">p</span> = 0.0006, ** <span class="html-italic">p</span> = 0.0023. (<b>c</b>) Profile of tremor amplitudes at multiple frequencies for <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>+/−</sup> mice (<b>left</b>)) and <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>−/−</sup> mice (<b>right</b>) at different ages. (<b>d</b>) Tremor amplitude at 20 Hz for <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>+/−</sup> and <span class="html-italic">hexb</span><sup>−/−</sup><span class="html-italic">Il1r1</span><sup>−/−</sup> mice at different ages. Data shown are mean ± SEM, n = 5–20. Statistical analysis, two-way ANOVA. Statistical significances are as indicated. Data are representative of two independent experiments.</p>
Full article ">Figure 8
<p>Blockade of IL-1β activity with anakinra significantly extends lifespan of <span class="html-italic">hexb</span><sup>−/−</sup> mice and improves neurological function: (<b>a</b>) Kaplan–Meier survival plot of <span class="html-italic">hexb</span><sup>−/−</sup> mice administered with anakinra (red columns, opentriangles) or vehicle (cyan columns, open circles). (<b>b</b>) <span class="html-italic">hexb</span><sup>−/−</sup> mice treated with anakinra (red triangles) have a significantly increased lifespan in comparison with vehicle-treated mice (cyan circles). Data are mean ± SEM, n = 5. ** <span class="html-italic">p</span> = 0.0039. Student’s <span class="html-italic">t</span>-test. (<b>c</b>) Frequency of center-rearing events by anakinra-treated <span class="html-italic">hexb</span><sup>−/−</sup> mice and vehicle-treated animals. Data are mean± SEM, n = 3–5. Statistical analysis, Student’s <span class="html-italic">t</span>-test; statistical significance values are as indicated. Data are representative of 2 independent experiments.</p>
Full article ">
24 pages, 5516 KiB  
Article
DAG-MAG-ΒHB: A Novel Ketone Diester Modulates NLRP3 Inflammasome Activation in Microglial Cells in Response to Beta-Amyloid and Low Glucose AD-like Conditions
by Valentina Gentili, Giovanna Schiuma, Latha Nagamani Dilliraj, Silvia Beltrami, Sabrina Rizzo, Djidjell Lara, Pier Paolo Giovannini, Matteo Marti, Daria Bortolotti, Claudio Trapella, Marco Narducci and Roberta Rizzo
Nutrients 2025, 17(1), 149; https://doi.org/10.3390/nu17010149 - 31 Dec 2024
Viewed by 267
Abstract
Background: A neuroinflammatory disease such as Alzheimer’s disease, presents a significant challenge in neurotherapeutics, particularly due to the complex etiology and allostatic factors, referred to as CNS stressors, that accelerate the development and progression of the disease. These CNS stressors include cerebral hypo-glucose [...] Read more.
Background: A neuroinflammatory disease such as Alzheimer’s disease, presents a significant challenge in neurotherapeutics, particularly due to the complex etiology and allostatic factors, referred to as CNS stressors, that accelerate the development and progression of the disease. These CNS stressors include cerebral hypo-glucose metabolism, hyperinsulinemia, mitochondrial dysfunction, oxidative stress, impairment of neuronal autophagy, hypoxic insults and neuroinflammation. This study aims to explore the efficacy and safety of DAG-MAG-ΒHB, a novel ketone diester, in mitigating these risk factors by sustaining therapeutic ketosis, independent of conventional metabolic pathways. Methods: We evaluated the intestinal absorption of DAG-MAG-ΒHB and the metabolic impact in human microglial cells. Utilizing the HMC3 human microglia cell line, we examined the compound’s effect on cellular viability, Acetyl-CoA and ATP levels, and key metabolic enzymes under hypoglycemia. Additionally, we assessed the impact of DAG-AG-ΒHB on inflammasome activation, mitochondrial activity, ROS levels, inflammation and phagocytic rates. Results: DAG-MAG-ΒHB showed a high rate of intestinal absorption and no cytotoxic effect. In vitro, DAG-MAG-ΒHB enhanced cell viability, preserved morphological integrity, and maintained elevated Acetyl-CoA and ATP levels under hypoglycemic conditions. DAG-MAG-ΒHB increased the activity of BDH1 and SCOT, indicating ATP production via a ketolytic pathway. DAG-MAG-ΒHB showed remarkable resilience against low glucose condition by inhibiting NLRP3 inflammasome activation. Conclusions: In summary, DAG-MAG-ΒHB emerges as a promising treatment for neuroinflammatory conditions. It enhances cellular health under varying metabolic states and exhibits neuroprotective properties against low glucose conditions. These attributes indicate its potential as an effective component in managing neuroinflammatory diseases, addressing their complex progression. Full article
(This article belongs to the Section Nutrition and Neuro Sciences)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>ESI-MS spectrum of (<b>a</b>) DAG-ΒHB and (<b>b</b>) MAG-ΒHB.</p>
Full article ">Figure 2
<p>(<b>a</b>) Evaluation of cytotoxicity by MTT assay after 24 h treatment with different concentrations of DAG/MAG-ΒHB of Caco-2cells (12.5, 25, 50, 100, 150, 200 mM). (<b>b</b>) Intestinal absorption of DAG/MAG-ΒHB determined using an in vitro human intestinal model based on human Caco-2 intestinal adenocarcinoma cells. The bioavailability was expressed as a percentage of uptake (%), calculated on the mass balance of the active ingredient (amount of active ingredient in apical + amount of active ingredient in basolateral). (<b>c</b>) Pharmacokinetics evaluation was performed in blood samples collected at the intervals of 30, 60, 90, and 120 min post-supplementation with 0.9% NaCl saline solution as the vehicle or DAG/MAG-ΒHB at the concentrations of 500 mg/kg, 1000 mg/kg, and 2000 mg/kg. Gas chromatography was used to measure blood DAG-MAG/ΒHB. (<b>d</b>) Paraffin-embedded tissue specimens were sectioned at 5 μm, and then haematoxylin and eosin stain were performed. For histological investigations, the sections were stained with haematoxylin and eosin and then observed under optical microscope.</p>
Full article ">Figure 3
<p>(<b>a</b>) Assessment of BBB passage of DAG/MAG-BHB (10 mM) and TMS (10 mM) through BBB in vitro system (Transwell endothelial cells (hBMECs) system) during 240 min observation. Gas chromatography was used to measure blood DAG-MAG/ΒHB and TMS. (<b>b</b>) Morphological evaluation of HMC3 cell line treated with different concentrations of glucose in the presence/absence of DAG/MAG-BHB. (<b>c</b>) Viability evaluation by MTT assay of HMC3 cell line treated with different concentrations of glucose in the presence/absence of DAG/MAG-BHB. (<b>d</b>) Levels of pyruvate, lactate, AcAc and acetylCoA in the presence of 5 mM glucose for 18 h in HMC3 cells. (<b>e</b>) BDH1, (<b>f</b>) SCOT, (<b>g</b>) ATP. Mean ± SEM for each group is reported. <span class="html-italic">p</span> values were obtained by statistical analysis with the Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 4
<p>(<b>a</b>) Evaluation of IL-1beta secretion in inflammasomes-primed HCM3 cells. Evaluation of IL-1beta secretion in Aβ1-42-primed HCM3 cells in (<b>b</b>) normal (25 mM) glucose medium and in (<b>c</b>) low (5.5 mM) glucose medium. (<b>d</b>) Evaluation of IL-1beta secretion in Aβ1-42-primed HCM3 cells in low glucose medium with pharmacological inhibition of TLR4 by CLI-095. (<b>e</b>) Evaluation of IL-1beta secretion in Aβ1-42-primed HCM3 cells in low glucose medium with 10 mM DAG/MAG-ΒHB (DAG/MAG-ΒHB). (<b>f</b>) Caspase-1 activation in Aβ1-42-primed HCM3 cells in low glucose medium with or without 10 mM DAG/MAG-ΒHB (DAG/MAG-ΒHB). (<b>g</b>) Caspase-1 and (<b>h</b>) IL-1beta cleavage in Aβ42-primed HCM3 in low glucose medium in the absence (Aβ1-42) or presence of 10 mM DAG/MAG-ΒHB (DAG/MAG-BHB). (<b>i</b>) NLRP3 mRNA expression of in Aβ42-primed HCM3 in low glucose medium in the absence (Aβ1-42) or presence of 10 mM DAG/MAG-ΒHB (DAG/MAG-BHB). (<b>j</b>) Immunofluorescence staining for ASC (red) speck and nuclei (DAPI) in Aβ42-primed HCM3 in low glucose medium in the absence (Aβ1-42) or presence of 10 mM DAG/MAG-ΒHB (DAG/MAG-BHB). Scale bar: 20 μm. (<b>k</b>) Percentages of microglia containing ASC foci were quantified. (<b>l</b>) Relative phagocytic activity of microglia measured by bead uptake assay in Aβ42-primed HCM3 in the absence (Aβ1-42) or presence of 10 mM DAG/MAG-ΒHB (DAG/MAG-BHB). Mean ± SEM for each group is reported. <span class="html-italic">p</span> values were obtained by statistical analysis with the Student’s <span class="html-italic">t</span>-test and Fisher’s exact test.</p>
Full article ">Figure 5
<p>(<b>a</b>) Mitochondrial membrane potential with Jc-1 staining method in Aβ1-42-primed HCM3 in low glucose medium. (<b>b</b>) Quantitative analysis of the ratio of red/green fluorescent intensity. (<b>c</b>) ATP content in in Aβ1-42-primed HCM3 in low glucose medium. (<b>d</b>) ROS levels in Aβ1-42-primed HCM3 in low glucose medium evaluated by the probe CM-H2DCFDA. (<b>e</b>) Quantitative analysis of fold intensity of CM-H2DCFDA intensity. Mean ± SEM for each group is reported. <span class="html-italic">p</span> values were obtained by statistical analysis with the Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Scheme 1
<p>Overall reaction scheme.</p>
Full article ">Scheme 2
<p>Mechanisms of (<b>A</b>) MAG- and (<b>B</b>) BAG-ΒHB formation.</p>
Full article ">
20 pages, 6040 KiB  
Article
Harnessing the Power of Machine Learning Guided Discovery of NLRP3 Inhibitors Towards the Effective Treatment of Rheumatoid Arthritis
by Sidra Ilyas, Abdul Manan, Chanyoon Park, Hee-Geun Jo and Donghun Lee
Cells 2025, 14(1), 27; https://doi.org/10.3390/cells14010027 - 30 Dec 2024
Viewed by 210
Abstract
The NLRP3 inflammasome, plays a critical role in the pathogenesis of rheumatoid arthritis (RA) by activating inflammatory cytokines such as IL1β and IL18. Targeting NLRP3 has emerged as a promising therapeutic strategy for RA. In this study, a multidisciplinary approach combining machine learning, [...] Read more.
The NLRP3 inflammasome, plays a critical role in the pathogenesis of rheumatoid arthritis (RA) by activating inflammatory cytokines such as IL1β and IL18. Targeting NLRP3 has emerged as a promising therapeutic strategy for RA. In this study, a multidisciplinary approach combining machine learning, quantitative structure–activity relationship (QSAR) modeling, structure–activity landscape index (SALI), docking, molecular dynamics (MD), and molecular mechanics Poisson–Boltzmann surface area MM/PBSA assays was employed to identify novel NLRP3 inhibitors. The ChEMBL database was used to retrieve compounds with known IC50 values to train machine learning (ML) models using the Lazy Predict package. After data pre-processing, 401 non-redundant structures were selected for exploratory data analysis (EDA). PubChem and MACCS fingerprints were used to predict the inhibitory activities of the compounds. SALI was used to identify structurally similar compounds with significantly different biological activities. The compounds were docked using MOE to assess their binding affinities and interactions with key residues in NLRP3. The models were evaluated, and a comparative analysis revealed that the ensemble Random Forest (RF) model (PubChem fingerprints) with RMSE (0.731), R2 (0.622), and MAPE (8.988) and bootstrap aggregating model (MACCS fingerprints) with RMSE (0.687), R2 (0.666), and MAPE (9.216) on the testing set performed well, in accordance with the Organization for Economic Cooperation and Development (OECD) guidelines. Out of all docked compounds, the two most promising compounds (ChEMBL5289544 and ChEMBL5219789) with binding scores of −7.5 and −8.2 kcal/mol were further investigated by MD to evaluate their stability and dynamic behavior within the binding site. MD simulations (200 ns) revealed strong structural stability, flexibility, and interactions in the selected complexes. MM/PBSA binding free energy calculations revealed that van der Waals and electrostatic forces were the key drivers of the binding of the protein with ligands. The outcomes obtained can be used to design more potent and selective NLRP3 inhibitors as therapeutic agents for the treatment of inflammatory diseases such as RA. However, concerns related to the lack of large datasets, experimental validation, and high computational costs remain. Full article
(This article belongs to the Special Issue Novel Therapeutic Targets of Rheumatoid Arthritis)
Show Figures

Figure 1

Figure 1
<p>Cathepsin release from bone activates NLRP3, which triggers caspase1 to cause cleavage of gasdermin D (GSDMD), and release of inflammatory cytokines (IL1β and IL18) leading to joint inflammation in RA.</p>
Full article ">Figure 2
<p>Research workflow of the study conducted.</p>
Full article ">Figure 3
<p>pIC<sub>50</sub> and chemical space of molecules. (<b>a</b>) pIC<sub>50</sub> of active and inactive compounds; (<b>b</b>) scatter plot of molecular weight (MW) vs. octanol–water partition coefficient (LogP) indicating the chemical space occupied by active (green) and inactive (blue) compounds.</p>
Full article ">Figure 4
<p>Box plots of physicochemical properties of drug-likeness between active and inactive compounds. (<b>a</b>–<b>f</b>) were showing the six distribution patterns of MW, LogP, nHA, nHD, nRot, and TPSA.</p>
Full article ">Figure 5
<p>Comparison of solvent accessible surface (SAS) maps using PubChem and MACCS fingerprints with activity cliffs quadrant set as X &gt; 0.9 and Y &gt; 2. The gradual transformation of color from green to red indicates a steady increase in the SALI value.</p>
Full article ">Figure 6
<p>Applicability domain (AD) visualization of (<b>a</b>) PubChem fingerprints and (<b>b</b>) MACCS fingerprints by PCA. The distribution of the training (blue circles) and test (red circles) datasets in the chemical space is represented. The two datasets are dispersed in the chemical space over the same area, and the test set falls within the AD of the model, indicating the model’s prediction efficiency.</p>
Full article ">Figure 7
<p>Two-dimensional illustration by Discovery studio visualizer. The reference and representative complexes (1 and 2) showed interactions with amino acid residues at the active site of NLRP3 where green: conventional hydrogen bond, light green: carbon–hydrogen bond, yellow: π-sulfur, orange: π-cation, pink: alkyl, light pink: π-alkyl bond.</p>
Full article ">Figure 8
<p>MD simulation of compounds during the 200 ns time period. The changes in RMSD, RMSF, Rg, and hydrogen bonding plots reveal the protein–ligand complex stability, flexibility, compactness, and interaction patterns.</p>
Full article ">Figure 9
<p>MM/PBSA analysis (<b>left</b>) binding free energy contribution by various interactions, (<b>right</b>) binding free energy contribution by active residues and ligand.</p>
Full article ">
16 pages, 5109 KiB  
Article
Zanthoxylum piperitum Attenuates Monosodium Urate-Induced Gouty Arthritis: A Network Pharmacology Investigation of Its Anti-Inflammatory Mechanisms
by Sung Wook Kim, Soo Hyun Jeong, Jong Uk Kim, Mi Hye Kim, Wonwoong Lee, Cheol-Jung Lee, Tae Han Yook and Gabsik Yang
Pharmaceuticals 2025, 18(1), 29; https://doi.org/10.3390/ph18010029 - 29 Dec 2024
Viewed by 317
Abstract
Background: Monosodium urate crystal accumulation in the joints is the cause of gout, an inflammatory arthritis that is initiated by elevated serum uric acid levels. It is the most prevalent form of inflammatory arthritis, affecting millions worldwide, and requires effective treatments. The necessity [...] Read more.
Background: Monosodium urate crystal accumulation in the joints is the cause of gout, an inflammatory arthritis that is initiated by elevated serum uric acid levels. It is the most prevalent form of inflammatory arthritis, affecting millions worldwide, and requires effective treatments. The necessity for alternatives with fewer side effects is underscored by the frequent adverse effects of conventional therapies, such as urate-lowering drugs. IL-1β is a potential therapeutic target due to its significant role in the inflammatory response induced by MSU. Zanthoxylum piperitum (ZP), a shrub that possesses antibacterial, antioxidant, and anti-inflammatory properties, has demonstrated potential in the treatment of inflammatory conditions. Methods: For anti-inflammatory properties of ZP, Raw264.7 cell stimulated LPS were treated ZP and using RNA-seq with Bone marrow derived macrophage, we observed to change inflammatory gene. Pharmacological networks were conducted to select target gene associated with ZP. For in vivo, mice were injected MSU in footpad for induce gouty arthritis model. The components of ZP were analyzed using GC-MS, and distilled extracts of ZP (deZP) were prepared. Results: In vitro, deZP decreased inflammatory cytokines. However, in vivo, it also decreased paw thickness and IL-1β levels. The anti-inflammatory effects of deZP are believed to be mediated through the NLRP3 inflammasome pathway, as indicated by RNA sequencing and network pharmacology analyses. Conclusions: ZP has an anti-inflammatory effect and regulation of the NLRP3 inflammasome in vitro and in vivo. Further research, including clinical trials, is required to confirm the safety of deZP, determine the optimal dosing, and evaluate its long-term effects. Full article
(This article belongs to the Special Issue Network Pharmacology of Natural Products)
Show Figures

Figure 1

Figure 1
<p>Anti-inflammatory effects of deZP. (<b>A</b>) RAW264.7 cells were treated with deZP at concentrations ranging from 0.32% to 5% for 24 h. (<b>B</b>–<b>D</b>) RAW264.7 cells were treated with deZP at concentrations of 0.63%, 1.25%, and 2.5% for 1 h, followed by stimulation with LPS (100 ng/mL) with deZP for 24 h. (<b>C</b>,<b>D</b>) Cell supernatants were collected for analysis of IL-6 and TNF-α using ELISA. (<b>E</b>) RAW264.7 cells were stimulated with LPS (100 ng/mL) and treated with deZP or colchicine (10 μM). The data are presented as mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle. †† <span class="html-italic">p</span> &lt; 0.01, ††† <span class="html-italic">p</span> &lt; 0.001 vs. LPS.</p>
Full article ">Figure 2
<p>Analysis of significantly differentially expressed genes (DEGs) and gene ontology (GO). (<b>A</b>,<b>B</b>) Heatmap showing genes with differential expression and exhibiting a scatter plot of DEG. They show inflammasome categories. (<b>A</b>) red boxes are presented as target genes. (<b>B</b>) red spots mean up regulated genes, and green are down regulated genes. (<b>C</b>) Gene ontology shows regulated genes by deZP in specific signaling (cytokine–cytokine receptor interactions and NF-kappa B signaling pathway). Red boxes are up-regulated genes, and blue are down-regulated genes.</p>
Full article ">Figure 2 Cont.
<p>Analysis of significantly differentially expressed genes (DEGs) and gene ontology (GO). (<b>A</b>,<b>B</b>) Heatmap showing genes with differential expression and exhibiting a scatter plot of DEG. They show inflammasome categories. (<b>A</b>) red boxes are presented as target genes. (<b>B</b>) red spots mean up regulated genes, and green are down regulated genes. (<b>C</b>) Gene ontology shows regulated genes by deZP in specific signaling (cytokine–cytokine receptor interactions and NF-kappa B signaling pathway). Red boxes are up-regulated genes, and blue are down-regulated genes.</p>
Full article ">Figure 3
<p>Network of Terpinen-4-ol as ZP’s active compound. (<b>A</b>) Whole network Terpine-4-ol as an active compound of ZP with 104 nodes and 1434 edges. (<b>B</b>) Venn diagram of intersection target genes between the Terpinen-4-ol network and ‘gout’ gene set. (<b>C</b>) Eight common genes of Terpinen-4-ol and gout. (<b>D</b>) Network of common genes of Terpinen-4-ol and gout with 8 nodes and 28 edges.</p>
Full article ">Figure 4
<p>Functional enrichment analysis of the ZP network and ‘gout’ gene set. (<b>A</b>) Biological terms of the intersecting genes of ZP and gout using a KEGG Pathways database in high FDR value order. (<b>B</b>) Significantly enriched pathway terms of ZP in gout on a KEGG Pathways database. (<b>C</b>) Biological terms of the intersecting genes of ZP and gout using a GO Biological Process database in high FDR value order. (<b>D</b>) Significantly enriched pathway terms of ZP in gout on a GO Biological Process database.</p>
Full article ">Figure 5
<p>A diagram of the NOD-like receptor signaling pathway in the KEGG Pathway database (hsa04621). All matched genes are marked in a red box.</p>
Full article ">Figure 6
<p>Treatment with deZP attenuates in vivo inflammatory symptoms. (<b>A</b>) Time course of footpad thickness after oral administration of deZP and injection of MSU crystals. (<b>B</b>) IL-1β levels in footpad tissue measured by ELISA. (<b>C</b>) Footpad tissues were analyzed by Western blot for NLRP3, caspase-1, and IL-1β. (<b>D</b>) Real-time PCR shows that mRNA expression levels of NLRP3, caspase-1, and IL-1β in footpad tissue. Data are presented as mean ± SD (<span class="html-italic">n</span> = 5 mice per group). * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle. † <span class="html-italic">p</span> &lt; 0.05 vs. MSU. # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01 vs. MSU.</p>
Full article ">Figure 7
<p>Identification of deZP and their functions. (<b>A</b>) Total ion chromatogram of deZP with labeled peaks: 1. Linalool; 2. Terpinen-4-ol; 3. Alpha-terpineol; 4. Delta 3-carene or Camphene; 5. Piperitone. (<b>B</b>) Function of the identified ingredients of deZP. (<b>C</b>) terpinene-4-ol, the main compound of ZP, was measured for concentrations.</p>
Full article ">
17 pages, 744 KiB  
Review
Mechanisms and Emerging Regulators of Neuroinflammation: Exploring New Therapeutic Strategies for Neurological Disorders
by Mi Eun Kim and Jun Sik Lee
Curr. Issues Mol. Biol. 2025, 47(1), 8; https://doi.org/10.3390/cimb47010008 - 26 Dec 2024
Viewed by 441
Abstract
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative [...] Read more.
Neuroinflammation is a complex and dynamic response of the central nervous system (CNS) to injury, infection, and disease. While acute neuroinflammation plays a protective role by facilitating pathogen clearance and tissue repair, chronic and dysregulated inflammation contributes significantly to the progression of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, and Multiple Sclerosis. This review explores the cellular and molecular mechanisms underlying neuroinflammation, focusing on the roles of microglia, astrocytes, and peripheral immune cells. Key signaling pathways, including NF-κB, JAK-STAT, and the NLRP3 inflammasome, are discussed alongside emerging regulators such as non-coding RNAs, epigenetic modifications, and the gut–brain axis. The therapeutic landscape is evolving, with traditional anti-inflammatory drugs like NSAIDs and corticosteroids offering limited efficacy in chronic conditions. Immunomodulators, gene and RNA-based therapeutics, and stem cell methods have all shown promise for more specific and effective interventions. Additionally, the modulation of metabolic states and gut microbiota has emerged as a novel strategy to regulate neuroinflammation. Despite significant progress, challenges remain in translating these findings into clinically viable therapies. Future studies should concentrate on integrated, interdisciplinary methods to reduce chronic neuroinflammation and slowing the progression of neurodegenerative disorders, providing opportunities for revolutionary advances in CNS therapies. Full article
(This article belongs to the Special Issue The Role of Neuroinflammation in Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>The major molecular pathways driving neuroinflammation in Alzheimer’s disease, highlighting their interconnected roles in perpetuating chronic inflammation. At the center of the diagram is neuroinflammation, which is fueled by four key pathways. The NF-κB pathway, activated by amyloid beta (Aβ) and tau proteins, promotes the production of pro-inflammatory cytokines such as TNF-α and IL-6, amplifying the inflammatory response. The NLRP3 inflammasome, triggered by Aβ, reactive oxygen species (ROS), and mitochondrial dysfunction, leads to the release of IL-1β and IL-18, further escalating neuroinflammation. The JAK-STAT pathway, induced by cytokines like IL-6, drives the activation of neurotoxic astrocytes, which contribute to neuronal damage. Finally, the MAPK pathway, stimulated by Aβ, tau, and oxidative stress, enhances ROS production and cytokine release, exacerbating oxidative damage and inflammation. Together, these pathways form a complex network that underpins the inflammatory processes observed in Alzheimer’s disease.</p>
Full article ">
31 pages, 1122 KiB  
Review
Therapeutic Significance of NLRP3 Inflammasome in Cancer: Friend or Foe?
by Aliea M. Jalali, Kenyon J. Mitchell, Christian Pompoco, Sudeep Poludasu, Sabrina Tran and Kota V. Ramana
Int. J. Mol. Sci. 2024, 25(24), 13689; https://doi.org/10.3390/ijms252413689 - 21 Dec 2024
Viewed by 577
Abstract
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the [...] Read more.
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the tumor microenvironment. The activation of NLRP3 inflammasome could increase immune surveillance and the efficacy of immunotherapy. It can also lead to the removal of tumor cells by the recruitment of phagocytic macrophages, T-lymphocytes, and other immune cells to the tumor site. On the other hand, NLRP3 activation can also be harmful, as chronic inflammation driven by NLRP3 supports tumor progression by creating an environment that facilitates cancer cell proliferation, migration, invasion, and metastasis. The release of pro-inflammatory cytokines such as IL-1β and IL-18 can promote tumor growth and angiogenesis, while sustained inflammation may lead to immune suppression, hindering effective anti-tumor responses. In this review article, we discuss the role of NLRP3 inflammasome-mediated inflammatory response in the pathophysiology of various cancer types; understanding this role is essential for the development of innovative therapeutic strategies for cancer growth and spread. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Canonical and non-canonical activation of NLRP3 inflammasome. The canonical pathway involves the activation of NLRP3 inflammasomes through signals such as mitochondrial ROS, calcium influx, and potassium efflux, leading to NF-κB activation and the production of pro-inflammatory cytokines (IL-1β and IL-18). This pathway ultimately activates caspase-1, resulting in cytokine release and pyroptosis. The non-canonical pathway involving LPS from Gram-negative bacteria triggers caspase-11, which indirectly activates NLRP3, leading to similar inflammasome responses, cytokine production, and pyroptosis. Both pathways generally play critical roles in innate immunity and inflammation.</p>
Full article ">Figure 2
<p>Significance of NLRP3 inflammasome in the melanoma progression. Various factors such as UV radiation, tumor microenvironment, and melanoma cells contribute to oxidative stress and cause DNA damage, immune cell activation, and cytokine release. Oxidative stress, in turn, triggers reactive oxygen species (ROS), mitochondrial DNA damage, potassium efflux, and NF-κB activation, which influence the activation of the NLRP3 inflammasome. NLRP3 activation promotes melanoma progression, metastasis, immune evasion, and therapy resistance. In contrast, inhibition of NLRP3 could enhance immunotherapy, inhibit tumor growth, and reduce metastasis.</p>
Full article ">Figure 3
<p>Significance of NLRP3 inflammasome in leukemias. Oxidative stress and mitochondrial dysfunction in leukemia cells could activate NLRP3 inflammasomes through mitochondrial ROS, potassium efflux, and NF-κB signaling pathways. NLRP3 activation leads to the generation of active IL-1β and IL-18 cytokines, and could cause pyroptosis. Further, NLRP3 activation plays various roles in different leukemias. For example, in Acute Myeloid Leukemia (AML), NLRP3 promotes immune evasion and survival, while inhibition reduces the disease burden. In Chronic Myeloid Leukemia (CML), NLRP3 is linked to KRAS mutations and therapy resistance. In Acute Lymphoblastic Leukemia (ALL), NLRP3 activation is correlated with glucocorticoid resistance, and in Chronic Lymphocytic Leukemia (CLL), P2X7R overexpression leads to increased NLRP3.</p>
Full article ">Figure 4
<p>Role of NLRP3 inflammasome in breast cancer growth and spread. Several factors such as reactive oxygen species (ROS)-induced mitochondrial damage, BRCA1-associated genetic mutations causing mitochondrial dysfunction, extracellular ATP leading to P2X7R overexpression, and inflammatory cytokines that activate NF-κB-mediated inflammasome components could lead to activation of NLRP3 inflammasome. NLRP3-mediated release of IL-1β and IL-18 promotes cancer cell proliferation, survival, migration, immune evasion, and resistance to therapy. Further, the outcomes also include increased tumor growth, metastasis, compromised immune surveillance, and drug resistance.</p>
Full article ">Figure 5
<p>Significance of NLRP3 inflammasome activation in cigarette smoke and COPD-induced lung cancer development. Cigarette smoke leads to reactive oxygen species (ROS), mitochondrial damage, and tissue dysfunction, activating the NLRP3 inflammasome. The release of pro-inflammatory cytokines IL-1β and IL-18 contributes to prolonged inflammation. COPD-induced inflammation and immune cell recruitment, such as macrophages, further amplify this process. The persistent inflammation and oxidative stress promote DNA damage, genetic mutations, and genomic instability, ultimately leading to lung carcinogenesis.</p>
Full article ">Figure 6
<p>Role of NLRP3 inflammasome activation in promoting colon cancer development. During inflammatory bowel disease (IBD), such as Crohn’s disease and ulcerative colitis, the inflammation of the gut lining activates immune cells, causing oxidative stress, mitochondrial damage, and the release of DAMPs. Gut microbiome imbalance (dysbiosis) leads to pathogenic bacterial growth and loss of gut-barrier integrity, allowing pathogen and toxin leakage, further driving oxidative stress. These pathways cause NLRP3 activation and trigger IL-1β and IL-18 release. Increased inflammasome response results in immune system imbalance, epithelial-barrier dysfunction, and neoplasia initiation, ultimately contributing to colon cancer growth and metastasis.</p>
Full article ">
16 pages, 6187 KiB  
Article
Aerosol Inhalation of Luteolin-7-O-Glucuronide Exerts Anti-Inflammatory Effects by Inhibiting NLRP3 Inflammasome Activation
by Jianliang Li, Ling Song, Han Li, Yunhang Gao, Tengfei Chen, Zhongxiu Zhang, Hongping Hon, Zuguang Ye and Guangping Zhang
Pharmaceuticals 2024, 17(12), 1731; https://doi.org/10.3390/ph17121731 - 21 Dec 2024
Viewed by 408
Abstract
Background: Luteolin-7-O-glucuronide (L7Gn) is a flavonoid isolated from numerous traditional Chinese herbal medicines that exerts anti-inflammatory effects. Previous research has revealed that aerosol inhalation is the most straightforward way of administration for the delivery of respiratory agents. Thus far, the impact of aerosol [...] Read more.
Background: Luteolin-7-O-glucuronide (L7Gn) is a flavonoid isolated from numerous traditional Chinese herbal medicines that exerts anti-inflammatory effects. Previous research has revealed that aerosol inhalation is the most straightforward way of administration for the delivery of respiratory agents. Thus far, the impact of aerosol inhalation of L7Gn on lung inflammation and the underlying mechanisms remain unknown. Methods: The real-time particle size for L7Gn aerosol inhalation was detected by the Spraytec spray droplet size measurement system, including transmission and size diameters. The acute lung injury (ALI) rat model was induced by aerosol inhalation of LPS to evaluate the protective effect of L7Gn. The inhibitory effect of NLRP3 inflammasome activation assays was conducted in LPS-induced MH-S cells. Elisa, Western blotting, and RT-PCR were utilized to investigate the expression of NLRP3 inflammasome-relevant proteins and genes. Results: In this study, we found that inhalation of L7Gn aerosol significantly reduced pulmonary injury by inhibiting inflammatory infiltration and enhancing lung function. Meanwhile, the NLR family pyrin domain containing 3 (NLRP3) inflammasome was activated dramatically, accompanied by upregulated expression of IL-1β and IL-18, both in the ALI rat model and in LPS-induced MH-S cells. Moreover, L7Gn was found to significantly downregulate the expression of NLRP3, ASC, caspase-1, and cleaved caspase-1, which are critical components of the NLRP3 inflammasome, as well as the expression of IL-1β and IL-18. Conclusions: Based on our findings, L7Gn could exert anti-inflammatory effects by inhibiting NLRP3 inflammasome activation, which may emerge as potential therapeutic agents for the treatment of ALI. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>The structure of L7Gn (<b>A</b>), the compound of interest, and the experimental design of the study (<b>B</b>).</p>
Full article ">Figure 2
<p>The parameter of real-time particle size of L7Gn. (<b>A</b>) The histogram of particles diameter; (<b>B</b>) Real-time transmission and percentiles of inhalation of aerosolized L7Gn, Trans (purple line) Dv(10) (dark green line), Dv(50) (light green line), and Dv(90) (green line).</p>
Full article ">Figure 3
<p>The effect of L7Gn on pulmonary function in LPS-induced ALI rats. Pulmonary function was measured quantitatively by assaying for TV, MV, Cdyn, and Ve (<b>A</b>–<b>D</b>). All the data presented as mean ± SEM. <span class="html-italic">n</span> = 5. **, <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>L7Gn inhibits LPS-induced pulmonary damage in rats. Tissue sections of a representative lung section from each group: normal group (<b>A</b>,<b>a</b>), LPS + vehicle group (<b>B</b>,<b>b</b>), LPS + L7Gn 10 min group (<b>C</b>,<b>c</b>), LPS + L7Gn 20 min group (<b>D</b>,<b>d</b>), and LPS + dexamethasone group (<b>E</b>,<b>e</b>), which were stained with hematoxylin and eosin (H&amp;E) ((<b>A</b>–<b>E</b>), 25×; (<b>a</b>–<b>e</b>), 100×), bronchiolar inflammation (red arrow), and interstitial thickening (green arrow).</p>
Full article ">Figure 5
<p>L7Gn inhibited NLRP3 inflammasome activation in LPS-induced rats. Sections of lung tissue were immunostained with CD68 (green) and Caspase-1 (red) antibodies shown as representative images (<b>A</b>); Representative Western blotting of NLRP3, Caspase-1, and ASC (<b>A</b>): The relative protein expression of NLRP3, Caspase-1, and ASC normalized to that of β-actin. (<b>B</b>–<b>E</b>). All data are presented as mean ± SEM., <span class="html-italic">n</span> = 3. * <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>L7Gn decreased the levels of inflammatory cytokines in LPS-induced bronchiolar inflammation. The levels of IL-1β in BALF (<b>A</b>) and lung homogenates (<b>B</b>) and IL-18 in BALF (<b>C</b>) and lung homogenates (<b>D</b>) were determined using ELISA kits. All data are presented as mean ± SEM., <span class="html-italic">n</span> = 6. * <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>L7Gn inhibited NLRP3 inflammasome activation in LPS-induced MH-S cells. Representative Western blotting of NLRP3, caspase-1, IL-1β, ASC, IL-18, cleaved IL-1β, and cleaved caspase-1 (<b>A</b>). The relative protein expression of NLRP3, caspase-1, IL-1β, ASC, IL-18, cleaved IL-1β, and cleaved caspase-1 was normalized to that of β-actin (<b>B</b>–<b>H</b>). Relative mRNA expression of IL-1β and IL-18 (<b>I</b>,<b>J</b>). All data are presented as mean ± SEM., <span class="html-italic">n</span> = 3. * <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 ">
17 pages, 3630 KiB  
Article
Porcine Epidemic Diarrhea Virus Infection of Porcine Intestinal Epithelial Cells Causes Mitochondrial DNA Release and the Activation of the NLRP3 Inflammasome to Mediate Interleukin-1β Secretion
by Di Bao, Shushuai Yi, Luobing Zhao, Han Zhao, Jiuyuan Liu, Yiming Wei, Guixue Hu and Xinxin Liu
Vet. Sci. 2024, 11(12), 643; https://doi.org/10.3390/vetsci11120643 - 12 Dec 2024
Viewed by 901
Abstract
Porcine epidemic diarrhea virus (PEDV) induces enteritis and diarrhea in piglets. Mitochondrial DNA (mtDNA) contributes to virus-induced inflammatory responses; however, the involvement of inflammasomes in PEDV infection responses remains unclear. We investigated the mechanism underlying inflammasome-mediated interleukin (IL)-1β secretion during the PEDV infection [...] Read more.
Porcine epidemic diarrhea virus (PEDV) induces enteritis and diarrhea in piglets. Mitochondrial DNA (mtDNA) contributes to virus-induced inflammatory responses; however, the involvement of inflammasomes in PEDV infection responses remains unclear. We investigated the mechanism underlying inflammasome-mediated interleukin (IL)-1β secretion during the PEDV infection of porcine intestinal epithelial (IPEC-J2) cells. IL-1β production and caspase-1 activity were assessed by quantitative PCR and enzyme-linked immunosorbent assay. NLRP3 inflammasome activation was assessed using immunoprecipitation experiments. Mitochondrial damage was evaluated by analyzing the mitochondrial membrane potential and ATP levels and by the flow cytometry examination of mitochondrial reactive oxygen species (mtROS). Mitochondria and mtDNA localization were observed using immunofluorescence. The inhibition of mtROS and mtDNA production allowed NLRP3 inflammasome and IL-1β expression detection and the evaluation of the pathway underlying NLRP3 inflammasome activation in PEDV-infected IPEC-J2 cells. IPEC-J2 cells upregulated IL-1β upon PEDV infection, where mature IL-1β secretion depended on caspase-1 activity, triggered NLRP3 inflammasome expression and assembly, and caused mitochondrial dysfunction, leading to mtDNA release and NLRP3 inflammasome activation, while mtROS contributed to NF-κB pathway activation, enhancing IL-1β secretion. This is the first demonstration of the mechanism underlying mtDNA release and NLRP3 inflammasome activation facilitating IL-1β secretion from PEDV-infected IPEC-J2 cells. These data enhance our understanding of the inflammatory mechanisms triggered by PEDV. Full article
Show Figures

Figure 1

Figure 1
<p>Porcine epidemic diarrhea virus (PEDV) infects IPEC-J2 cells to promote the secretion of mature interleukin (IL)-1β. PEDV (multiplicity of infection (MOI) = 1) was used to infect IPEC-J2 cells for the specified periods of time. Detection of PEDV N protein expression via Western blot following PEDV infection in IPEC-J2 cells (<b>a</b>). mRNA expression (<b>b</b>) and Western blot (<b>f</b>) analyses of IL-1β levels, as well as the results of an enzyme-linked immunosorbent assay (ELISA) to assess IL-1β secretion in the cell supernatants (<b>c</b>). IPEC-J2 cells were infected with PEDV at the specified dose for 24 h, followed by fluorescence quantification (<b>d</b>), Western blotting (<b>g</b>), and the ELISA detection of IL-1β secretion in the cell supernatant (<b>e</b>). Data represent the mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>PEDV promotes caspase-1 enzymatic activity in IPEC-J2 cells. Caspase-1 enzyme activity (<b>a</b>) and Western blot results (<b>b</b>) at the specified time points after the PEDV infection of IPEC-J2 cells (MOI = 1). Caspase-1 enzyme activity (<b>c</b>) and Western blot results (<b>d</b>) of IPEC-J2 cells infected with PEDV at the specified doses for 24 h. IPEC-J2 cells were treated with the caspase-1 inhibitor, Ac-YVAD-cmk, at the specified concentration for 1 h and then inoculated with PEDV (MOI = 1). ELISA was used to detect IL-1β secretion in the cell supernatant 24 h after infection (<b>e</b>). Data represent the mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>PEDV infection of IPEC-J2 cells activates the NLRP3 inflammasome. PEDV was used to infect IPEC-J2 cells at the specified doses for 24 h and then the <span class="html-italic">NLRP3</span> inflammasome (<b>a</b>) and <span class="html-italic">ASC</span> (<b>c</b>) mRNA expression were analyzed. <span class="html-italic">NLRP3</span> (<b>b</b>) and <span class="html-italic">ASC</span> (<b>d</b>) mRNA expression in IPEC-J2 cells infected with PEDV (MOI = 1) for the specified times. NLRP3 (<b>e</b>) and ASC (<b>f</b>) rabbit-derived primary antibodies were used as bait antibodies and rabbit serum was used as the negative control antibody in a co-immunoprecipitation experiment. Data represent the mean ± SD (<span class="html-italic">n</span> = 3). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>PEDV-infected IPEC-J2 cells secrete IL-1β through NLRP3 inflammasome activity. IPEC-J2 cells were treated with 10 µM MCC950 for 1 h, and negative control cells were treated with DMSO for the same time. After 24 h of cell infection with PEDV (MOI = 1), IL-1β secretion in the cell supernatants was detected by ELISA (<b>a</b>) and <span class="html-italic">IL-1β</span> mRNA quantified by fluorescence (<b>c</b>). siNLRP3 and siCtrl (control) were transfected into IPEC-J2 cells. PEDV was used to infect cells (MOI = 1) for 24 h, IL-1β secretion in the cell supernatants was detected by ELISA (<b>b</b>), and <span class="html-italic">IL-1β</span> mRNA expression was quantified by fluorescence (<b>d</b>). Western blot of IPEC-J2 cells treated with 10 µM MCC950 (<b>e</b>) and siNLRP3 transfection of IPEC-J2 cells to detect the expression of inflammasome proteins (<b>f</b>). Data represent the mean ± SD (<span class="html-italic">n</span> = 3),** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>PEDV infection causes mitochondrial dysfunction and results in mitochondrial reactive oxygen species (mtROS) production and mitochondrial DNA (mtDNA) release. Mitochondrial membrane potential was decreased (<b>a</b>). Reduced ATP production after PEDV infection (<b>b</b>). Flow cytometry fluorescence intensity analysis of mtROS production after PEDV infection (<b>c</b>). Immunofluorescence showing mtDNA release after PEDV infection (<b>d</b>). Data represent mean ± SD (<span class="html-italic">n</span> = 3), ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>mtROS are involved in NF-κB activation in PEDV-infected IPEC-J2 cells. IPEC-J2 cells were infected with PEDV at the specified doses for 24 h and the expression levels of pp65 and p65 detected by Western blot (<b>a</b>). IPEC-J2 cells were treated with the NF-κB inhibitor BAY11-7082 (10 μM). <span class="html-italic">IL-1β</span> mRNA expression was detected 24 h after PEDV infection (MOI = 1) (<b>b</b>) and IL-1β secretion in the cell supernatant detected by ELISA (<b>c</b>). Western blot to assess the expression of pp65, p65, and IL-1β proteins (<b>d</b>). After IPEC-J2 cells were treated with 10 µM Mito-TEMPO for 1 h, <span class="html-italic">IL-1β</span> mRNA expression was detected following PEDV infection (MOI = 1) for 24 h (<b>e</b>) and IL-1β secretion in the cell supernatant detected by ELISA (<b>f</b>). Data represent mean ± SD (<span class="html-italic">n</span> = 3), ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>mtDNA participates in NLRP3 inflammasome activation in PEDV-infected IPEC-J2 cells. After transfection with DNase I, IPEC-J2 cells were infected with PEDV (MOI = 1). IL-1β mRNA expression (<b>a</b>) and secretion in cell supernatants detected by ELISA (<b>b</b>). <span class="html-italic">NLRP3</span> inflammasome mRNA expression (<b>c</b>) and caspase-1 enzyme activity (<b>d</b>). Western blot detection of NLRP3 inflammasome and downstream protein expression after transfection with DNase I protein (<b>e</b>). Data represent mean ± SD (<span class="html-italic">n</span> = 3). ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Schematic showing that PEDV infection leads to cytoplasmic mitochondrial DNA release and the activation of the NLPR3 inflammasome.</p>
Full article ">
11 pages, 1000 KiB  
Review
Eastern Equine Encephalitis Virus: The Importance of Metabolism and Aging
by Pawel Kordowitzki
Int. J. Mol. Sci. 2024, 25(24), 13318; https://doi.org/10.3390/ijms252413318 - 12 Dec 2024
Viewed by 674
Abstract
Eastern equine encephalitis virus (EEEV) is a mosquito-transmitted alphavirus that, among humans, can cause a severe and often fatal illness. The zoonotic EEEV enzootic cycle involves a cycle of transmission between Culiseta melanura and avian hosts, frequently resulting in spillover to dead-end vertebrate [...] Read more.
Eastern equine encephalitis virus (EEEV) is a mosquito-transmitted alphavirus that, among humans, can cause a severe and often fatal illness. The zoonotic EEEV enzootic cycle involves a cycle of transmission between Culiseta melanura and avian hosts, frequently resulting in spillover to dead-end vertebrate hosts such as humans and horses. Interestingly, it has been described that the W132G mutation of the very low-density lipoprotein receptor (VLDLR), the receptor of EEEV, significantly enhanced the VLDLR-mediated cell attachment of EEEV. The patient’s metabolism plays a pivotal role in shaping the complex landscape of viral zoonosis. EEEV represents a significant public health concern due to its severe clinical outcomes, challenging epidemiological characteristics, and certain risk factors that heighten susceptibility among specific populations or age groups. Age is one of several predictors that can impact the outcome of EEEV infection; juvenile animals appear to be particularly vulnerable to severe disease. This has also been observed in natural infections, as children are often the most severely impacted humans. The aim of this piece is to shed light on the intricate relationship between human metabolism and the Eastern equine encephalitis virus. Full article
Show Figures

Figure 1

Figure 1
<p>The scheme shows the Eastern equine encephalitis virus (EEEV) transmission.</p>
Full article ">Figure 2
<p>Scheme showing the interaction between EEEV, mitochondria, and the NLRP3 inflammasome. The left part of the scheme represents the priming step, and the right part of the scheme depicts the activation step of the NLRP3 inflammasome. The priming process leads to the activation of the transcription factor NF-κB and the subsequent transcription of canonical and non-canonical components of the NLRP3 inflammasome, whereas the activation process is responsible for the assembly of the NLRP3 complex and the subsequent release of inflammatory cytokines such as IL-1β and IL-18 and others. Priming starts once pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharide (LPS) or cytokines (endogenous) activate the respective receptors on the cell membrane. The activation of the NLRP3 inflammasome during the viral infection upon viral RNA, proteins, and other components can directly or indirectly trigger the NLRP3 inflammasome, resulting in the release of inflammatory cytokines and pyroptosis.</p>
Full article ">
35 pages, 3440 KiB  
Review
Clinical Potential of Misshapen/NIKs-Related Kinase (MINK) 1—A Many-Sided Element of Cell Physiology and Pathology
by Anna Kot, Dominika Koszewska, Błażej Ochman and Elżbieta Świętochowska
Curr. Issues Mol. Biol. 2024, 46(12), 13811-13845; https://doi.org/10.3390/cimb46120826 - 5 Dec 2024
Viewed by 751
Abstract
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of [...] Read more.
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of cellular signaling pathways, which include Wnt signaling, JNK signaling, pathways engaging Ras proteins, the Hippo pathway, and STRIPAK complexes. It thus contributes to regulating the cell cycle, apoptosis, cytoskeleton organization, cell migration, embryogenesis, or tissue homeostasis. MINK1 plays an important role in immunological responses, inhibiting Th17 and Th1 cell differentiation and regulating NLRP3 inflammasome function. It may be considered a link between ROS and the immunological system, and a potential antiviral target for human enteroviruses. The kinase has been implicated in the pathogenesis of sepsis, rheumatoid arthritis, asthma, SLE, and more. It is also involved in tumorigenesis and drug resistance in cancer. Silencing MINK1 reduces cancer cell migration, suggesting potential for new therapeutic approaches. Targeting MINK1 could be a promising treatment strategy for patients insensitive to current chemotherapies, and could improve their prognosis. Moreover, MINK1 plays an important role in the nervous system and the cardiovascular system development and function. The modulation of MINK1 activity could influence the course of neurodegenerative diseases, including Alzheimer’s disease. Further exploration of the activity of the kinase could also help in gaining more insight into factors involved in thrombosis or congenital heart disease. This review aims to summarize the current knowledge on MINK1, highlight its therapeutic and prognostic potential, and encourage more studies in this area. Full article
(This article belongs to the Special Issue Advances in Molecular Pathogenesis Regulation in Cancer 2024)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of the MINK1 protein structure, highlighting its major domains. The protein consists of an N-terminal domain (purple), a conserved kinase domain (blue) responsible for enzymatic activity, a coiled-coil region (residues 394–495; red) involved in actin regulation, a proline-rich region (yellow), and a GCK C-terminal domain (residues 953–1295; orange), containing WD-40 motifs crucial for substrate binding and protein–protein interactions. The GCK domain also includes a Citron-NIK-Homology (CNH) domain (green), implicated in additional regulatory functions.</p>
Full article ">Figure 2
<p>The signaling interactions between the Wnt/Frizzled pathway, MINK1, Prickle1, and their downstream effects on cellular processes. Fz—Frizzled; Dsh—Dishevelled; MINK1—Misshapen-like kinase 1; mTORC2—mammalian target of rapamycin complex 2; AKT—protein Kinase B; Vangl—Van Gogh-like protein; Prickle1—Prickle-like protein 1; Rab5—Ras-related protein Rab-5; CE—convergent extension; CLASP2—cytoplasmic linker associated protein 2; PHLDB2—pleckstrin homology-like domain Family B Member 2; and A/P axis—anterior–posterior axis, *—the possible inhibitors of the relevant components in the presented signaling pathways.</p>
Full article ">Figure 3
<p>MINK1 and RAS signaling interactions. ERK—extracellular signal-regulated kinase; MEK—mitogen-activated protein kinase kinase; RAS-GTP—Ras guanosine triphosphate; Raf—rapidly accelerated fibrosarcoma kinase; pERK—phosphorylated extracellular signal-regulated kinase; MINK1—Misshapen-like kinase 1; ROS—reactive oxygen species; MAP3K5—mitogen-activated protein kinase kinase kinase 5; MMK3/6—mitogen-activated protein kinase kinase 3/6; p38 MAPK—p38 mitogen-activated protein kinase; p21 WAF1/CIP1—p21 wild-type p53-activated fragment 1/cyclin-dependent kinase-interacting protein 1; Rap2—Ras-related protein Rap2; and TANC1—tetratricopeptide repeat, ankyrin repeat, and coiled-coil containing 1.</p>
Full article ">Figure 4
<p>MINK1 and JNK signaling pathway interaction. MAP4K4—mitogen-activated protein kinase kinase kinase kinase 4; MINK1—Misshapen-like kinase 1; TNIK—Traf2- and Nck-interacting kinase; DLK—dual leucine zipper kinase; JNK—c-Jun N-terminal kinase; c-JUN—c-Jun proto-oncogene; and SNHG14—small nucleolar RNA host gene 14.</p>
Full article ">Figure 5
<p>MINK1 and Hippo pathway regulation. ECM—extracellular matrix; Rap2—Ras-related protein 2; MINK1—Misshapen-like kinase 1; LATS1/2—large tumor suppressor kinase 1/2; and YAP/TAZ—Yes-associated protein/WW domain-containing transcription regulator 1.</p>
Full article ">Figure 6
<p>MINK1 and STRIPAK complex. STRIPAK—striatin-interacting phosphatase and kinase; STRN4—Striatin 4; PLK1—Polo-like kinase 1; CDK1—cyclin-dependent kinase 1; MINK1—Misshapen-like kinase 1; and PPP2CA—protein phosphatase 2 catalytic subunit A.</p>
Full article ">Figure 7
<p>MINK1 in immunity. SLE—systemic lupus erythematosus; OCR—open chromatin regions; TCR—T-cell receptor; ROS—reactive oxygen species; NLRP3—NOD-like receptor family pyrin domain containing 3; MINK1—Misshapen-like kinase 1; RA—rheumatoid arthritis; and SNHG14—small nucleolar RNA host gene 14.</p>
Full article ">Figure 8
<p>MINK1 as a key component in different cancer types. HNSC—head and neck squamous cell carcinoma; GBM—glioblastoma multiforme; OSCC—oral squamous cell carcinoma; PRICKLE—Prickle planar cell polarity protein; MINK1—Misshapen-like kinase 1; RICTOR—rapamycin-insensitive companion of mTOR; CLASP2—cytoplasmic linker-associated protein 2; LL5β—pleckstrin homology domain-containing family L member 5β; APC—adenomatous polyposis coli; and CRC—colorectal cancer.</p>
Full article ">Figure 9
<p>MINK1 in cardiovascular diseases. MINK1—Misshapen-like kinase 1; MAPK—mitogen-activated protein kinase; PI3K—phosphoinositide 3-kinase; AKT—AKT serine/threonine kinase; and OFT—outflow tract.</p>
Full article ">Figure 10
<p>MINK1 role in the nervous system and in neurodegenerative diseases. DLK—dual leucine zipper kinase; JNK—c-Jun N-terminal kinase; AMPA-R—alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor; AD—Alzheimer’s disease; and MINK1—Misshapen-like kinase 1.</p>
Full article ">
9 pages, 1324 KiB  
Article
The Role of Beta-Hydroxybutyrate in Mitigating the Inflammatory and Metabolic Consequences of Uric Acid
by Nicole P. Remund, John G. Larsen, Marley J. Shin, Cali E. Warren, Isabelle L. Palmer, Iris J. Kim, Elijah T. Cooper-Leavitt, Derek M. Clarke, Colson G. Beus, Richard J. Johnson, Juan A. Arroyo, Paul R. Reynolds and Benjamin T. Bikman
Metabolites 2024, 14(12), 679; https://doi.org/10.3390/metabo14120679 - 4 Dec 2024
Viewed by 3437
Abstract
Background: Uric acid (UA), a metabolite of purine and fructose metabolism, is linked to inflammation and metabolic disorders, including gout and cardiovascular disease. Its pro-inflammatory effects are largely driven by the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) [...] Read more.
Background: Uric acid (UA), a metabolite of purine and fructose metabolism, is linked to inflammation and metabolic disorders, including gout and cardiovascular disease. Its pro-inflammatory effects are largely driven by the activation of the nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, leading to increased cytokine production. Beta-hydroxybutyrate (BHB), a ketone produced during fasting or carbohydrate restriction, has been shown to reduce inflammation. This study explores the role of BHB in mitigating the inflammatory and metabolic effects of elevated uric acid levels. Methods: We utilized a murine muscle cell culture treated with UA and BHB. Results: Muscle cells treated with UA had increased production of pro-inflammatory cytokines and reduced cell viability. Co-treatment with BHB reversed these effects, improving cell survival and reducing cytokine levels. Additionally, uric acid impaired mitochondrial function and increased oxidative stress, which were mitigated by BHB. Furthermore, uric acid disrupted insulin signaling, but BHB co-treatment restored insulin sensitivity. Conclusions: These findings suggest that BHB holds therapeutic potential by counteracting the inflammatory and metabolic disruptions caused by elevated uric acid, making it a promising target for conditions such as hyperuricemia and metabolic syndrome. Full article
(This article belongs to the Special Issue Exploring Uric Acid and Beyond)
Show Figures

Figure 1

Figure 1
<p>Myoblasts were incubated for 24 h in either uric acid (500 µM), β-hydroxybutyrate (BHB; 5 mM), or both U + B (<span class="html-italic">n</span> = 10). Interferon-γ (IFN-γ; (<b>A</b>) and tumor necrosis factor-α (TNF-α; (<b>B</b>) were measured via dot blot. (<b>C</b>) NLRP3 activity was determined via activity assay. * <span class="html-italic">p</span> &lt; 0.05 vs. CON. # <span class="html-italic">p</span> &lt; 0.05 vs. UA.</p>
Full article ">Figure 2
<p>Myoblasts were incubated for 24 h in either uric acid (500 µM), β-hydroxybutyrate (BHB; 5 mM), or both U + B. Cell viability was measured via the MTT assay (<span class="html-italic">n</span> = 8). * <span class="html-italic">p</span> &lt; 0.05 vs. CON.</p>
Full article ">Figure 3
<p>Myoblasts were incubated for 24 h in either uric acid (500 µM), β-hydroxybutyrate (BHB; 5 mM), or both U + B (<span class="html-italic">n</span> = 5). Mitochondrial respiration (<b>A</b>) was measured by the following strategy: GM indicates glutamate (10 mM) + malate (2 mM); +ADP (2.5 mM); +succinate (S; 10 mM); +FCCP (0.05 μM). H<sub>2</sub>O<sub>2</sub> generation (<b>B</b>) was measured via Amplex Red. The ratio of H<sub>2</sub>O<sub>2</sub> generation to O<sub>2</sub> consumed (<b>C</b>) was also determined. * <span class="html-italic">p</span> &lt; 0.05 for treatment versus CON. # <span class="html-italic">p</span> &lt; 0.05 vs. UA.</p>
Full article ">Figure 4
<p>Myoblasts were incubated for 24 h in either uric acid (500 µM), β-hydroxybutyrate (BHB; 5 mM), or both (U + B) followed by 10 min of insulin treatment (100 nM) (<span class="html-italic">n</span> = 5). Levels of phosphorylated Akt (pAkt; (<b>A</b>) and glycogen synthase kinase 3β (pGSK3β; (<b>B</b>) were measured compared with no insulin stimulation. * <span class="html-italic">p</span> &lt; 0.05 for treatment vs. CON.</p>
Full article ">
7 pages, 828 KiB  
Brief Report
Association of Inflammasome Gene Expression Levels with Pathogenesis of Familial Mediterranean Fever in Armenians
by Varduhi Hayrapetyan, Lana Karapetyan, Lilit Ghukasyan, Sofi Atshemyan, Hovsep Ghazaryan, Valentina Vardanyan, Vahan Mukuchyan, Arsen Arakelyan and Roksana Zakharyan
Int. J. Mol. Sci. 2024, 25(23), 12958; https://doi.org/10.3390/ijms252312958 - 2 Dec 2024
Viewed by 512
Abstract
Familial Mediterranean fever (FMF) is a genetically determined autoinflammatory disease transmitted mostly by an autosomal recessive mechanism and caused by point mutations of the MEFV (Mediterranean FeVer) gene. The aim of this study was to evaluate the expression of inflammasome genes (p65 [...] Read more.
Familial Mediterranean fever (FMF) is a genetically determined autoinflammatory disease transmitted mostly by an autosomal recessive mechanism and caused by point mutations of the MEFV (Mediterranean FeVer) gene. The aim of this study was to evaluate the expression of inflammasome genes (p65, Casp1, MEFV, and NLRP3) in patients with FMF compared to controls to understand the changes playing a key role in disease development. We found altered expression levels of the full-length MEFV isoform as well as Casp1 and p65 in FMF patients versus controls. This, once again, highlighted the significance of inflammasome genes in terms of FMF. Full article
(This article belongs to the Section Molecular Genetics and Genomics)
Show Figures

Figure 1

Figure 1
<p>mRNA expression levels of <span class="html-italic">p65</span> (<b>a</b>), <span class="html-italic">NLRP3</span> (<b>b</b>), and <span class="html-italic">Casp1</span> (<b>c</b>) in patients with FMF and healthy controls. The horizontal line that splits the box in two represents the median; the lower and upper sides of the box represent the 1st (Q1) and 3rd (Q3) quartiles; and whiskers represent the 1.5 interquartile range from Q1 and Q3.</p>
Full article ">Figure 2
<p>mRNA expression levels of <span class="html-italic">MEFV</span>-f1 (<b>a</b>), <span class="html-italic">MEFV</span>-d2 (<b>b</b>), and <span class="html-italic">MEFV</span> full-length (<b>c</b>) isoforms in patients with FMF and healthy controls. The horizontal lines that splits the box in two represents the median; the lower and upper sides of the box represent the 1st (Q1) and 3rd (Q3) quartiles; and whiskers represent the 1.5 interquartile range from Q1 and Q3.</p>
Full article ">
16 pages, 2423 KiB  
Article
DENV-1 Infection of Macrophages Induces Pyroptosis and Causes Changes in MicroRNA Expression Profiles
by Qinyi Zhang, Sicong Yu, Zhangnv Yang, Xingxing Wang, Jianhua Li, Lingxuan Su, Huijun Zhang, Xiuyu Lou, Haiyan Mao, Yi Sun, Lei Fang, Hao Yan and Yanjun Zhang
Biomedicines 2024, 12(12), 2752; https://doi.org/10.3390/biomedicines12122752 - 30 Nov 2024
Viewed by 607
Abstract
Background: Dengue virus (DENV) is the most widespread mosquito-borne virus, which can cause dengue fever with mild symptoms, or progress to fatal dengue hemorrhagic fever and dengue shock syndrome. As the main target cells of DENV, macrophages are responsible for the innate immune [...] Read more.
Background: Dengue virus (DENV) is the most widespread mosquito-borne virus, which can cause dengue fever with mild symptoms, or progress to fatal dengue hemorrhagic fever and dengue shock syndrome. As the main target cells of DENV, macrophages are responsible for the innate immune response against the virus. Methods: In this study, we investigated the role of pyroptosis in the pathogenic mechanism of dengue fever by examining the level of pyroptosis in DENV-1-infected macrophages and further screened differentially expressed microRNAs by high-throughput sequencing to predict microRNAs that could affect the pyroptosis of the macrophage. Results: Macrophages infected with DENV-1 were induced with decreased cell viability, decreased release of lactate dehydrogenase and IL-1β, activation of NLRP3 inflammasome and caspase-1, cleavage of GSDMD to produce an N-terminal fragment bound to cell membrane, and finally induced macrophage pyroptosis. MicroRNA expression profiles were obtained by sequencing macrophages from all periods of DENV-1 infection and comparing with the negative control. Sixty-three microRNAs differentially expressed in both the early and later stages of infection were also identified. In particular, miR-223-3p, miR-148a-3p, miR-125a-5p, miR-146a-5p and miR-34a-5p were recognized as small molecules that may be involved in the regulation of inflammation. Conclusions: In summary, this study aimed to understand the pathogenic mechanism of DENV through relevant molecular mechanisms and provide new targets for dengue-specific therapy. Full article
(This article belongs to the Special Issue Pathogenic Mechanism and Biosafety of Pathogenic Microorganisms)
Show Figures

Figure 1

Figure 1
<p>Results of macrophage cell viability and LDH release. (<b>A</b>,<b>B</b>) Macrophage cell viability was detected using CCK-8; (<b>C</b>,<b>D</b>) LDH release results were detected by collecting cell supernatants in different culture states. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the negative control group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared between the two groups.</p>
Full article ">Figure 2
<p>Results of macrophage IL-1β release and mRNA and protein expression level. (<b>A</b>,<b>B</b>) Supernatants were collected under different culture conditions, and IL-1β content was detected using an ELISA kit; (<b>C</b>,<b>D</b>) Relative quantification of <span class="html-italic">IL-1β</span> mRNA expression level by RT-qPCR. (<b>E</b>) IL-1β protein expression levels were detected using Western blot, and β-actin was used as an internal reference control. * <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 compared with the negative control group; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared between the two groups.</p>
Full article ">Figure 3
<p>Results of the detection of macrophage pyroptosis-related gene mRNA and its protein expression level. (<b>A</b>–<b>F</b>) Detection results of the expression levels of pyroptosis-related gene mRNA under different culture conditions; (<b>G</b>) The expression levels of pyroptosis-related proteins in macrophages infected with DENV-1 (MOI = 1) for 24 h, 48 h and 72 h, where β-actin was used as an internal reference control. * <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 compared with the negative control group; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared between the two groups.</p>
Full article ">Figure 4
<p>Transcriptome sequencing results and analysis after DENV-1 infection in macrophages. (<b>A</b>) Differentially expressed miRNAs in the infected group and the negative control group at different infection times, where blue indicates downregulation and red indicates upregulation; (<b>B</b>) GO enrichment analysis of differentially expressed miRNA target genes at h.p.i = 72 h, showing the top 10 entries of difference in each classification; (<b>C</b>) KEGG enrichment analysis of differentially expressed miRNA target genes at h.p.i = 72 h, showing the entries in the top 20 differences.</p>
Full article ">Figure 5
<p>Analysis and validation of differentially expressed miRNAs after DENV-1 infection of macrophages. (<b>A</b>) Venn diagram of miRNAs differentially expressed at 24 h, 48 h and 72 h of infection; (<b>B</b>) Heatmap of miRNA clustering for miRNAs that were differentially expressed at all infection periods, with the red colored-frame indicating upregulation and blue colored-frame indicating downregulation; (<b>C</b>) Detection of the expression levels of <span class="html-italic">miR-223-3p</span>, <span class="html-italic">miR-148a-3p</span>, <span class="html-italic">miR-125a-5p</span>, <span class="html-italic">miR-146a-5p</span> and <span class="html-italic">miR-34a-5p</span> by RT-qPCR in macrophages at all infection periods. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the negative control group.</p>
Full article ">
15 pages, 10237 KiB  
Article
Irisin Attenuates Neuroinflammation Targeting the NLRP3 Inflammasome
by Francesca Martina Filannino, Melania Ruggiero, Maria Antonietta Panaro, Dario Domenico Lofrumento, Teresa Trotta, Tarek Benameur, Antonia Cianciulli, Rosa Calvello, Federico Zoila and Chiara Porro
Molecules 2024, 29(23), 5623; https://doi.org/10.3390/molecules29235623 - 28 Nov 2024
Viewed by 754
Abstract
Neuroinflammation is defined as an immune response involving various cell types, particularly microglia, which monitor the neuroimmune axis. Microglia activate in two distinct ways: M1, which is pro-inflammatory and capable of inducing phagocytosis and releasing pro-inflammatory factors, and M2, which has anti-inflammatory properties. [...] Read more.
Neuroinflammation is defined as an immune response involving various cell types, particularly microglia, which monitor the neuroimmune axis. Microglia activate in two distinct ways: M1, which is pro-inflammatory and capable of inducing phagocytosis and releasing pro-inflammatory factors, and M2, which has anti-inflammatory properties. Inflammasomes are large protein complexes that form in response to internal danger signals, activating caspase-1 and leading to the release of pro-inflammatory cytokines such as interleukin 1β. Irisin, a peptide primarily released by muscles during exercise, was examined for its effects on BV2 microglial cells in vitro. Even at low concentrations, irisin was observed to influence the NLRP3 inflammasome, showing potential as a neuroprotective and anti-inflammatory agent after stimulation with lipopolysaccharides (LPSs). Irisin helped maintain microglia in their typical physiological state and reduced their migratory capacity. Irisin also increased Arg-1 protein expression, a marker of M2 polarization, while downregulating NLRP3, Pycard, caspase-1, IL-1β, and CD14. The results of this study indicate that irisin may serve as a crucial mediator of neuroprotection, thus representing an innovative tool for the prevention of neurodegenerative diseases. Full article
(This article belongs to the Section Bioorganic Chemistry)
Show Figures

Figure 1

Figure 1
<p>Effect of irisin on cell viability (MTT assay). BV-2 cells were incubated with irisin at concentrations spanning a dose–response curve, ranging from 5 nM to 20 nM (<b>A</b>). Irisin at a concentration of 5 nM was used to treat cells, either in the absence or presence of 1 µg/mL LPSs (<b>B</b>). Data are reported as percentages compared to control values and are expressed as means ± SDs. ** <span class="html-italic">p</span> &lt; 0.01 compared to the control. *** <span class="html-italic">p</span> &lt; 0.001 compared to the control.</p>
Full article ">Figure 2
<p>Morphological analysis of the BV2 cells following the administration of irisin, either alone or after LPS stimulation. The morphological analysis was conducted on BV2 cells in the control condition (<b>A</b>) and following the administration of 1 µg/mL LPSs (<b>B</b>), 5 nM irisin (<b>C</b>), or 5 nM irisin in the presence of 1 µg/mL LPSs (<b>D</b>). The scale bar is 100 µm (10× objective). The arrows in the images indicate cells that have undergone a morphological change. The cell areas (µm<sup>2</sup>) were quantified using the ImageJ 1.8.0 software, which was bound with Java 8 64-bit (<b>E</b>). The data are expressed as means ± standard deviations. A significant difference can be observed between the control and LPS groups (*** <span class="html-italic">p</span> &lt; 0.001), as well as between the IRISIN + LPS and LPS groups (### <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>The analysis of the migratory capacity of microglia after the administration of irisin in the presence or absence of LPSs. A wound was generated in a sub-confluent layer of BV2 cells, and the resulting space was captured at the wound site 0 and 24 h after the treatment: (<b>A</b>) BV2 cells at time 0, (<b>B</b>) 24 h after the cut in the control condition, (<b>C</b>) treated with 1 µg/mL LPSs, (<b>D</b>) with 5 nM irisin, and (<b>E</b>) with 5 nM irisin in the presence of 1 µg/mL LPSs. The images are representative of an experiment with three independent replicates. The percentage of the wound gap was analyzed using the ImageJ software and subsequently plotted and statistically analyzed as the percentage of wound closure compared to the 0 time condition (<b>F</b>). The values are presented as means ± standard deviations. Bar: 75 µm (20× objective). ** <span class="html-italic">p</span> &lt; 0.01 compared to the control. # <span class="html-italic">p</span> &lt; 0.05 compared to the LPS condition.</p>
Full article ">Figure 4
<p>Evaluation of NLRP3 and Pycard expression following the administration of irisin, with or without LPSs. Western blotting detection and densitometric analysis of the expression of the pro-inflammatory NLRP3 (<b>A</b>) and Pycard (<b>B</b>) in control cells (CTR), BV2 cells treated with irisin, BV2 cells treated with LPSs (LPS), and BV2 cells treated with irisin + LPSs. The protein expression values are expressed in arbitrary units after normalization against β-actin. The data are presented as means ± SDs (* <span class="html-italic">p</span> &lt; 0.05 vs. CTR; *** <span class="html-italic">p</span> &lt; 0.001 compared to the control; and ### <span class="html-italic">p</span> &lt; 0.001 compared to the condition of LPS-stimulated microglia).</p>
Full article ">Figure 5
<p>Evaluation of caspase-1, IL1Beta, and CD-14 expression following the administration of irisin, with or without LPSs. Western blotting detection and densitometric analysis of the expression of pro-inflammatory caspase-1 (<b>A</b>), IL1B (<b>B</b>), and CD-14 (<b>C</b>) in control cells (CTR), BV2 cells treated with irisin, BV2 cells treated with LPSs (LPS), and BV2 cells treated with irisin + LPSs (*** <span class="html-italic">p</span> &lt; 0.001 compared to the control condition; and ### <span class="html-italic">p</span> &lt; 0.001 compared to the condition of LPS-stimulated cells).</p>
Full article ">Figure 6
<p>Evaluation of Arginase 1 expression following the administration of irisin, with or without LPSs. Western blotting detection and densitometric analysis of the expression of the anti-inflammatory agent in control cells (CTR), BV2 cells treated with LPSs (LPS), BV2 cells treated with irisin, and BV2 cells treated with irisin + LPSs (*** <span class="html-italic">p</span> &lt; 0.001 compared to the control condition; and ### <span class="html-italic">p</span> &lt; 0.001 compared to the condition of LPS-stimulated cells).</p>
Full article ">
19 pages, 11411 KiB  
Article
Ginsenoside Rg1 Alleviates Blood–Milk Barrier Disruption in Subclinical Bovine Mastitis by Regulating Oxidative Stress-Induced Excessive Autophagy
by Shanshan Yang, Zihao Fang, Hongwei Duan, Weitao Dong and Longfei Xiao
Antioxidants 2024, 13(12), 1446; https://doi.org/10.3390/antiox13121446 - 24 Nov 2024
Viewed by 570
Abstract
As a critical disease usually infected by Staphylococcus aureus, with a worldwide effect on dairy animals, subclinical mastitis is characterized by persistence and treatment resistance. During mastitis, the blood–milk barrier (BMB)’s integrity is impaired, resulting in pathogen invasion and milk quality decline. [...] Read more.
As a critical disease usually infected by Staphylococcus aureus, with a worldwide effect on dairy animals, subclinical mastitis is characterized by persistence and treatment resistance. During mastitis, the blood–milk barrier (BMB)’s integrity is impaired, resulting in pathogen invasion and milk quality decline. In this study, it was found that ginsenoside Rg1 (Rg1), a natural anti-inflammatory and antioxidant compound derived from ginseng, inhibited the onset of tight junction (TJ) dysfunction and ameliorated lipoteichoic acid (LTA)-induced BMB disruption inside and outside the organisms. According to subsequent mechanistic studies, Rg1 inhibited excessive autophagy and inactivated the NLRP3 inflammasome by blockading ROS generation, thereby alleviating TJ dysfunction. Peroxisome proliferator-activated receptor gamma (PPARγ) was identified as a potential target of Rg1 by means of molecular docking plus network pharmacology analysis. Furthermore, it was demonstrated that Rg1 inhibited the oxidative stress levels by activating PPARγ, and regulating the upstream autophagy-related AMPK/mTOR signaling pathway, thus decreasing excessive in vivo and in vitro autophagy. The ROS/autophagy/NLRP3 inflammasome axis was identified as a promising target for treating subclinical bovine mastitis in this study. In conclusion, Rg1 is proven to alleviate BMB disruption by activating PPARγ to inhibit oxidative stress and subsequent excessive autophagy in the case of subclinical bovine mastitis. Full article
Show Figures

Figure 1

Figure 1
<p>Excessive autophagy and BMB disruption in mammary gland with subclinical bovine mastitis. (<b>A</b>) HE staining for histological changes in mammary glands. Original magnification = 200×, scale bar = 100 μm (n = 3). (<b>B</b>) Ultrastructure of autophagy and TJs in bMECs observed by TEM (n = 3). Original magnification = 10,000×, scale bar = 500 nm. The red arrowheads indicated TJs, the red arrows indicated autophagic structures. (<b>C</b>) Typical immunofluorescence images for localization of LC3 (green) in the mammary glands. Original magnification = 200×, scale bar = 100 μm (n = 3). (<b>D</b>) Typical immunofluorescence images for localization of claudin-1, ZO-1, and occludin (green) in the mammary glands. Original magnification = 200×, scale bar = 100 μm (n = 3). (<b>E</b>) The relative protein expressions of claudin-1, beclin-1, occludin, ZO-1 and LC3 in mammary glands detected by Western blotting (n = 3). The loading control is set as β-actin, and mean ± SD is used for value expression. ** <span class="html-italic">p</span> &lt; 0.01 vs. the healthy group.</p>
Full article ">Figure 2
<p>Rg1 inhibited LTA-induced excessive autophagy to alleviate TJ dysfunction. (<b>A</b>) MAC-T cells processed for 24 h by LTA at 0.1, 1 and 10 μg/mL. Western blotting for proteins associated with TJs (ZO-1, occludin, and claudin-1). (<b>B</b>) MAC-T cells undergoing 24 h LTA (10 μg/mL) and/or Rg1 (at required concentrations) treatment. Western blotting for proteins in relation to autophagy and TJs. (<b>C</b>) MAC-T cells subjected to 24 h of LTA (10 μg/mL) and/or Rg1 (20 μM) processing. After the transfection of MAC-T cells with adenovirus plus autolysosome quantitation via mCherry-GFP-LC3, autophagy was visually observable. Original magnification = 400×. (<b>D</b>) MAC-T cells under 24 h of treatment by LTA (10 μg/mL), Rg1 (20 μM), and/or CQ (50 μM). Western blotting for proteins correlated with autophagy and TJs. The mean ± SD is used for value presentation; compared with the control group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; compared to the LTA treatment group, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Rg1 inhibited LTA-induced TJ dysfunction by blocking the ROS/autophagy/NLRP3 inflammasome axis. (<b>A</b>) SOD, MDA, and CAT content in subclinical bovine mastitis and healthy mammary tissues detected (n = 4). MAC-T cells that had undergone treatment by LTA (10 μg/mL), Rg1 (20 μM), and/or NAC (5 mM) for 24 h. (<b>B</b>) DCFH-DA probe for ROS level evaluation in MAC-T cells (original magnification = 400×, scale bar = 50 μm.). (<b>C</b>) Quantification of MAC-T cells for ROS content (n = 6). (<b>D</b>) Calculated GSH level in MAC-T cells (n = 6). (<b>E</b>) Western blotting for ZO-1, occludin, and claudin-1 (proteins related to TJs). (<b>F</b>) Relative protein expressions concerning ASC, IL-1β, NLRP3, and caspase-1 in mammary glands determined by means of Western blotting. (<b>G</b>) Western blotting for the TJ-related and NLRP3 inflammasome-related proteins. (<b>H</b>) MAC-T cells subjected to LTA (10 μg/mL), Rg1 (20 μM), and/or MCC950 (10 μM) processing for 24 h. Western blotting for the NLRP3 inflammasome-related and TJ-related proteins. (<b>I</b>) MAC-T cells under LTA (10 μg/mL), Rg1 (20 μM), and/or CQ (50 μM) treatment for 24 h. Western blotting for NLRP3 and IL-1β proteins. (<b>J</b>) Twenty-four hour treatment for MAC-T cells using LTA (10 μg/mL), and/or MCC950 (10 μM). Western blotting for beclin-1 and LC3 proteins. The mean ± SD is the expression format for values; compared with the control group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; compared to the LTA treatment group, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Rg1 inhibited LTA-induced excessive autophagy by regulating the ROS/AMPK/mTOR signaling pathway. (<b>A</b>) Relative protein expressions of mTOR, AMPK, p-AMPK, and p-mTOR in mammary glands determined by means of Western blotting (n = 3). (<b>B</b>) Rg1 (20 μM), LTA (10 μg/mL), and/or NAC (5 mM) added to treat MAC-T cells for 24 h. Western blotting for AMPK, mTOR, p-AMPK, and p-mTOR protein. (<b>C</b>) MAC-T cells subjected to 24 h of LTA (10 μg/mL), Rg1 (20 μM), and/or CC (10 μM) treatment. Protein expressions of AMPK, mTOR, beclin-1, p-mTOR, LC3, and p-AMPK detected by Western blotting. The mean ± SD is adopted to describe values; compared to the control group, ** <span class="html-italic">p</span> &lt; 0.01; compared to the LTA treatment group, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Repressed oxidative stress plus raised PPARγ are necessary for Rg1 to inhibit LTA-induced excessive autophagy and TJ recovery. (<b>A</b>) Number of intersecting NAFLD, mastitis and R&amp;M target genes. (<b>B</b>) Relative expressions of PPARα, PPARβ/δ, and PPARγ proteins in mammary glands determined by means of Western blotting (n = 3). (<b>C</b>) MAC-T cells subjected to 24 h of LTA (10 μg/mL) and/or Rg1 (20 μM) treatment. Western blotting for PPARα, PPARβ/δ, and PPARγ protein. (<b>D</b>) MAC-T cells undergoing 24 h disposal by LTA (10 μg/mL), Rg1 (20 μM), and/or GW6471 (10 μM). Western blotting for PPARα and TJ-related proteins. (<b>E</b>) MAC-T cells under 24 h of treatment using LTA (10 μg/mL), Rg1 (20 μM), and/or T0070907 (10 μM). Western blotting for PPARγ and TJ-related proteins. (<b>F</b>) Docking of PPARγ with Rg1. Rg1 interacts with 2 amino acids, THR 440 and GLN 437, adjacent to the PPARγ active site, forming crucial binding forces in the periphery of the active site. (<b>G</b>) ROS level quantification in MAC-T cells (n = 6). (<b>H</b>) GSH level in MAC-T cells was calculated (n = 6). (<b>I</b>) Western blotting for autophagy-related plus NLRP3 inflammasome-related proteins like AMPK, mTOR, p-AMPK, and p-mTOR. The mean ± SD is used to express values; compared with the control group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; compared with the LTA treatment group, ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Rg1 alleviated BMB disruption through regulating autophagy signaling in organisms. Five groups were set up for the mice: control group (no treatment with LTA), Rg1 (no LTA, 200 mg/kg) group, model group (LTA injection, no treatment), Rg1 (200 mg/kg) + LTA treatment group, and rosiglitazone (10 mg/kg) + LTA treatment group. (<b>A</b>) Hematoxylin-eosin (HE) staining for general examination of mammary glands (original magnification = 200×, scale bar = 100 μm), together with immunofluorescence images for LC3, ZO-1, occludin and claudin-1 (original magnification = 400×, scale bar = 50 μm). (<b>B</b>) Examined variations in SOD, MDA, and CAT content. (<b>C</b>,<b>D</b>) Western blotting for determining the relative protein expressions of AMPK, PPARγ, p-AMPK, mTOR, p-mTOR, autophagy-related, NLRP3 inflammasome-related and TJ-related proteins. The mean ± SD as the value format and β-actin as the loading control. Compared with the control group, ** <span class="html-italic">p</span> &lt; 0.01; Compared with the LTA treatment group, and ## <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>Proposed mechanism of Rg1 in alleviating BMB disruption in cases of subclinical bovine mastitis by regulating oxidative stress-induced excessive autophagy.</p>
Full article ">
Back to TopTop