www.fgks.org   »   [go: up one dir, main page]

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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
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 (1,490)

Search Parameters:
Keywords = inflammasome

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 338 KiB  
Review
Emerging Therapeutic Potential of Fluoxetine on Cognitive Decline in Alzheimer’s Disease: Systematic Review
by Anastasia Bougea, Efthalia Angelopoulou, Efthimios Vasilopoulos, Philippos Gourzis and Sokratis Papageorgiou
Int. J. Mol. Sci. 2024, 25(12), 6542; https://doi.org/10.3390/ijms25126542 (registering DOI) - 13 Jun 2024
Abstract
Fluoxetine, a commonly prescribed medication for depression, has been studied in Alzheimer’s disease (AD) patients for its effectiveness on cognitive symptoms. The aim of this systematic review is to investigate the therapeutic potential of fluoxetine in cognitive decline in AD, focusing on its [...] Read more.
Fluoxetine, a commonly prescribed medication for depression, has been studied in Alzheimer’s disease (AD) patients for its effectiveness on cognitive symptoms. The aim of this systematic review is to investigate the therapeutic potential of fluoxetine in cognitive decline in AD, focusing on its anti-degenerative mechanisms of action and clinical implications. According to PRISMA, we searched MEDLINE, up to 1 April 2024, for animal and human studies examining the efficacy of fluoxetine with regard to the recovery of cognitive function in AD. Methodological quality was evaluated using the ARRIVE tool for animal AD studies and the Cochrane tool for clinical trials. In total, 22 studies were analyzed (19 animal AD studies and 3 clinical studies). Fluoxetine promoted neurogenesis and enhanced synaptic plasticity in preclinical models of AD, through a decrease in Aβ pathology and increase in BDNF, by activating diverse pathways (such as the DAF-16-mediated, TGF-beta1, ILK-AKT-GSK3beta, and CREB/p-CREB/BDNF). In addition, fluoxetine has anti-inflammatory properties/antioxidant effects via targeting antioxidant Nrf2/HO-1 and hindering TLR4/NLRP3 inflammasome. Only three clinical studies showed that fluoxetine ameliorated the cognitive performance of people with AD; however, several methodological issues limited the generalizability of these results. Overall, the high-quality preclinical evidence suggests that fluoxetine may have neuroprotective, antioxidant, and anti-inflammatory effects in AD animal models. While more high-quality clinical research is needed to fully understand the mechanisms underlying these effects, fluoxetine is a promising potential treatment for AD patients. If future clinical trials confirm its anti-degenerative and neuroprotective effects, fluoxetine could offer a new therapeutic approach for slowing down the progression of AD. Full article
(This article belongs to the Special Issue Latest Review Papers in Molecular Neurobiology 2024)
21 pages, 923 KiB  
Review
CARD8: A Novel Inflammasome Sensor with Well-Known Anti-Inflammatory and Anti-Apoptotic Activity
by Tugay Karakaya, Marta Slaufova, Michela Di Filippo, Paulina Hennig, Thomas Kündig and Hans-Dietmar Beer
Cells 2024, 13(12), 1032; https://doi.org/10.3390/cells13121032 - 13 Jun 2024
Viewed by 62
Abstract
Inflammasomes comprise a group of protein complexes with fundamental roles in the induction of inflammation. Upon sensing stress factors, their assembly induces the activation and release of the pro-inflammatory cytokines interleukin (IL)-1β and -18 and a lytic type of cell death, termed pyroptosis. [...] Read more.
Inflammasomes comprise a group of protein complexes with fundamental roles in the induction of inflammation. Upon sensing stress factors, their assembly induces the activation and release of the pro-inflammatory cytokines interleukin (IL)-1β and -18 and a lytic type of cell death, termed pyroptosis. Recently, CARD8 has joined the group of inflammasome sensors. The carboxy-terminal part of CARD8, consisting of a function-to-find-domain (FIIND) and a caspase activation and recruitment domain (CARD), resembles that of NLR family pyrin domain containing 1 (NLRP1), which is recognized as the main inflammasome sensor in human keratinocytes. The interaction with dipeptidyl peptidases 8 and 9 (DPP8/9) represents an activation checkpoint for both sensors. CARD8 and NLRP1 are activated by viral protease activity targeting their amino-terminal region. However, CARD8 also has some unique features compared to the established inflammasome sensors. Activation of CARD8 occurs independently of the inflammasome adaptor protein apoptosis-associated speck-like protein containing a CARD (ASC), leading mainly to pyroptosis rather than the activation and secretion of pro-inflammatory cytokines. CARD8 was also shown to have anti-inflammatory and anti-apoptotic activity. It interacts with, and inhibits, several proteins involved in inflammation and cell death, such as the inflammasome sensor NLRP3, CARD-containing proteins caspase-1 and -9, nucleotide-binding oligomerization domain containing 2 (NOD2), or nuclear factor kappa B (NF-κB). Single nucleotide polymorphisms (SNPs) of CARD8, some of them occurring at high frequencies, are associated with various inflammatory diseases. The molecular mechanisms underlying the different pro- and anti-inflammatory activities of CARD8 are incompletely understood. Alternative splicing leads to the generation of multiple CARD8 protein isoforms. Although the functional properties of these isoforms are poorly characterized, there is evidence that suggests isoform-specific roles. The characterization of the functions of these isoforms, together with their cell- and disease-specific expression, might be the key to a better understanding of CARD8’s different roles in inflammation and inflammatory diseases. Full article
(This article belongs to the Special Issue Role of Inflammasome Activation in Innate and Adaptive Immunity)
14 pages, 959 KiB  
Article
Evaluating Single-Nucleotide Polymorphisms in Inflammasome Proteins and Serum Levels of IL-18 and IL-1β in Kidney Interstitial Damage in Anti-Neutrophilic Cytoplasmic Antibody-Associated Vasculitis
by Laura Martinez Valenzuela, Anna Vidal-Alabró, Belén Rubio, Paula Antón-Pàmpols, Francisco Gómez-Preciado, Xavier Fulladosa, Josep Maria Cruzado, Juan Torras, Nuria Lloberas and Juliana Draibe
Int. J. Mol. Sci. 2024, 25(12), 6479; https://doi.org/10.3390/ijms25126479 - 12 Jun 2024
Viewed by 133
Abstract
The inflammasome regulates the innate inflammatory response and is involved in autoimmune diseases. In this study, we explored the levels of IL-18 and IL-1β in serum and urine and the influence of various single-nucleotide polymorphisms (SNPs) on kidney lesions at diagnosis in patients [...] Read more.
The inflammasome regulates the innate inflammatory response and is involved in autoimmune diseases. In this study, we explored the levels of IL-18 and IL-1β in serum and urine and the influence of various single-nucleotide polymorphisms (SNPs) on kidney lesions at diagnosis in patients with ANCA-associated vasculitis (AAV) and their clinical outcomes. Ninety-two patients with renal AAV were recruited, and blood and urine were collected at diagnosis. Serum and urine cytokine levels were measured by ELISA. DNA was extracted and genotyped using TaqMan assays for SNPs in several inflammasome genes. Lower serum IL-18 (p = 0.049) and the IL-18 rs187238 G-carrier genotype (p = 0.042) were associated with severe fibrosis. The IL-18 rs1946518 TT genotype was associated with an increased risk of relapse (p = 0.05), whereas GG was related to better renal outcomes (p = 0.031). The rs187238 GG genotype was identified as a risk factor for mortality within the first year after AAV diagnosis, independent of the requirement for dialysis or lung involvement (p = 0.013). We suggest that decreased cytokine levels could be a surrogate marker of scarring and chronicity of the renal lesions, together with the rs187238 GG genotype. If our results are validated, the rs1946518 TT genotype predicts the risk of relapse and renal outcomes during follow-up. Full article
(This article belongs to the Special Issue New Advances in Inflammasomes)
Show Figures

Figure 1

Figure 1
<p>Levels of IL-18 in serum according to the rs187238 and rs1946518 genotypes.</p>
Full article ">Figure 2
<p>Kaplan–Meier survival curve according to the <span class="html-italic">IL-18</span> rs187238 genotype.</p>
Full article ">
32 pages, 1440 KiB  
Review
Mitochondrial Reactive Oxygen Species in Infection and Immunity
by Arunima Mukherjee, Krishna Kanta Ghosh, Sabyasachi Chakrabortty, Balázs Gulyás, Parasuraman Padmanabhan and Writoban Basu Ball
Biomolecules 2024, 14(6), 670; https://doi.org/10.3390/biom14060670 - 8 Jun 2024
Viewed by 371
Abstract
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including [...] Read more.
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including ultra-violet (UV) radiation, electron leakage during aerobic respiration, inflammatory responses mediated by macrophages, and other external stimuli or stress. The enhanced production of ROS is termed oxidative stress and this leads to cellular damage, such as protein carbonylation, lipid peroxidation, deoxyribonucleic acid (DNA) damage, and base modifications. This damage may manifest in various pathological states, including ageing, cancer, neurological diseases, and metabolic disorders like diabetes. On the other hand, the optimum levels of ROS have been implicated in the regulation of many important physiological processes. For example, the ROS generated in the mitochondria (mitochondrial ROS or mt-ROS), as a byproduct of the electron transport chain (ETC), participate in a plethora of physiological functions, which include ageing, cell growth, cell proliferation, and immune response and regulation. In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection by bacteria, protozoan parasites, viruses, and fungi. We will also discuss how these pathogens, in turn, modulate mt-ROS to evade host immunity. We will conclude by briefly giving an overview of the potential therapeutic approaches involving mt-ROS in infectious diseases. Full article
(This article belongs to the Special Issue Mitochondrial ROS in Health and Disease)
18 pages, 8310 KiB  
Protocol
A Dynamic Protocol to Explore NLRP3 Inflammasome Activation in Cerebral Organoids
by Dana El Soufi El Sabbagh, Liliana Attisano, Ana Cristina Andreazza and Alencar Kolinski Machado
Int. J. Mol. Sci. 2024, 25(12), 6335; https://doi.org/10.3390/ijms25126335 - 7 Jun 2024
Viewed by 272
Abstract
The NLRP3 inflammasome plays a crucial role in the inflammatory response, reacting to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). This response is essential for combating infections and restoring tissue homeostasis. However, chronic activation can lead to detrimental effects, particularly in [...] Read more.
The NLRP3 inflammasome plays a crucial role in the inflammatory response, reacting to pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). This response is essential for combating infections and restoring tissue homeostasis. However, chronic activation can lead to detrimental effects, particularly in neuropsychiatric and neurodegenerative diseases. Our study seeks to provide a method to effectively measure the NLRP3 inflammasome’s activation within cerebral organoids (COs), providing insights into the underlying pathophysiology of these conditions and enabling future studies to investigate the development of targeted therapies. Full article
(This article belongs to the Special Issue Research in iPSC-Based Disease Models)
Show Figures

Figure 1

Figure 1
<p>Overview of CO generation protocol and long-term growth. ESCs or iPSCs are singularized and plated to form embryo bodies and then grown into a neural induction phase. Once the 2D spheres are intact, organoids are embedded in Matrigel to form 3D structures and then excised and placed in rotating cultures that grow into uniform spheroids. At the 1-month time point, they primarily contain radial glia cells, while at 4 months and 6 months, astrocyte progenitors emerge. After 6 months, COs contain mature deep-layer and upper-layer neurons, mature astrocytes.</p>
Full article ">Figure 2
<p>Overview of the NLRP3 inflammasome. Step 1 is a priming process, and step 2 involves the activation of the inflammasome which results in increased inflammatory cytokine production.</p>
Full article ">Figure 3
<p>The experimental scheme of the time-response curve in CO slices in groups I, II, and III. After the LPS-priming stimulus and MCC950 exposures, group I was treated with nigericin for 1 h, group II for 4 h, and group III for 16 h. Slices were then fixed and immunostained, while the supernatant was collected for various assays of inflammatory activation.</p>
Full article ">Figure 4
<p>Time-response curve of the LPS + Nigericin protocol using groups I, II, and III. (<b>A</b>) Group I: 3 h LPS treatment followed by 1 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 1 h Nigericin treatment (<b>bottom</b>). (<b>B</b>) Group II: 3 h LPS treatment followed by 4 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 4 h Nigericin treatment (<b>bottom</b>). (<b>C</b>) Group III: 3 h LPS treatment followed by 16 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 16 h Nigericin treatment (<b>bottom</b>). Slices were stained with SOX2 (neural progenitor cells), GFAP (astrocytes), and ASC (ASC speck formation in the NLRP3). Scale bar = 100 µm. (<b>D</b>) By counting cells with HALO and ASC specks with ImageJ. Results are shown as a ratio of cells expressing ASC specks in each group. Data were analyzed by two-way ANOVA with Sidak’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) most cells containing ASC specks in group II were found to be double positive ASC+/GFAP+.</p>
Full article ">Figure 4 Cont.
<p>Time-response curve of the LPS + Nigericin protocol using groups I, II, and III. (<b>A</b>) Group I: 3 h LPS treatment followed by 1 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 1 h Nigericin treatment (<b>bottom</b>). (<b>B</b>) Group II: 3 h LPS treatment followed by 4 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 4 h Nigericin treatment (<b>bottom</b>). (<b>C</b>) Group III: 3 h LPS treatment followed by 16 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 16 h Nigericin treatment (<b>bottom</b>). Slices were stained with SOX2 (neural progenitor cells), GFAP (astrocytes), and ASC (ASC speck formation in the NLRP3). Scale bar = 100 µm. (<b>D</b>) By counting cells with HALO and ASC specks with ImageJ. Results are shown as a ratio of cells expressing ASC specks in each group. Data were analyzed by two-way ANOVA with Sidak’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) most cells containing ASC specks in group II were found to be double positive ASC+/GFAP+.</p>
Full article ">Figure 4 Cont.
<p>Time-response curve of the LPS + Nigericin protocol using groups I, II, and III. (<b>A</b>) Group I: 3 h LPS treatment followed by 1 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 1 h Nigericin treatment (<b>bottom</b>). (<b>B</b>) Group II: 3 h LPS treatment followed by 4 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 4 h Nigericin treatment (<b>bottom</b>). (<b>C</b>) Group III: 3 h LPS treatment followed by 16 h Nigericin treatment (<b>top</b>). 3 h LPS treatment, 2 h MCC950 pre-treatment, and 16 h Nigericin treatment (<b>bottom</b>). Slices were stained with SOX2 (neural progenitor cells), GFAP (astrocytes), and ASC (ASC speck formation in the NLRP3). Scale bar = 100 µm. (<b>D</b>) By counting cells with HALO and ASC specks with ImageJ. Results are shown as a ratio of cells expressing ASC specks in each group. Data were analyzed by two-way ANOVA with Sidak’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05. (<b>E</b>) most cells containing ASC specks in group II were found to be double positive ASC+/GFAP+.</p>
Full article ">Figure 5
<p>Method reproducibility for group II: Locating the ASC specks in astrocytes. (<b>A</b>) (<b>i</b>) Control slice. (<b>ii</b>) LPS (3 h) + Nigericin (4 h). (<b>iii</b>) LPS (3 h) + MCC950 (2 h) + Nigericin (4 h). Staining performed using MAP2 (neurons) GFAP (astrocytes) ASC (ASC activation in NLRP3). Scale bar = 10 µm. (<b>B</b>) Images quantified for ASC specks using HALO and ImageJ. Data were analyzed by two-way ANOVA with Sidak’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05 CTL vs. L + N and # <span class="html-italic">p</span> &lt; 0.05 L + N vs. L + M + N.</p>
Full article ">Figure 5 Cont.
<p>Method reproducibility for group II: Locating the ASC specks in astrocytes. (<b>A</b>) (<b>i</b>) Control slice. (<b>ii</b>) LPS (3 h) + Nigericin (4 h). (<b>iii</b>) LPS (3 h) + MCC950 (2 h) + Nigericin (4 h). Staining performed using MAP2 (neurons) GFAP (astrocytes) ASC (ASC activation in NLRP3). Scale bar = 10 µm. (<b>B</b>) Images quantified for ASC specks using HALO and ImageJ. Data were analyzed by two-way ANOVA with Sidak’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05 CTL vs. L + N and # <span class="html-italic">p</span> &lt; 0.05 L + N vs. L + M + N.</p>
Full article ">Figure 6
<p>Validation on NLRP3 inflammasome endpoints of group II. (<b>A</b>) Measurement of ccf mtDNA; (<b>B</b>) IL-1β protein expression analysis; (<b>C</b>) Caspase-1 protein expression; (<b>D</b>) Indirect measurement of NO levels; (<b>E</b>) Semi-quantitative evaluation of ROS levels. (<b>F</b>) Measurement of dsDNA release. Data in (<b>A</b>–<b>F</b>) are expressed as means and standard deviation of three technical replicates of a single experiment. Statistical analyses were performed using a One-way ANOVA to examine differences followed by Tukey’s multiple comparison test (* <span class="html-italic">p</span> &lt; 0.005 CTL vs. L + N and # <span class="html-italic">p</span> &lt; 0.005 L + N vs. L + M + N).</p>
Full article ">
21 pages, 2966 KiB  
Article
Elevated NLRP3 Inflammasome Activation Is Associated with Motor Neuron Degeneration in ALS
by Hilal Cihankaya, Verian Bader, Konstanze F. Winklhofer, Matthias Vorgerd, Johann Matschke, Sarah Stahlke, Carsten Theiss and Veronika Matschke
Cells 2024, 13(12), 995; https://doi.org/10.3390/cells13120995 - 7 Jun 2024
Viewed by 345
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron degeneration in the central nervous system. Recent research has increasingly linked the activation of nucleotide oligomerization domain-like receptor protein 3 (NLRP3) inflammasome to ALS pathogenesis. NLRP3 activation triggers Caspase 1 [...] Read more.
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by motor neuron degeneration in the central nervous system. Recent research has increasingly linked the activation of nucleotide oligomerization domain-like receptor protein 3 (NLRP3) inflammasome to ALS pathogenesis. NLRP3 activation triggers Caspase 1 (CASP 1) auto-activation, leading to the cleavage of Gasdermin D (GSDMD) and pore formation on the cellular membrane. This process facilitates cytokine secretion and ultimately results in pyroptotic cell death, highlighting the complex interplay of inflammation and neurodegeneration in ALS. This study aimed to characterize the NLRP3 inflammasome components and their colocalization with cellular markers using the wobbler mouse as an ALS animal model. Firstly, we checked the levels of miR-223-3p because of its association with NLRP3 inflammasome activity. The wobbler mice showed an increased expression of miR-223-3p in the ventral horn, spinal cord, and cerebellum tissues. Next, increased levels of NLRP3, pro-CASP 1, cleaved CASP 1 (c-CASP 1), full-length GSDMD, and cleaved GDSMD revealed NLRP3 inflammasome activation in wobbler spinal cords, but not in the cerebellum. Furthermore, we investigated the colocalization of the aforementioned proteins with neurons, microglia, and astrocyte markers in the spinal cord tissue. Evidently, the wobbler mice displayed microgliosis, astrogliosis, and motor neuron degeneration in this tissue. Additionally, we showed the upregulation of protein levels and the colocalization of NLRP3, c-CASP1, and GSDMD in neurons, as well as in microglia and astrocytes. Overall, this study demonstrated the involvement of NLRP3 inflammasome activation and pyroptotic cell death in the spinal cord tissue of wobbler mice, which could further exacerbate the motor neuron degeneration and neuroinflammation in this ALS mouse model. Full article
(This article belongs to the Special Issue Molecular Insights into Neurodegenerative Diseases)
Show Figures

Figure 1

Figure 1
<p>Wobbler mice demonstrated elevated levels of miR-223-3p in laser-microdissected ventral horn, cervical spinal cord, and cerebellum. (<b>A</b>) Image of cresyl-violet-stained spinal cord section. Gray matter can be seen as dark purple, and white matter can be seen as light purple/pink. Blunt-ended lines show ventral and dorsal horns. Arrows show individual motor neurons. Scale: 400 µm. Close-up image of cresyl-violet-stained gray matter of ventral horn (<b>B</b>) before and (<b>C</b>) after LMD. Scale: 200 µm. RT-qPCR analysis of miR-223-3p using samples from (<b>D</b>) laser-microdissected ventral horn, (<b>E</b>) cervical spinal cord, and (<b>F</b>) cerebellum of p40 wild-type and wobbler mice. The differences in the expression levels were calculated using the 2<sup>−ΔΔCt</sup> method with endogenous normalization to UniSp6. Data are presented as mean ± SD, n = 4–8. *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 2
<p>Increased levels of NLRP3, pro-CASP 1, c-CASP 1, GSDMD-FL, C-GSDMD, IL-1β, and IL-18 proteins revealed NLRP3 inflammasome activation and thus pyroptotic cell death in wobbler spinal cords, but not in the cerebellum, despite increased NLRP3 expression. (<b>A</b>) Western blot image and semi-quantitative analyses of (<b>B</b>) NLRP3, (<b>C</b>) pro-CASP 1, (<b>D</b>) c-CASP 1, (<b>E</b>) GSDMD-FL, (<b>F</b>) C-GSDMD, (<b>G</b>) IL-1β, and (<b>H</b>) IL-18 in cervical spinal cord tissues of p40 wild-type and wobbler mice. (<b>I</b>) Western blot image and quantitative analyses of (<b>J</b>) NLRP3, (<b>K</b>) pro-CASP 1, (<b>L</b>) c-CASP 1, and (<b>M</b>) C-GSDMD in cerebellum tissues of p40 wild-type and wobbler mice. Calnexin or actin was used as a loading control. Data are presented as mean ± SD, n varies between 4 and 12 per group. ns: not significant. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Representative images of double immunofluorescence staining for NLRP3 with NeuN, Iba1, or GFAP in the cervical spinal cord tissues of p40 wild-type and wobbler mice. NLRP3 was predominantly expressed in wobbler NeuN<sup>+</sup> cells (motor neuronal cells), as well as in Iba1<sup>+</sup> (microglia) and GFAP<sup>+</sup> (astrocytes), which were in close proximity to motor neurons. Arrows indicate double-labeled cells. White frames indicate the areas of magnification. Scale: 50 µm; scale in the close-up images: 20 µm.</p>
Full article ">Figure 4
<p>Representative images of double immunofluorescence staining for CASP 1 with NeuN, Iba1, or GFAP in the cervical spinal cord tissues of p40 wild-type and wobbler mice. CASP 1 was predominantly expressed in wobbler NeuN<sup>+</sup> (motor neuronal cells), as well as in Iba1<sup>+</sup> (microglia) and GFAP<sup>+</sup> (astrocytes), which were in close proximity to motor neurons. Arrows indicate double-labeled cells. White frames indicate the areas of magnification. Scale: 50 µm; scale in the close-up images: 20 µm.</p>
Full article ">Figure 5
<p>Representative images of double immunofluorescence staining for GSDMD with Neuro Trace, Iba1, or GFAP in the cervical spinal cord tissues of p40 wild-type and wobbler mice. GSDMD was predominantly expressed in wobbler NeuN+ (motor neuronal cells), as well as in GFAP<sup>+</sup> (astrocytes), which were in close proximity to motor neurons. Arrows indicate double-labeled cells. White frames indicate the areas of magnification. Scale: 50 µm; scale in the close-up images: 20 µm.</p>
Full article ">Figure 6
<p>The statistical mean intensity analysis of (<b>A</b>) NeuN, (<b>B</b>) Iba1, (<b>C</b>) GFAP, (<b>D</b>) NLRP3, (<b>E</b>) CASP 1, and (<b>F</b>) GSDMD in the spinal cord of p40 wild-type and wobbler mice. Data are presented as mean ± SD, n is at least 30 per group. ** <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>Activation of NLRP3 inflammasome and pyroptotic cell death in motor neurons of the wobbler spinal cord. During the priming step, DAMP and PAMP molecules activate several different receptors such as TLR4, IL-1R, TNFR, and RAGE. This activation leads to ubiquitination and thereby degradation of IκB, the inhibitor of NFκB. As a consequence, P50 and RelA subunits of NFκB can be translocated into the nucleus, where they can upregulate the expression of inflammasome genes such as Nlrp3, pro-Il-1b, and pro-Il-18. (<b>A</b>) Increasing levels of miR-223-3p has been shown in the lasered ventral horn and cervical spinal cord of the wobbler mouse. However, it is unclear whether miR-223-3p targets NLRP3 or an alternative protein(s) in the NFκB translocation pathway in wobbler mice. (<b>B</b>) Elevated levels of ROS and (<b>C</b>) mitochondrial fragmentation in the spinal cord of the wobbler motor neurons trigger (<b>D</b>) NLRP3 inflammasome formation, leading to (<b>E</b>) auto-cleavage of pro-CASP 1 into c-CASP 1. On one hand, c-CASP 1 is able to cleave pro-IL-1β and pro-IL-18 into their mature forms, and on the other hand, it can cleave GSDMD protein. N-GSDMD subunits can translocate into the cellular membrane and form pores, which cause membrane rupture and eventually (<b>F</b>) pyroptotic motor neuronal death. Through these pores, mature cytokines can be released, leading to (<b>G</b>) inflammation for the neighboring cells. This image was created with BioRender.com.</p>
Full article ">
13 pages, 2573 KiB  
Article
The Anti-Inflammatory Effect of SDF-1 Derived Peptide on Porphyromonas gingivalis Infection via Regulation of NLRP3 and AIM2 Inflammasome
by Si Yeong Kim, Min Kee Son, Jung Hwa Park, Hee Sam Na and Jin Chung
Pathogens 2024, 13(6), 474; https://doi.org/10.3390/pathogens13060474 - 4 Jun 2024
Viewed by 315
Abstract
(1) Background: Peptides are appealing as pharmacological materials because they are easily produced, safe, and tolerable. Despite increasing gum-care awareness, periodontitis is still prevalent and is influenced by factors like high sugar consumption, smoking, and aging. Porphyromonas gingivalis is considered a major [...] Read more.
(1) Background: Peptides are appealing as pharmacological materials because they are easily produced, safe, and tolerable. Despite increasing gum-care awareness, periodontitis is still prevalent and is influenced by factors like high sugar consumption, smoking, and aging. Porphyromonas gingivalis is considered a major etiologic agent of periodontitis and activates the NLR family pyrin domain containing 3 (NLRP3) but is absent in melanoma 2 (AIM2) inflammasomes, resulting in pro-inflammatory cytokine release. (2) Methods: We examined the anti-inflammatory effects of 18 peptides derived from human stromal cell-derived factor-1 (SDF-1) on THP-1 macrophages. Inflammation was induced by P. gingivalis, and the anti-inflammatory effects were analyzed using molecular biological techniques. In a mouse periodontitis model, alveolar bone resorption was assessed using micro-CT. (3) Results: Of the 18 SDF-1-derived peptides, S10 notably reduced IL-1β and TNF-α secretion. S10 also diminished the P. gingivalis-induced expression of NLRP3, AIM2, ASC (apoptosis-associated speck-like protein), caspase-1, and IL-1β. Furthermore, S10 attenuated the enhanced TLR (toll-like receptor) signaling pathway and decreased the phosphorylation of nuclear factor-κB (NF-κB) and mitogen-activated protein kinases (MAPKs). In addition, S10 mitigated alveolar bone loss in our P. gingivalis-induced mouse model of periodontitis. (4) Conclusions: S10 suppressed TLR/NF-κB/NLRP3 inflammasome signaling and the AIM2 inflammasome in our P. gingivalis-induced murine periodontitis model, which suggests that it has potential use as a therapeutic treatment for periodontitis. Full article
(This article belongs to the Special Issue Nosocomial Infection and Antimicrobial Resistance)
Show Figures

Figure 1

Figure 1
<p>SDF-1-derived peptides suppressed the secretion of IL-1β and TNF-α. (<b>A</b>,<b>B</b>) THP-1 macrophages were pretreated with each SDF-1 derived peptide (1 μg/mL) for 30 min and were then infected with <span class="html-italic">P. gingivalis</span> (MOI 100). IL-1β (<b>A</b>) and TNF-α (<b>B</b>) secretion levels were measured by ELISA (<span class="html-italic">n =</span> 3). *** <span class="html-italic">p</span>-value &lt; 0.001 versus untreated group (Con); <sup>#</sup> <span class="html-italic">p</span>-value &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span>-value &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span>-value &lt; 0.001 versus <span class="html-italic">P. gingivalis</span> infection (Pg).</p>
Full article ">Figure 2
<p>S10 inhibited the activation of NLRP3 and AIM2 inflammasomes by <span class="html-italic">P. gingivalis</span>. (<b>A</b>) THP-1 macrophages were pretreated with S10 (1 μg/mL) for 30 min and were then infected with <span class="html-italic">P. gingivalis</span> (MOI 100) for 18 h. The secreted levels of IL-1β and TNF-α in the supernatant were determined by an ELISA (<span class="html-italic">n =</span> 3). (<b>B</b>) Real-time PCR was conducted to quantify its effects on NLRP3 and AIM2 inflammasome components; the results are presented in the graph (<span class="html-italic">n =</span> 3). (<b>C</b>) Cell lysates were subjected to Western blot analysis, and representative immunoblots and graphs of protein levels versus β-actin for each protein are shown (<span class="html-italic">n =</span> 3). Blot densities are expressed relative to treatment-naïve controls. (<b>D</b>) PMA-primed ASC-GFP-THP-1 cells were pretreated with S10 for 30 min and then infected with <span class="html-italic">P. gingivalis</span> for 18 h. “Pg” in the images indicates the ASC speck, indicated by yellow arrows (original magnification 200×). The graph shows the percentage of total cells containing ASC. * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 3
<p>S10 suppressed the <span class="html-italic">P. gingivalis</span>-induced activation of the TLR2/4 and NF-κB/MAPK signaling pathways. (<b>A</b>) Cell lysates of THP-1-derived macrophages, pretreated with S10 (1 μg/mL) for 30 min and infected with <span class="html-italic">P. gingivalis</span> (MOI 100) for 18 h, were studied via Western blot analysis. TLR2/4 and their downstream signaling proteins were analyzed, and representative figures, along with densitometric graphs normalized versus β-actin, are shown (<span class="html-italic">n =</span> 3). (<b>B</b>,<b>C</b>) The phosphorylation levels of NF-Κb and MAPK were evaluated using dedicated antibodies targeting each phosphorylated site, and normalization was conducted versus total protein levels (<span class="html-italic">n =</span> 3). * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01; *** <span class="html-italic">p</span>-value &lt; 0.001.</p>
Full article ">Figure 4
<p>S10 alleviated the alveolar bone loss caused by <span class="html-italic">P. gingivalis</span> infection. (<b>A</b>) Details of the S10 treatment protocol. (<b>B</b>,<b>C</b>) Micro-CT images were used to measure alveolar bone areas after orally administering <span class="html-italic">P. gingivalis</span> with or without S10 treatment. Areas between the cementoenamel junctions and alveolar bone crests of the three molars in the captured images were measured using ImageJ. Images are representative of the three groups, and graphs summarize the measured alveolar bone areas (<span class="html-italic">n =</span> 6). * <span class="html-italic">p</span>-value &lt; 0.05; ** <span class="html-italic">p</span>-value &lt; 0.01.</p>
Full article ">
21 pages, 937 KiB  
Review
Inflammasomes Are Influenced by Epigenetic and Autophagy Mechanisms in Colorectal Cancer Signaling
by Györgyi Műzes and Ferenc Sipos
Int. J. Mol. Sci. 2024, 25(11), 6167; https://doi.org/10.3390/ijms25116167 - 3 Jun 2024
Viewed by 101
Abstract
Inflammasomes contribute to colorectal cancer signaling by primarily inducing inflammation in the surrounding tumor microenvironment. Its role in inflammation is receiving increasing attention, as inflammation has a protumor effect in addition to inducing tissue damage. The inflammasome’s function is complex and controlled by [...] Read more.
Inflammasomes contribute to colorectal cancer signaling by primarily inducing inflammation in the surrounding tumor microenvironment. Its role in inflammation is receiving increasing attention, as inflammation has a protumor effect in addition to inducing tissue damage. The inflammasome’s function is complex and controlled by several layers of regulation. Epigenetic processes impact the functioning or manifestation of genes that are involved in the control of inflammasomes or the subsequent signaling cascades. Researchers have intensively studied the significance of epigenetic mechanisms in regulation, as they encompass several potential therapeutic targets. The regulatory interactions between the inflammasome and autophagy are intricate, exhibiting both advantageous and harmful consequences. The regulatory aspects between the two entities also encompass several therapeutic targets. The relationship between the activation of the inflammasome, autophagy, and epigenetic alterations in CRC is complex and involves several interrelated pathways. This article provides a brief summary of the newest studies on how epigenetics and autophagy control the inflammasome, with a special focus on their role in colorectal cancer. Based on the latest findings, we also provide an overview of the latest therapeutic ideas for this complex network. Full article
(This article belongs to the Special Issue Immunomodulatory Molecules in Cancer)
Show Figures

Figure 1

Figure 1
<p>Classification and mode of action of various inflammasomes. There are different PAMPs that can turn on the NLRP3, NLRP1, NLRC4, AIM2, and pyrin inflammasomes. NLRP3 is also capable of detecting uric acid crystals, mtDNA, ROS, ATP, and mineral particulates (e.g., asbestos or silica), among others. AIM2 interacts structurally with particular ligands, including dsDNA. Rho modification triggers the activation of pyrins in response to toxins produced by bacteria. Upon activation, the sensors recruit and stimulate caspase-1. If an inflammasome has a CARD domain, then it can interact directly with pro-caspase-1. If it only has a pyrin domain, then it does so through the adaptor protein ASC. IL1β and IL18 are produced when pro-IL1β and pro-IL18 are cleaved and activated by caspase-1, respectively. Additionally, caspase-1 mediates the cleavage of gasdermin D to induce pyroptosis. The figure was partially created with <a href="https://www.biorender.com" target="_blank">https://www.biorender.com</a> (accessed on 28 April 2024).</p>
Full article ">Figure 2
<p>The NLRP3 inflammasome pathway in CRC. PAMPs and DAMPs have the ability to trigger the activation of NLRP3 inflammasomes. The activation of NFκB and subsequent overexpression of NLRP3, triggered by microbial components or endogenous cytokines, initiates the generation of pro-IL1β and pro-IL18, leading to chronic inflammation in CRC. The inflammasome is under multi-level regulation. This regulatory network includes epigenetics and autophagy machinery. The figure was partially created using <a href="https://www.biorender.com" target="_blank">https://www.biorender.com</a> (accessed on 28 April 2024).</p>
Full article ">Figure 3
<p>Inflammasomes and autophagy engage in cross-communication. By eliminating endogenous inflammasome activators, such as damaged mitochondria that generate reactive ROS, inflammasome components, and cytokines, autophagy can negatively regulate NLRP3 inflammasome activation. In addition to regulating the inflammatory response, autophagic machinery prevents the unusual secretion of IL1β. On the other hand, when the NLRP3 inflammasome activates, a number of distinct mechanisms govern the formation of autophagosomes. The maintenance of equilibrium between the necessary inflammatory response of the host’s defenses and the prevention of excessive inflammation require cross-communication between inflammasomes and autophagy. The figure was partly created with <a href="https://www.biorender.com" target="_blank">https://www.biorender.com</a> (accessed on 28 April 2024).</p>
Full article ">
14 pages, 4631 KiB  
Article
TBHQ Alleviates Particulate Matter-Induced Pyroptosis in Human Nasal Epithelial Cells
by Ji-Sun Kim, Hyunsu Choi, Jeong-Min Oh, Sung Won Kim, Soo Whan Kim, Byung Guk Kim, Jin Hee Cho, Joohyung Lee and Dong Chang Lee
Toxics 2024, 12(6), 407; https://doi.org/10.3390/toxics12060407 - 3 Jun 2024
Viewed by 119
Abstract
Pyroptosis represents a type of cell death mechanism notable for its cell membrane disruption and the subsequent release of proinflammatory cytokines. The Nod-like receptor family pyrin domain containing inflammasome 3 (NLRP3) plays a critical role in the pyroptosis mechanism associated with various diseases [...] Read more.
Pyroptosis represents a type of cell death mechanism notable for its cell membrane disruption and the subsequent release of proinflammatory cytokines. The Nod-like receptor family pyrin domain containing inflammasome 3 (NLRP3) plays a critical role in the pyroptosis mechanism associated with various diseases resulting from particulate matter (PM) exposure. Tert-butylhydroquinone (tBHQ) is a synthetic antioxidant commonly used in a variety of foods and products. The aim of this study is to examine the potential of tBHQ as a therapeutic agent for managing sinonasal diseases induced by PM exposure. The occurrence of NLRP3 inflammasome-dependent pyroptosis in RPMI 2650 cells treated with PM < 4 µm in size was confirmed using Western blot analysis and enzyme-linked immunosorbent assay results for the pyroptosis metabolites IL-1β and IL-18. In addition, the inhibitory effect of tBHQ on PM-induced pyroptosis was confirmed using Western blot and immunofluorescence techniques. The inhibition of tBHQ-mediated pyroptosis was abolished upon nuclear factor erythroid 2-related factor 2 (Nrf2) knockdown, indicating its involvement in the antioxidant mechanism. tBHQ showed potential as a therapeutic agent for sinonasal diseases induced by PM because NLRP3 inflammasome activation was effectively suppressed via the Nrf2 pathway. Full article
(This article belongs to the Special Issue Air Pollutant Exposure and Respiratory Diseases)
Show Figures

Graphical abstract

Graphical abstract
Full article ">
28 pages, 3300 KiB  
Article
Inhibition of SARS-CoV-2-Induced NLRP3 Inflammasome-Mediated Lung Cell Inflammation by Triphala-Loaded Nanoparticle Targeting Spike Glycoprotein S1
by Chuda Chittasupho, Sonthaya Umsumarng, Kamonwan Srisawad, Punnida Arjsri, Rungsinee Phongpradist, Weerasak Samee, Wipawan Tingya, Chadarat Ampasavate and Pornngarm Dejkriengkraikul
Pharmaceutics 2024, 16(6), 751; https://doi.org/10.3390/pharmaceutics16060751 - 2 Jun 2024
Viewed by 255
Abstract
The COVID-19 pandemic, caused by SARS-CoV-2, poses a significant global health threat. The spike glycoprotein S1 of the SARS-CoV-2 virus is known to induce the production of pro-inflammatory mediators, contributing to hyperinflammation in COVID-19 patients. Triphala, an ancient Ayurvedic remedy composed of dried [...] Read more.
The COVID-19 pandemic, caused by SARS-CoV-2, poses a significant global health threat. The spike glycoprotein S1 of the SARS-CoV-2 virus is known to induce the production of pro-inflammatory mediators, contributing to hyperinflammation in COVID-19 patients. Triphala, an ancient Ayurvedic remedy composed of dried fruits from three plant species—Emblica officinalis (Family Euphorbiaceae), Terminalia bellerica (Family Combretaceae), and Terminalia chebula (Family Combretaceae)—shows promise in addressing inflammation. However, the limited water solubility of its ethanolic extract impedes its bioavailability. In this study, we aimed to develop nanoparticles loaded with Triphala extract, termed “nanotriphala”, as a drug delivery system. Additionally, we investigated the in vitro anti-inflammatory properties of nanotriphala and its major compounds, namely gallic acid, chebulagic acid, and chebulinic acid, in lung epithelial cells (A549) induced by CoV2-SP. The nanotriphala formulation was prepared using the solvent displacement method. The encapsulation efficiency of Triphala in nanotriphala was determined to be 87.96 ± 2.60% based on total phenolic content. In terms of in vitro release, nanotriphala exhibited a biphasic release profile with zero-order kinetics over 0–8 h. A549 cells were treated with nanotriphala or its active compounds and then induced with 100 ng/mL of spike S1 subunit (CoV2-SP). The results demonstrate that chebulagic acid and chebulinic acid are the active compounds in nanotriphala, which significantly reduced cytokine release (IL-6, IL-1β, and IL-18) and suppressed the expression of inflammatory genes (IL-6, IL-1β, IL-18, and NLRP3) (p < 0.05). Mechanistically, nanotriphala and its active compounds notably attenuated the expression of inflammasome machinery proteins (NLRP3, ASC, and Caspase-1) (p < 0.05). In conclusion, the nanoparticle formulation of Triphala enhances its stability and exhibits anti-inflammatory properties against CoV2-SP-induction. This was achieved by suppressing inflammatory mediators and the NLRP3 inflammasome machinery. Thus, nanotriphala holds promise as a supportive preventive anti-inflammatory therapy for COVID-19-related chronic inflammation. Full article
16 pages, 4382 KiB  
Article
Combined Insults of a MASH Diet and Alcohol Binges Activate Intercellular Communication and Neutrophil Recruitment via the NLRP3-IL-1β Axis in the Liver
by Mrigya Babuta, Prashanth Thevkar Nagesh, Aditi Ashish Datta, Victoria Remotti, Yuan Zhuang, Jeeval Mehta, Francesca Lami, Yanbo Wang and Gyongyi Szabo
Cells 2024, 13(11), 960; https://doi.org/10.3390/cells13110960 - 1 Jun 2024
Viewed by 298
Abstract
Binge drinking in obese patients positively correlates with accelerated liver damage and liver-related death. However, the underlying mechanism and the effect of alcohol use on the progression of metabolic-dysfunction-associated steatotic liver disease (MASLD) remain unexplored. Here, we show that short-term feeding of a [...] Read more.
Binge drinking in obese patients positively correlates with accelerated liver damage and liver-related death. However, the underlying mechanism and the effect of alcohol use on the progression of metabolic-dysfunction-associated steatotic liver disease (MASLD) remain unexplored. Here, we show that short-term feeding of a metabolic-dysfunction-associated steatohepatitis (MASH) diet plus daily acute alcohol binges for three days induce liver injury and activation of the NLRP3 inflammasome. We identify that a MASH diet plus acute alcohol binges promote liver inflammation via increased infiltration of monocyte-derived macrophages, neutrophil recruitment, and NET release in the liver. Our results suggest that both monocyte-derived macrophages and neutrophils are activated via NLRP3, while the administration of MCC950, an NLRP3 inhibitor, dampens these effects.In this study, we reveal important intercellular communication between hepatocytes and neutrophils. We discover that the MASH diet plus alcohol induces IL-1β via NLRP3 activation and that IL-1β acts on hepatocytes and promotes the production of CXCL1 and LCN2. In turn, the increase in these neutrophils recruits chemokines and causes further infiltration and activation of neutrophils in the liver. In vivo administration of the NLRP3 inhibitor, MCC950, improves the early phase of MetALD by preventing liver damage, steatosis, inflammation, and immune cells recruitment. Full article
(This article belongs to the Special Issue Inflammation in Target Organs)
Show Figures

Figure 1

Figure 1
<p>Short-term feeding of a MASH diet and acute alcohol binges promote liver injury and steatosis. (<b>A</b>) Feeding schematics for combined liver injury. C57BL/6 wild-type (WT) mice (n = 6–8) were fed on a MASH diet plus alcohol binges with a standard laboratory chow diet as a control for three days. (<b>B</b>,<b>C</b>) ALT and AST levels were measured from serum. (<b>D</b>) Formalin-fixed liver sections were stained with hematoxylin and eosin and representative slides are shown, scale bar = 50 μm. ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 2
<p>MASH diet and alcohol binges upregulate NLRP3 inflammasome. Liver RNA was used to determine <span class="html-italic">Nlrp3</span> (<b>A</b>), <span class="html-italic">Pycard</span> (<b>B</b>), <span class="html-italic">Casp1</span> (<b>C</b>), <span class="html-italic">Il1b</span> (<b>D</b>) and <span class="html-italic">Il18</span> (<b>E</b>) mRNA levels by qPCR. Here, 18s was used to normalize the Cq values. Liver lysates were used to detect cleaved caspase-1 (<b>F</b>), cleaved IL-1β (<b>G</b>) and cleaved gasdermin D (<b>H</b>) by Western blot. (<b>I</b>,<b>J</b>) Flow cytometry analysis of monocyte-derived macrophages, CD45<sup>+</sup>CD11b<sup>hi</sup>F4/80<sup>low</sup>CD86<sup>+</sup>, CD45<sup>+</sup>CD11b<sup>hi</sup>F4/80<sup>low</sup>CD206<sup>+</sup>. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 3
<p>Short-term feeding of MASH diet and alcohol induces hepatic neutrophil infiltration and NET formation. Liver RNA was used to determine <span class="html-italic">Cxcr1</span> (<b>A</b>), <span class="html-italic">Cxcr2</span> (<b>B</b>), <span class="html-italic">Cxcl1</span> (<b>C</b>), <span class="html-italic">Cxcl2</span> (<b>D</b>), <span class="html-italic">Lcn2</span> (<b>H</b>) and <span class="html-italic">Itgam</span> (<b>F</b>) mRNA levels by qPCR. Here, 18s was used to normalize Cq values. Whole-cell liver lysates were used to detect CXCL1 (<b>E</b>) by ELISA. (<b>G</b>) Flow cytometry analysis of neutrophils (CD11b<sup>+</sup>Ly6G<sup>+</sup>) in liver immune cells. (<b>I</b>–<b>K</b>) Whole-cell liver lysates were used to detect LCN2 (<b>I</b>), NE (<b>J</b>), and Cit-H3 (<b>K</b>) by ELISA. * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 4
<p>Inhibition of NLRP3 inflammasome by MCC950 ameliorates liver injury in short-term feeding of MASH diet and alcohol binges. (<b>A</b>) Feeding schematics for combined liver injury. C57BL/6 wild-type (WT) mice (n = 6–8) were fed with a MASH diet plus acute alcohol binges in the presence and absence of MCC950, with a standard laboratory chow diet as a control, for three days. (<b>B</b>,<b>C</b>) ALT and AST levels were measured from serum. (<b>D</b>) Formalin-fixed liver sections were stained with hematoxylin and eosin and representative slides are shown, scale bar = 50 μm. Liver RNA was used to determine <span class="html-italic">Casp1</span> (<b>E</b>), and <span class="html-italic">Il1b</span> (<b>F</b>), mRNA levels by qPCR. Here, 18s was used to normalize Cq values. (<b>G</b>) IL-1β level in the mice serum and whole-cell liver lysate was detected by ELISA. (<b>H</b>) ASC level in mice serum was detected by ELISA. (<b>I</b>) Liver RNA was used to determine <span class="html-italic">Tnf</span> and <span class="html-italic">Ccl2</span>, mRNA levels by qPCR. Moreover, 18s was used to normalize Cq values. Flow cytometry analysis of monocyte- derived macrophages, CD45<sup>+</sup>CD11b<sup>hi</sup>F4/80<sup>low</sup>CD86<sup>+</sup> (<b>J</b>), CD45<sup>+</sup>CD11b<sup>hi</sup>F4/80<sup>low</sup>CD206<sup>+</sup> (<b>K</b>). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.00005.ns; not significant.</p>
Full article ">Figure 5
<p>Inhibiting NLRP3 attenuates neutrophil infiltration and NET formation in short-term feeding of MASH diet plus alcohol binges. (<b>A</b>) Flow cytometry analysis of neutrophils (CD11b<sup>+</sup>Ly6G<sup>+</sup>) in liver immune cells. (<b>B</b>,<b>D</b>) CXCL1 and LCN2 were detected in serum by ELISA. (<b>C</b>,<b>E</b>) Liver lysates were used to detect CXCL1 (<b>B</b>), and LCN2 (<b>D</b>), by ELISA. (<b>F</b>) NE was detected in serum by ELISA. (<b>G</b>,<b>H</b>) Whole-cell liver lysates were used to detect the level of NE (<b>G</b>) and Cit-H3 (<b>H</b>) by ELISA. (<b>I</b>) Bone-marrow-derived neutrophils from WT and NLRP3-KO were treated with PA or EtOH, and NET-associated NE was measured to quantify NETs. (<b>J</b>) Contour plot depeciting the Ly6G<sup>+</sup> cells. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, *** <span class="html-italic">p</span> &lt; 0.0005, **** <span class="html-italic">p</span> &lt; 0.00005.</p>
Full article ">Figure 6
<p>Exogenous IL-1β regulates production of CXCL1 and LCN2 from primary hepatocytes. (<b>A</b>,<b>B</b>) Supernatant of WT and NLRP3-KO primary hepatocytes after exogenous IL-1β treatment was used to detect CXCL1 (<b>A</b>) and LCN2 (<b>B</b>) by ELISA. mRNA of WT and NLRP3-KO primary hepatocytes after exogenous IL-1β treatment was used to determine <span class="html-italic">Cxcl1</span> (<b>C</b>), and <span class="html-italic">Lcn2</span> (<b>D</b>), mRNA levels by qPCR. Here, 18s was used to normalize Cq values. Whole-cell lysate of * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005, **** <span class="html-italic">p</span> &lt; 0.00005 (<b>E</b>) Briefly, MASH diet and alcohol binges lead to increased production of DAMPs and PAMPs, which activate NLRP3 inflammasome in monocyte-derived macrophages. NLRP3-dependent IL-1β promotes the production of CXCL1 and LCN2 from damaged hepatocytes, which in turn leads to increased neutrophil infiltration. NET formation by infiltrated neutrophils occurs in an NLRP3-depedent manner.</p>
Full article ">
30 pages, 1499 KiB  
Review
Crosstalk between Inflammation and Atherosclerosis in Rheumatoid Arthritis and Systemic Lupus Erythematosus: Is There a Common Basis?
by Marta Chiara Sircana, Gian Luca Erre, Floriana Castagna and Roberto Manetti
Life 2024, 14(6), 716; https://doi.org/10.3390/life14060716 - 31 May 2024
Viewed by 185
Abstract
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the [...] Read more.
Cardiovascular disease is the leading cause of morbidity and mortality in patients with rheumatoid arthritis and systemic lupus erythematosus. Traditional cardiovascular risk factors, although present in lupus and rheumatoid arthritis, do not explain such a high burden of early cardiovascular disease in the context of these systemic connective tissue diseases. Over the past few years, our understanding of the pathophysiology of atherosclerosis has changed from it being a lipid-centric to an inflammation-centric process. In this review, we examine the pathogenesis of atherosclerosis in systemic lupus erythematosus and rheumatoid arthritis, the two most common systemic connective tissue diseases, and consider them as emblematic models of the effect of chronic inflammation on the human body. We explore the roles of the inflammasome, cells of the innate and acquired immune system, neutrophils, macrophages, lymphocytes, chemokines and soluble pro-inflammatory cytokines in rheumatoid arthritis and systemic lupus erythematosus, and the roles of certain autoantigens and autoantibodies, such as oxidized low-density lipoprotein and beta2-glycoprotein, which may play a pathogenetic role in atherosclerosis progression. Full article
(This article belongs to the Special Issue Clinical Manifestations and Treatment of Autoimmune Diseases)
Show Figures

Figure 1

Figure 1
<p>Pro-atherogenic pathways in RA and SLE.</p>
Full article ">
13 pages, 2374 KiB  
Article
Bovine DDX3X Restrains Bovine SP110c-Mediated Activation of Inflammasome in Macrophages
by Zhunan Li, Jing Han, Jiayi Jing, Ajiao Fan, Yong Zhang and Yuanpeng Gao
Animals 2024, 14(11), 1650; https://doi.org/10.3390/ani14111650 - 31 May 2024
Viewed by 158
Abstract
The inflammasome is a vital part of the host’s innate immunity activated by cellular infection or stress. Our previous research identified the bovine SP110c isoform (bSP110c) as a novel activator of the inflammasome that promoted the secretion of proinflammatory cytokines IL-1β and IL-18 [...] Read more.
The inflammasome is a vital part of the host’s innate immunity activated by cellular infection or stress. Our previous research identified the bovine SP110c isoform (bSP110c) as a novel activator of the inflammasome that promoted the secretion of proinflammatory cytokines IL-1β and IL-18 in macrophages infected with Listeria monocytogenes or stimulated with lipopolysaccharide (LPS). However, the exact molecular mechanism for inhibiting bSP110c-induced inflammasome activation requires further clarification. Here, the researchers identified bovine DDX3X (bDDX3X) as an NLRP3-associated protein and an inhibitor of the bSP110c-induced inflammasome in the human THP1 macrophage cell line. Immunoprecipitation showed that bDDX3X interacted with the bSP110c CARD domain via its helicase domain. The co-expression of bSP110c and bDDX3X in THP1 macrophages significantly prevented the bSP110c-induced activation of inflammasomes. In addition, both bDDX3X and bSP110c interacted with bovine NLRP3 (bNLRP3), and bDDX3X enhanced the interaction between bSP110c and bNLRP3. The expression of bDDX3X in nigericin-stimulated THP1 macrophages significantly suppressed NLRP3 inflammasome activation, ASC speck formation, and pyroptosis. These findings demonstrate that bDDX3X negatively regulates the bSP110c-mediated inflammatory response by restricting the activation of the NLRP3 inflammasome. This discovery unveils a novel regulatory mechanism involving bDDX3X and bSP110c in coordinating inflammasome activation and subsequent cell-fate decisions in LPS-treated macrophages and, in turn, constitutes a step forward toward the implementation of marker-assisted selection in breeding programs aimed at utilizing cattle’s immune defenses. Full article
(This article belongs to the Section Cattle)
Show Figures

Figure 1

Figure 1
<p>Bovine DDX3X interacts with bSP110c: (<b>a</b>) HEK293T cells were transfected with recombinant vectors for indicated expression of bDDX3X-HA and bSP110c-FLAG. Whole-cell extracts were co-IP with anti-Flag antibody or anti-HA antibody and analyzed by Western blot (WB) with respective antibodies. (<b>b</b>) Diagram of HA-tagged bDDX3X domain-deletion expression recombinant vectors. (<b>c</b>) bSP110c-FLAG and HA-labeled bDDX3X domain-deletion mutants were co-expressed in HEK293T cells. Whole-cell lysis underwent co-IP with anti-Flag antibody and was tested by WB with respective antibodies. (<b>d</b>) Diagram of FLAG-tagged bSP110c domain-deletion expression recombinant vectors. (<b>e</b>) bDDX3X-HA and Flag-labeled bSP110c domain-deletion mutants were co-expressed in HEK293T cells. Whole-cell lysis was subjected to co-IP with anti-Flag antibody and detected by WB with respective antibodies.</p>
Full article ">Figure 2
<p>Bovine DDX3X suppresses bSP110c-induced inflammatory response: (<b>a</b>) WB of cleaved IL-1β, caspase-1, and GSDMD in whole-cell extracts of THP1 cells expressing bSP110c or co-expressing bSP110c and bDDX3X, primed with LPS. β-tubulin was also detected as loading control. (<b>b</b>) ELISA of IL-1β and IL18 in supernatants from LPS primed THP1 cells expressed of bSP110c or bSP110c and bDDX3X. **** <span class="html-italic">p</span> &lt; 0.0001. (<b>c</b>) bSP110c expressed or bSP110c and bDDX3X co-expressed THP1 cells, treated with LPS were evaluated using an ATP-based luminescent cell viability assay. *** <span class="html-italic">p</span> &lt; 0.0004.</p>
Full article ">Figure 3
<p>Bovine NLRP3 interacts with bDDX3X and bSP110c: (<b>a</b>) HEK293T cells were transfected with recombined constructs for indicated expression of bDDX3X-FLAG, bNLRP3-HA, and bASC-HA. Whole-cell extracts were subjected to co-IP with anti-Flag antibody or anti-HA antibody and detected by WB with respective antibodies. (<b>b</b>) HEK293T cells were transfected with recombined constructs for indicated expression of bSP110c-FLAG, bNLRP3-HA, bASC-HA, and bDDX3X-HA. Whole-cell extracts underwent co-IP with anti-Flag antibody or anti-HA antibody and were detected by WB with respective antibodies.</p>
Full article ">Figure 4
<p>Bovine DDX3X inhibits pyroptosis and activation of NLRP3 inflammasomes in macrophages: (<b>a</b>) Confocal microscopy of THP1 cells expressing bDDX3X, pretreated with LPS, stimulated with nigericin, and stained for NLRP3 (green) ASC (red) and DAPI (blue). Scale bars, 10 μm. (<b>b</b>) THP1 cells expressing bDDX3X were treated with LPS and nigericin and evaluated using an ATP-based luminescent cell viability assay. ** <span class="html-italic">p</span> = 0.0014. (<b>c</b>) WB of cleaved IL-1β, caspase-1, and GSDMD in whole-cell extracts of bDDX3X-expressing THP1 cells, pretreated with LPS, and then stimulated with nigericin is shown. β-tubulin was also run as a loading control. (<b>d</b>) ELISA of IL-1β and IL18 in supernatants from THP1 cells expressing bDDX3X, incubated with LPS followed by nigericin ** <span class="html-italic">p</span> &lt; 0.0078.</p>
Full article ">Figure 5
<p>Diagram of the effects of interaction between bDDX3X and bSP110c and its relationship with inflammasome activation and cell-fate decisions. bDDX3X inhibits NLRP3 inflammasome activation probably by interacting with NLRP3. bDDX3X restrains bSP110c activation of NLRP3 inflammasomes in macrophages most likely by interacting with the bSP110c CARD domain through its helicase domain and regulating bSP110c interaction with NLRP3.</p>
Full article ">
25 pages, 1605 KiB  
Review
NLRP3 Inflammasome Inhibitors for Antiepileptogenic Drug Discovery and Development
by Inamul Haque, Pritam Thapa, Douglas M. Burns, Jianping Zhou, Mukut Sharma, Ram Sharma and Vikas Singh
Int. J. Mol. Sci. 2024, 25(11), 6078; https://doi.org/10.3390/ijms25116078 - 31 May 2024
Viewed by 265
Abstract
Epilepsy is one of the most prevalent and serious brain disorders and affects over 70 million people globally. Antiseizure medications (ASMs) relieve symptoms and prevent the occurrence of future seizures in epileptic patients but have a limited effect on epileptogenesis. Addressing the multifaceted [...] Read more.
Epilepsy is one of the most prevalent and serious brain disorders and affects over 70 million people globally. Antiseizure medications (ASMs) relieve symptoms and prevent the occurrence of future seizures in epileptic patients but have a limited effect on epileptogenesis. Addressing the multifaceted nature of epileptogenesis and its association with the Nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated neuroinflammation requires a comprehensive understanding of the underlying mechanisms of these medications for the development of targeted therapeutic strategies beyond conventional antiseizure treatments. Several types of NLRP3 inhibitors have been developed and their effect has been validated both in in vitro and in vivo models of epileptogenesis. In this review, we discuss the advances in understanding the regulatory mechanisms of NLRP3 activation as well as progress made, and challenges faced in the development of NLRP3 inhibitors for the treatment of epilepsy. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the mechanism regulating NLRP3 inflammasome activation in canonical, non-canonical, and alternate pathway. Optimal activation of NLRP3 requires two steps. The first step is priming, which is initiated by extracellular PAMPs and endogenous cytokines by the PRRs, which then upregulates the NF-κB-mediated transcription of NLRP3, pro-IL-1β, and pro-IL-18. The second step is activation, which includes canonical and non-canonical pathways. The canonical pathway is triggered by multiple pathogens and inflammatory agents through a combination of important and related events such as activation of the purinergic receptor P2X7 by ATP, cathepsin release following lysozyme rupture, opening of Ca2+ channels to allow ion flux, mitochondrial dysfunction, ROS formation, Golgi apparatus disassembly, and endoplasmic reticulum stress. Once activated, oligomerization of the NLRP3 inflammasome is thought to induce conformational changes that generate active caspase-1, which converts pro-pro-IL-1β and pro-IL-18 to mature bioactive IL-1β and IL-18. Additionally, Casp1 cleaves the protein gasdermin D to generate N-terminal gasdermin D to form pores, allowing IL-1β and IL-18 to leave the cell and effectively execute a highly inflammatory form of cell death that is termed pyroptosis. In the non-canonical pathway, intracellular LPS is directly recognized by the CARD domain of caspase-11 in mice and caspase-4/5 in humans, ultimately leading to IL-1β and IL-18 release through the activation of the NLRP3-ASC-Casp1 pathway. The alternative pathway of activation is caused by TLR4 agonists like LPS, which activates the TLR4-TRIF-RIPK1-FADD-Casp8 signaling pathway. Casp8 activates the NLRP3 inflammasome but lacks ASC speck formation, pyroptosis induction, or K+ efflux.</p>
Full article ">Figure 2
<p>Chemical structures of clinically approved antiseizure medications discussed in this review.</p>
Full article ">Figure 3
<p>Chemical structure of different NLRP3 inflammasome inhibitors discussed in this review.</p>
Full article ">
11 pages, 249 KiB  
Review
Schnitzler Syndrome: Insights into Its Pathogenesis, Clinical Manifestations, and Current Management
by Antoine Braud and Dan Lipsker
Biomolecules 2024, 14(6), 646; https://doi.org/10.3390/biom14060646 - 31 May 2024
Viewed by 349
Abstract
Schnitzler syndrome is a rare disorder characterized by a chronic urticarial rash associated with immunoglobulin M (IgM) monoclonal gammopathy. Schnitzler syndrome shares strong clinicopathologic similarities with monogenic IL-1-mediated autoinflammatory disorders and is now considered an acquired adult-onset autoinflammatory disease. The spectacular effect of [...] Read more.
Schnitzler syndrome is a rare disorder characterized by a chronic urticarial rash associated with immunoglobulin M (IgM) monoclonal gammopathy. Schnitzler syndrome shares strong clinicopathologic similarities with monogenic IL-1-mediated autoinflammatory disorders and is now considered an acquired adult-onset autoinflammatory disease. The spectacular effect of interleukin-1 inhibitors demonstrates the key role of this cytokine in the pathogenesis of the disease. However, the physiopathology of Schnitzler syndrome remains elusive, and the main question regarding the relationship between autoinflammatory features and monoclonal gammopathy is still unanswered. The purpose of this narrative review is to describe what is currently known about the pathogenesis of this peculiar disease, as well as to address its diagnosis and management. Full article
(This article belongs to the Special Issue Novel Insights into Autoimmune/Autoinflammatory Skin Diseases)
Back to TopTop