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
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,255)

Search Parameters:
Keywords = AIM2 like receptor

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 9137 KiB  
Article
Utilizing Immunoinformatics for mRNA Vaccine Design against Influenza D Virus
by Elijah Kolawole Oladipo, Stephen Feranmi Adeyemo, Modinat Wuraola Akinboade, Temitope Michael Akinleye, Kehinde Favour Siyanbola, Precious Ayomide Adeogun, Victor Michael Ogunfidodo, Christiana Adewumi Adekunle, Olubunmi Ayobami Elutade, Esther Eghogho Omoathebu, Blessing Oluwatunmise Taiwo, Elizabeth Olawumi Akindiya, Lucy Ochola and Helen Onyeaka
BioMedInformatics 2024, 4(2), 1572-1588; https://doi.org/10.3390/biomedinformatics4020086 - 12 Jun 2024
Viewed by 126
Abstract
Background: Influenza D Virus (IDV) presents a possible threat to animal and human health, necessitating the development of effective vaccines. Although no human illness linked to IDV has been reported, the possibility of human susceptibility to infection remains uncertain. Hence, there is a [...] Read more.
Background: Influenza D Virus (IDV) presents a possible threat to animal and human health, necessitating the development of effective vaccines. Although no human illness linked to IDV has been reported, the possibility of human susceptibility to infection remains uncertain. Hence, there is a need for an animal vaccine to be designed. Such a vaccine will contribute to preventing and controlling IDV outbreaks and developing effective countermeasures against this emerging pathogen. This study, therefore, aimed to design an mRNA vaccine construct against IDV using immunoinformatic methods and evaluate its potential efficacy. Methods: A comprehensive methodology involving epitope prediction, vaccine construction, and structural analysis was employed. Viral sequences from six continents were collected and analyzed. A total of 88 Hemagglutinin Esterase Fusion (HEF) sequences from IDV isolates were obtained, of which 76 were identified as antigenic. Different bioinformatics tools were used to identify preferred CTL, HTL, and B-cell epitopes. The epitopes underwent thorough analysis, and those that can induce a lasting immunological response were selected for the construction. Results: The vaccine prototype comprised nine epitopes, an adjuvant, MHC I-targeting domain (MITD), Kozaq, 3′ UTR, 5′ UTR, and specific linkers. The mRNA vaccine construct exhibited antigenicity, non-toxicity, and non-allergenicity, with favourable physicochemical properties. The secondary and tertiary structure analyses revealed a stable and accurate vaccine construct. Molecular docking simulations also demonstrated strong binding affinity with toll-like receptors. Conclusions: The study provides a promising framework for developing an effective mRNA vaccine against IDV, highlighting its potential for mitigating the global impact of this viral infection. Further experimental studies are needed to confirm the vaccine’s efficacy and safety. Full article
(This article belongs to the Special Issue Computational Biology and Artificial Intelligence in Medicine)
Show Figures

Figure 1

Figure 1
<p>Workflow of the Methodology used in the study.</p>
Full article ">Figure 2
<p>Three-dimensional structures of eight predicted conformational B-cell epitopes: The yellow portion represents the B-cell epitope, while the grey portion represents the surrounding residues. The pI scores for each epitope are as follows: (<b>A</b>) 0.848 with eight residues, (<b>B</b>) 0.691 with 9 residues, (<b>C</b>) 0.831 with 10 residues, (<b>D</b>) 0.861 with 11 residues, (<b>E</b>) 0.885 with 11 residues, (<b>F</b>) 0.815 with 12 residues, (<b>G</b>) 0.923 with 13 residues, and (<b>H</b>) 0.799 with 20 residues.</p>
Full article ">Figure 3
<p>The schematic diagram showing the final mRNA vaccine construct. The 1058-amino acid-designed vaccine consists of 5′ Cap, 5′ UTR, Kozak sequence, tPA, adjuvant (purple) and three HTL (orange) epitopes linked by the GPGPG linker (black). The HTLs are joined together by GPGPG, the last HTL epitope and the first LBL (yellow) epitope are linked by the KK linker, including the other LBLs. The last LBL and the CTLs (green) are linked by EAAAK. MITD, 3′ UTR and Poly-A tail (121 alanine) are added at the C-terminal end of the vaccine construct for stability and purification.</p>
Full article ">Figure 4
<p>A graph showing the solubility prediction of the vaccine construct (QuerySol) and the average soluble <span class="html-italic">E. coli</span> protein (PopAvrSol).</p>
Full article ">Figure 5
<p>Secondary structure prediction of the vaccine construct.</p>
Full article ">Figure 6
<p>Three-dimensional structure of the vaccine construct. (<b>A</b>) Structure (Rank 1) of the vaccine from AlphaFold (<b>B</b>) Refined vaccine construct (in ribbon) from Galaxy Refine (<b>C</b>) Surface structure of the refined vaccine construct.</p>
Full article ">Figure 7
<p>Validation of the 3-D structure. (<b>A</b>) Results of the Ramachandran plot generated by the PROCHECK (<b>B</b>) Analysis of the Ramachandran plot.</p>
Full article ">Figure 8
<p>Molecular docking results. (<b>A</b>) Tertiary structure of the construct. (<b>B</b>) Toll-like receptor-2 (TLR-2). (<b>C</b>) Docked complex of TLR-2 (Green) and the construct (Orange).</p>
Full article ">Figure 9
<p>Molecular docking results. (<b>A</b>) Tertiary structure of the construct. (<b>B</b>) Toll-like receptor-4 (TLR-4). (<b>C</b>) Docked complex of TLR-4 (Blue) and the vaccine construct (Orange).</p>
Full article ">
15 pages, 2550 KiB  
Article
Multi-Cohort Transcriptomic Profiling of Medical Gas Plasma-Treated Cancers Reveals the Role of Immunogenic Cell Death
by Antonios Gkantaras, Charalampos Kotzamanidis, Konstantinos Kyriakidis, Evangelia Farmaki, Kali Makedou, Georgios Tzimagiorgis, Sander Bekeschus and Andigoni Malousi
Cancers 2024, 16(12), 2186; https://doi.org/10.3390/cancers16122186 - 10 Jun 2024
Viewed by 301
Abstract
The therapeutic potential of cold physical gas plasma operated at atmospheric pressure in oncology has been thoroughly demonstrated in numerous preclinical studies. The cytotoxic effect on malignant cells has been attributed mainly to biologically active plasma-generated compounds, namely, reactive oxygen and nitrogen species. [...] Read more.
The therapeutic potential of cold physical gas plasma operated at atmospheric pressure in oncology has been thoroughly demonstrated in numerous preclinical studies. The cytotoxic effect on malignant cells has been attributed mainly to biologically active plasma-generated compounds, namely, reactive oxygen and nitrogen species. The intracellular accumulation of reactive oxygen and nitrogen species interferes strongly with the antioxidant defense system of malignant cells, activating multiple signaling cascades and inevitably leading to oxidative stress-induced cell death. This study aims to determine whether plasma-induced cancer cell death operates through a universal molecular mechanism that is independent of the cancer cell type. Using whole transcriptome data, we sought to investigate the activation mechanism of plasma-treated samples in patient-derived prostate cell cultures, melanoma, breast, lymphoma, and lung cancer cells. The results from the standardized single-cohort gene expression analysis and parallel multi-cohort meta-analysis strongly indicate that plasma treatment globally induces cancer cell death through immune-mediated mechanisms, such as interleukin signaling, Toll-like receptor cascades, and MyD88 activation leading to pro-inflammatory cytokine release and tumor antigen presentation. Full article
(This article belongs to the Section Cancer Immunology and Immunotherapy)
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the search strategy and gene expression dataset selection process.</p>
Full article ">Figure 2
<p>log<sub>2</sub>-scaled, quantile normalized expression levels (log<sub>2</sub>Gex) of the statistically significant genes that were found to be up-regulated in the plasma-treated samples of four cohorts.</p>
Full article ">Figure 3
<p>(<b>a</b>) Flow visualization of linking plasma-treated cancer cells and pathway classes. (<b>b</b>) Immune response pathways involved in plasma treatment and the number of cohorts, out of eight, showed statistically significant deregulation.</p>
Full article ">Figure 4
<p>Pathways affected by the up-regulated genes of the meta-analysis with FDR ≤ 0.05.</p>
Full article ">Figure 5
<p>GO biological processes that are affected by the up-regulated genes.</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 320
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, 670 KiB  
Review
Management of Fibromyalgia: An Update
by Eric A. Jones, Farrah Asaad, Nishil Patel, Esha Jain and Alaa Abd-Elsayed
Biomedicines 2024, 12(6), 1266; https://doi.org/10.3390/biomedicines12061266 - 6 Jun 2024
Viewed by 456
Abstract
Fibromyalgia, a chronic pain condition marked by abnormal pain processing, impacts a significant part of the population, leading to reduced quality of life and function. Hallmark symptoms include widespread persistent pain, sleep disturbances, fatigue, cognitive dysfunction, and mood changes. Through this updated review, [...] Read more.
Fibromyalgia, a chronic pain condition marked by abnormal pain processing, impacts a significant part of the population, leading to reduced quality of life and function. Hallmark symptoms include widespread persistent pain, sleep disturbances, fatigue, cognitive dysfunction, and mood changes. Through this updated review, we aim to contribute to the evolving understanding and management of fibromyalgia, offering insights into the diverse tools available to improve the lives of those affected by this challenging condition. Management begins with educating patients to ultimately relieve them of unnecessary testing and provide reassurance. Treatment emphasizes a comprehensive approach, combining nonpharmacological interventions such as aforementioned education, exercise, and psychotherapy, alongside pharmacologic management—namely duloxetine, milnacipran, pregabalin, and amitriptyline—which have consistent benefits for a range of symptoms across the spectrum of fibromyalgia. Notably, drugs like nonsteroidal anti-inflammatory drugs (NSAIDs) and acetaminophen are generally not recommended due to limited efficacy and associated risks. Lastly, a variety of other medications have shown promise, including NMDA-receptor antagonists, naltrexone, and cannabinoids; however, they should be used with caution due to a small amount of evidence and potential for adverse effects. Full article
(This article belongs to the Collection Neurogenic Neuroinflammation in Fibromyalgia)
Show Figures

Figure 1

Figure 1
<p>Flow diagram illustrating the multifaceted treatment of fibromyalgia.</p>
Full article ">
28 pages, 1160 KiB  
Review
New Molecules in Type 2 Diabetes: Advancements, Challenges and Future Directions
by Kyriazoula Chatzianagnostou, Melania Gaggini, Adrian Suman Florentin, Ludovica Simonini and Cristina Vassalle
Int. J. Mol. Sci. 2024, 25(11), 6218; https://doi.org/10.3390/ijms25116218 - 5 Jun 2024
Viewed by 236
Abstract
Although good glycemic control in patients with type 2 diabetes (T2D) can prevent cardiovascular complications, many diabetic patients still have poor optimal control. A new class of antidiabetic drugs (e.g., glucagon-like peptide-1-GLP-1 receptor agonists, sodium-glucose co-transporters-SGLT2 inhibitors), in addition to the low hypoglycemic [...] Read more.
Although good glycemic control in patients with type 2 diabetes (T2D) can prevent cardiovascular complications, many diabetic patients still have poor optimal control. A new class of antidiabetic drugs (e.g., glucagon-like peptide-1-GLP-1 receptor agonists, sodium-glucose co-transporters-SGLT2 inhibitors), in addition to the low hypoglycemic effect, exert multiple beneficial effects at a metabolic and cardiovascular level, through mechanisms other than antihyperglycemic agents. This review aims to discuss the effects of these new antidiabetic drugs, highlighting cardiovascular and metabolic benefits, through the description of their action mechanisms as well as available data by preclinical and clinical studies. Moreover, new innovative tools in the T2D field will be described which may help to advance towards a better targeted T2D personalized care in future. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Summary of the GLP-1RA in preclinical studies.</p>
Full article ">Figure 2
<p>Summary of the SGLT2i in preclinical studies.</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 308
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 ">
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 114
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 ">
16 pages, 4308 KiB  
Article
The Role of TLR7 and TLR9 in the Pathogenesis of Systemic Sclerosis
by Chenyang Wang, Kyosuke Oishi, Tadahiro Kobayashi, Ko Fujii, Motoki Horii, Natsumi Fushida, Tasuku Kitano, Shintaro Maeda, Yuichi Ikawa, Akito Komuro, Yasuhito Hamaguchi and Takashi Matsushita
Int. J. Mol. Sci. 2024, 25(11), 6133; https://doi.org/10.3390/ijms25116133 - 1 Jun 2024
Viewed by 176
Abstract
The bleomycin-induced scleroderma model is a well-established and dependable method for creating a mouse model of SSc (systemic sclerosis). In the field of skin connective tissue diseases, increasing evidence from clinical and animal experiments suggests that TLRs (Toll-like receptors) play an important role [...] Read more.
The bleomycin-induced scleroderma model is a well-established and dependable method for creating a mouse model of SSc (systemic sclerosis). In the field of skin connective tissue diseases, increasing evidence from clinical and animal experiments suggests that TLRs (Toll-like receptors) play an important role in several diseases. This study aimed to determine the role of TLR7 (Toll-like receptor 7) and TLR9 (Toll-like receptor 9) in the mechanisms of immune abnormalities and fibrosis in SSc. This study used TLR7-KO mice (TLR7-knockout mice with a balb/c background) and TLR9-KO mice (TLR9-knockout mice with a balb/c background) as well as WT mice (wild-type balb/c mice). All three kinds of mice were induced by BLM (bleomycin) in a scleroderma model as the experimental group; meanwhile, WT mice treated with PBS (phosphate-buffered saline) were used as the control group. We analyzed the fibrotic phenotype and the immunological abnormality phenotype of TLR7-deficient and TLR9-deficient mice in the SSc disease model using flow cytometry, RT-PCR (reverse transcription–polymerase chain reaction), a histological examination, and IHC (immunohistochemical staining). In a mouse model of SSc disease, the deletion of TLR7 attenuated skin and lung fibrosis, while the deletion of TLR9 exacerbated skin and lung fibrosis. The deletion of TLR7 resulted in a relative decrease in the infiltration and expression of various pro-inflammatory and fibrotic cells and cytokines in the skin. On the other hand, the deletion of TLR9 resulted in a relative increase in the infiltration and expression of various pro-inflammatory and cytokine-inhibiting cells and cytokines in the skin. Under the influence of pDCs (plasmacytoid dendritic cells), the balances of Beff/Breg (IL-6 + CD19 + B cell/IL-10 + CD19 + B cell), Th17/Treg (IL-17A + CD4 + T cell/Foxp3 + CD25 + CD4 + T cell), M1/M2 (CD86 + macrophage/CD206 + macrophage), and Th1/Th2 (TNFα + CD3 + CD4 + T cell/IL-4 + CD3 + CD4 + T cell) were biased towards the suppression of inflammation and fibrosis as a result of the TLR7 deletion. Comparatively, the balance was biased towards promoting inflammation and fibrosis due to the TLR9 deletion. In the SSc model, TLR7 promoted inflammation and fibrosis progression, while TLR9 played a protective role. These results suggest that TLR7 and TLR9 play opposite roles in triggering SSc to produce immune system abnormalities and skin fibrosis. Full article
(This article belongs to the Special Issue News in Skin Diseases: From Basic Mechanisms to Therapies)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Examples of Masson’s trichrome-stained skin tissue sections. The scale bars are all 50 μm. The dermal thicknesses in the blue sections were all measured at the same location, as indicated by the red scale bar. (<b>B</b>) The left panel corresponds to a bar chart of the dermal thickness of each mouse skin. The right panel shows the mRNA expression of TGFβ in the skin. (<b>C</b>) Immunohistochemical sections of αSMA-positive cell infiltration in the skin. The red part is αSMA-positive cells. The scale bars are all 20 μm. (<b>D</b>) Bar graphs of each type of mouse, corresponding to the C graph. (<b>E</b>): Example of a section of mouse lung tissue stained with Masson’s trichrome. All are microscopic images of the same site in the lower lateral part of the left lung. The scale bars are all 50 μm. (<b>F</b>) The degree of fibrosis in the (<b>E</b>) was quantified by using the Ashcroft Score and made into a bar chart. * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, and **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with a sample size of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left-hand corner of each example figure.</p>
Full article ">Figure 2
<p>(<b>A</b>) Flow cytometry results from an independent experiment, reflecting the proportion of CD11c-staining-positive cells with positive PDCA-1 staining. (<b>B</b>) Bar graphs for each mouse species, corresponding to the left panel. **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with a sample size of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left corner of each example graph.</p>
Full article ">Figure 3
<p>(<b>A</b>) Flow cytometry results, reflecting the proportion of cells positive for IL-6 staining in an independent experiment. (<b>B</b>) Bar graphs of CD19 + IL6 + for each mouse species, corresponding to (<b>A</b>). (<b>C</b>) The mRNA expression of IL-6 in skin tissue. (<b>D</b>) The flow cytometry results, reflecting the proportion of IL-10 staining in CD19-positive cells in an independent experiment. (<b>E</b>) The mRNA expression of IL-10 in skin tissue. (<b>F</b>) Bar graphs for each type of mouse corresponding to the graph. * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, and **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with sample sizes of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left corner of each example graph.</p>
Full article ">Figure 4
<p>(<b>A</b>) Flow cytometry results, reflecting the proportion of cells positive for Foxp3 staining in an independent experiment with positive CD4 and CD25 staining. (<b>B</b>) Bar graphs for each mouse species, corresponding to (<b>A</b>). (<b>C</b>) The flow cytometry results, reflecting the proportion of IL-17A-positive cells in CD4-positive cells in an independent experiment. (<b>D</b>) Corresponding bar charts for each mouse species. * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, and **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with sample sizes of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left corner of each example graph.</p>
Full article ">Figure 5
<p>(<b>A</b>) Flow cytometry results, reflecting the proportion of cells positive for IL-4 staining in an independent experiment. (<b>B</b>) Bar charts for each mouse species, corresponding to (<b>A</b>). (<b>C</b>) The flow cytometry results, reflecting the proportion of CD4-positive cells staining positive for TNFα in an independent experiment. (<b>D</b>) Corresponding bar charts for each mouse species. * = <span class="html-italic">p</span> &lt; 0.05, ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, and **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with sample sizes of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left corner of each example graph.</p>
Full article ">Figure 6
<p>(<b>A</b>) Flow cytometry results, reflecting the proportion of CD206-positive cells in an experiment with CD11b-positive cells. (<b>B</b>) The left panel shows the bar graph corresponding to (<b>A</b>). The right panel shows the flow cytometry results, reflecting the proportion of CD86-positive cells in the same experiment. The right panel shows the bar graph corresponding to the flow cytometry results, reflecting the proportion of CD86-positive cells in the same experiment. (<b>C</b>) An example of an immunohistochemical section of skin infiltrated with F4/80-positive cells. (<b>D</b>) The upper panel is the bar chart corresponding to (<b>C</b>) for each type of mouse skin. In the lower left and lower right are bars reflecting the infiltration of CD86-positive cells and CD206-positive cells in the skin of each of the four mouse species, respectively. ** = <span class="html-italic">p</span> &lt; 0.01, *** = <span class="html-italic">p</span> &lt; 0.001, and **** = <span class="html-italic">p</span> &lt; 0.0001 for each type of mouse in each type of experiment, with a sample size of 4–5 mice. The bar denotes the mean SEM. The name of each type of mouse is shown in the upper left corner of each example plot.</p>
Full article ">
29 pages, 2013 KiB  
Review
Na+/K+-ATPase: More than an Electrogenic Pump
by Ruben G. Contreras, Antonio Torres-Carrillo, Catalina Flores-Maldonado, Liora Shoshani and Arturo Ponce
Int. J. Mol. Sci. 2024, 25(11), 6122; https://doi.org/10.3390/ijms25116122 - 1 Jun 2024
Viewed by 242
Abstract
The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, [...] Read more.
The sodium pump, or Na+/K+-ATPase (NKA), is an essential enzyme found in the plasma membrane of all animal cells. Its primary role is to transport sodium (Na+) and potassium (K+) ions across the cell membrane, using energy from ATP hydrolysis. This transport creates and maintains an electrochemical gradient, which is crucial for various cellular processes, including cell volume regulation, electrical excitability, and secondary active transport. Although the role of NKA as a pump was discovered and demonstrated several decades ago, it remains the subject of intense research. Current studies aim to delve deeper into several aspects of this molecular entity, such as describing its structure and mode of operation in atomic detail, understanding its molecular and functional diversity, and examining the consequences of its malfunction due to structural alterations. Additionally, researchers are investigating the effects of various substances that amplify or decrease its pumping activity. Beyond its role as a pump, growing evidence indicates that in various cell types, NKA also functions as a receptor for cardiac glycosides like ouabain. This receptor activity triggers the activation of various signaling pathways, producing significant morphological and physiological effects. In this report, we present the results of a comprehensive review of the most outstanding studies of the past five years. We highlight the progress made regarding this new concept of NKA and the various cardiac glycosides that influence it. Furthermore, we emphasize NKA’s role in epithelial physiology, particularly its function as a receptor for cardiac glycosides that trigger intracellular signals regulating cell–cell contacts, proliferation, differentiation, and adhesion. We also analyze the role of NKA β-subunits as cell adhesion molecules in glia and epithelial cells. Full article
(This article belongs to the Special Issue Ion Pumps: Molecular Mechanisms, Structure, Physiology)
Show Figures

Figure 1

Figure 1
<p>Structure and function of NKA pump. (<b>A</b>) The diagram depicts the conformational changes of various domains within the α subunit of NKA, along with the associated ligands for each transition. The red circled arrow denotes the physiological progression of the Post-Alberts cycle. (<b>B</b>) Representative crystal images demonstrate structural disparities between the two primary stages, E1 and E2 (created from PB IDs 3WGV and 7WYT [<a href="#B78-ijms-25-06122" class="html-bibr">78</a>,<a href="#B79-ijms-25-06122" class="html-bibr">79</a>]). (<b>C</b>) The topological profile of the α subunit is depicted to highlight its intracellular domains: actuator (A), nucleotide binding (N), and phosphorylation (P) domains.</p>
Full article ">Figure 2
<p>Cardiac glycosides. The figure illustrates the basic chemical structure of CGs, comprising a steroid group with attached sugar and lactone groups, with each containing either five or six carbons. Additionally, it highlights the origins of notable CGs, such as ouabain, digoxin, and marinobufagenin.</p>
Full article ">Figure 3
<p>Different signaling pathways activated by the binding of CGs and ROS to NKS. (<b>A</b>) The diagram depicts several signaling pathways, including the positive inotropic effect shown in the left inset, as well as those activated by SRC and PI3K transactivation within caveolae. Each signaling pathway is represented by arrows of varying colors. Additionally, the diagram includes the representation of the ROS amplification loop. (<b>B</b>) The diagram illustrates the generation of intracellular calcium oscillations and the involvement of molecular components, such as IP3R, ORAi1, and STIM1, along with voltage-dependent calcium channels (VDCCs). Arrows indicate the pathways. The “X” on NCX and NKA indicate diminished activity.</p>
Full article ">Figure 4
<p>Ouabain–NKA-induced changes in epithelial cultured cells MDCK. The figure outlines the diverse signaling pathways triggered by the binding of ouabain, at nanomolar concentrations, to NKA. It illustrates the resulting effects on cellular components, including voltage-dependent potassium ion channels (VDKC) and TRPV4 channels, as well as the acceleration of ciliogenesis.</p>
Full article ">
12 pages, 775 KiB  
Systematic Review
Triple Negative Breast Cancer: Molecular Subtype-Specific Immune Landscapes with Therapeutic Implications
by Antonia Syrnioti, Stamatios Petousis, Lisa A. Newman, Chrysoula Margioula-Siarkou, Theodora Papamitsou, Konstantinos Dinas and Triantafyllia Koletsa
Cancers 2024, 16(11), 2094; https://doi.org/10.3390/cancers16112094 - 31 May 2024
Viewed by 186
Abstract
Triple Negative Breast Cancer (TNBC) is characterized by distinct molecular subtypes with unique biological and clinical features. This systematic review aimed to identify articles examining the differences in the tumor immune microenvironment (TIME) across different TNBC molecular subtypes. Six studies meeting inclusion criteria [...] Read more.
Triple Negative Breast Cancer (TNBC) is characterized by distinct molecular subtypes with unique biological and clinical features. This systematic review aimed to identify articles examining the differences in the tumor immune microenvironment (TIME) across different TNBC molecular subtypes. Six studies meeting inclusion criteria were analyzed, utilizing gene expression profiling and bioinformatic analyses to classify TNBC samples into molecular subtypes, as well as immunohistochemistry and cell deconvolution methods to characterize the TIME. Results revealed significant heterogeneity in immune cell composition among TNBC subtypes, with the immunomodulatory (IM) subtype demonstrating robust immune infiltration, composed mainly of adaptive immune cells along with an increased density of CTLA-4+ and PD-1+ TILs, high PD-L1 tumor cell expression, and upregulation of FOXP3+ Tregs. A more immunosuppressive TIME with a predominance of innate immune cells and lower levels of tumor-infiltrating lymphocytes (TILs) was observed in luminal androgen receptor (LAR) tumors. In mesenchymal stem-like (MSL) tumors, the TIME was mainly composed of innate immune cells, with a high number of M2 tumor-associated macrophages (TAMs), while the BL and M tumors displayed poor adaptive and innate immune responses, indicating an “immune-cold” phenotype. Differential activation of signaling pathways, genomic diversity, and metabolic reprogramming were identified as contributors to TIME heterogeneity. Understanding this interplay is crucial for tailoring therapeutic strategies, especially regarding immunotherapy. Full article
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the study selection process.</p>
Full article ">
23 pages, 2387 KiB  
Review
Hypoglycemic Drugs in Patients with Diabetes Mellitus and Heart Failure: A Narrative Review
by Anastasia Nikolaidou, Ioannis Ventoulis, Georgios Karakoulidis, Vasileios Anastasiou, Stylianos Daios, Spyridon-Filippos Papadopoulos, Matthaios Didagelos, John Parissis, Theodoros Karamitsos, Kalliopi Kotsa, Antonios Ziakas and Vasileios Kamperidis
Medicina 2024, 60(6), 912; https://doi.org/10.3390/medicina60060912 - 30 May 2024
Viewed by 202
Abstract
Over the last few years, given the increase in the incidence and prevalence of both type 2 diabetes mellitus (T2DM) and heart failure (HF), it became crucial to develop guidelines for the optimal preventive and treatment strategies for individuals facing these coexisting conditions. [...] Read more.
Over the last few years, given the increase in the incidence and prevalence of both type 2 diabetes mellitus (T2DM) and heart failure (HF), it became crucial to develop guidelines for the optimal preventive and treatment strategies for individuals facing these coexisting conditions. In patients aged over 65, HF hospitalization stands out as the predominant reason for hospital admissions, with their prognosis being associated with the presence or absence of T2DM. Historically, certain classes of glucose-lowering drugs, such as thiazolidinediones (rosiglitazone), raised concerns due to an observed increased risk of myocardial infarction (MI) and cardiovascular (CV)-related mortality. In response to these concerns, regulatory agencies started requiring CV outcome trials for all novel antidiabetic agents [i.e., dipeptidyl peptidase-4 inhibitors (DPP-4 inhibitors), glucagon-like peptide-1 receptor agonists (GLP-1 RAs), and sodium-glucose cotransporter-2 inhibitors (SGLT2is)] with the aim to assess the CV safety of these drugs beyond glycemic control. This narrative review aims to address the current knowledge about the impact of glucose-lowering agents used in T2DM on HF prevention, prognosis, and outcome. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

Figure 1
<p>Antidiabetic drugs’ impact on cardiovascular and heart failure outcomes. CV, cardiovascular; HF, heart failure; T2DM, type 2 diabetes mellitus; DPP-4i, dipeptidyl peptidase-4 inhibitor; GLP-1 RA, glucagon-like peptide-1 receptor agonist; SGLT2i, sodium-glucose cotransporter 2 inhibitor.</p>
Full article ">Figure 2
<p>Dipeptidyl peptidase-4 inhibitors’ impact on cardiovascular and heart failure outcomes [<a href="#B70-medicina-60-00912" class="html-bibr">70</a>]. DPP-4i, dipeptidyl peptidase-4 inhibitors CV, cardiovascular; HF, heart failure; LV, left ventricular; T2DM, type 2 diabetes mellitus; HFpEF, heart failure with preserved ejection fraction; HFmrEF, heart failure with mild reduced ejection fraction; HFrEF, heart failure with reduced ejection fraction; SE, stress echocardiography; CAD, coronary artery disease.</p>
Full article ">Figure 3
<p>Glucagon-like peptide-1 receptor agonists’ impact on cardiovascular and heart failure outcomes [<a href="#B97-medicina-60-00912" class="html-bibr">97</a>]. GLP-1 RA, glucagon-like peptide-1 receptor agonist; CV, cardiovascular; HF, heart failure; T2DM, type 2 diabetes mellitus; LV, left ventricular.</p>
Full article ">Figure 4
<p>Sodium-glucose cotransporter-2 inhibitors’ impact on cardiovascular and heart failure outcomes [<a href="#B120-medicina-60-00912" class="html-bibr">120</a>]. SGLT2i, sodium-glucose cotransporter-2 inhibitor; CV, cardiovascular; HF, heart failure; T2DM, type 2 diabetes mellitus; HFpEF, heart failure with preserved ejection fraction; HFmrEF, heart failure with mild reduced ejection fraction; HFrEF, heart failure with reduced ejection fraction; LV, left ventricular; CKD, chronic kidney disease.</p>
Full article ">Figure 5
<p>Indications of antidiabetic drugs in multiple heart failure phenotypes according to guidelines. CAD, coronary artery disease; HF, heart failure; DPP-4 inhibitors, dipeptidyl peptidase-4 inhibitors; GLP-1 RA, glucagon-like peptide-1 receptor agonist; SGLT2i, sodium-glucose cotransporter 2 inhibitor, TZDs, thiazolidinediones; HFpEF, heart failure with preserved ejection fraction; HFmrEF, heart failure with mild reduced ejection fraction; HFrEF, heart failure with reduced ejection fraction; CKD, chronic kidney disease.</p>
Full article ">
13 pages, 2019 KiB  
Article
Synergistic Steatosis Induction in Mice: Exploring the Interactions and Underlying Mechanisms between PFOA and Tributyltin
by Yannick Dauwe, Lucile Mary, Fabiana Oliviero, Louise Dubois, Elodie Rousseau-Bacquie, Jelskey Gomez, Véronique Gayrard and Laïla Mselli-Lakhal
Cells 2024, 13(11), 940; https://doi.org/10.3390/cells13110940 - 30 May 2024
Viewed by 372
Abstract
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially [...] Read more.
This study explores the impact of environmental pollutants on nuclear receptors (CAR, PXR, PPARα, PPARγ, FXR, and LXR) and their heterodimerization partner, the Retinoid X Receptor (RXR). Such interaction may contribute to the onset of non-alcoholic fatty liver disease (NAFLD), which is initially characterized by steatosis and potentially progresses to steatohepatitis and fibrosis. Epidemiological studies have linked NAFLD occurrence to the exposure to environmental contaminants like PFAS. This study aims to assess the simultaneous activation of nuclear receptors via perfluorooctanoic acid (PFOA) and RXR coactivation via Tributyltin (TBT), examining their combined effects on steatogenic mechanisms. Mice were exposed to PFOA (10 mg/kg/day), TBT (5 mg/kg/day) or a combination of them for three days. Mechanisms underlying hepatic steatosis were explored by measuring nuclear receptor target gene and lipid metabolism key gene expressions, by quantifying plasma lipids and hepatic damage markers. This study elucidated the involvement of the Liver X Receptor (LXR) in the combined effect on steatosis and highlighted the permissive nature of the LXR/RXR heterodimer. Antagonistic effects of TBT on the PFOA-induced activation of the Pregnane X Receptor (PXR) and Peroxisome Proliferator-Activated Receptor Gamma (PPARγ) were also observed. Overall, this study revealed complex interactions between PFOA and TBT, shedding light on their combined impact on liver health. Full article
(This article belongs to the Collection Functions of Nuclear Receptors)
Show Figures

Figure 1

Figure 1
<p>Histologic hepatic steatosis assessment. Histology was conducted on frozen liver sections stained with Harris Haematoxylin and red oil stain. (<b>A</b>) An accumulation of multiple small red droplets in the cytoplasm allows to assess steatosis. Zoom focus: 63×. (<b>B</b>) Optic quantification and morphological assessment of steatosis. Area of lipid droplets and droplet diameters are presented as mean ± standard error of the mean. The <span class="html-italic">p</span>-values indicate the level of significance, with * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 being calculated via the one-way ANOVA test followed by Tukey’s multiple comparisons test.</p>
Full article ">Figure 2
<p>Nuclear receptor activation in the liver. RT-qPCR analysis was performed on (<b>A</b>) CAR prototypical target genes (<span class="html-italic">Cyp2b10</span> and <span class="html-italic">Cyp2c29</span>); (<b>B</b>) PPARα target genes <span class="html-italic">Cyp4a10</span> and <span class="html-italic">Cyp4a14</span>; (<b>C</b>) PXR prototypical target gene <span class="html-italic">Cyp3a11</span>; (<b>D</b>) <span class="html-italic">Pparγ</span> expression; (<b>E</b>) LXR target gene <span class="html-italic">Cyp7a1</span>; (<b>F</b>) FXR target gene <span class="html-italic">Abcb11</span>. The results are presented as a graph, showing the expression levels in fold changes of the DMSO group. The data are presented as mean ± standard error of the means. The statistical analysis used was a one-way ANOVA test followed by Tukey’s multiple comparisons test. The <span class="html-italic">p</span>-values indicate the level of significance, with * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 indicating significant differences.</p>
Full article ">Figure 3
<p>RT-qPCR of genes involved in lipid metabolism and transport. RT-qPCR of genes involved in (<b>A</b>) lipid synthesis and lipid droplet formation, (<b>B</b>) in fatty acid β-oxidation, (<b>C</b>) in fatty acid and triglyceride transport, and (<b>D</b>) in cholesteryl ester uptake. The results are expressed as fold changes of the DMSO group. Data are presented as mean ± standard error of the mean. * <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. <span class="html-italic">p</span>-values represent significant differences between each treatment group according to one-way ANOVA test followed by Tukey’s multiple comparisons test. # for <span class="html-italic">p</span> &lt; 0.05 represents a significant difference, with additivity calculated using a one-sample t-test.</p>
Full article ">
26 pages, 4577 KiB  
Review
Novel Insights into Psychosis and Antipsychotic Interventions: From Managing Symptoms to Improving Outcomes
by Adonis Sfera, Hassan Imran, Dan O. Sfera, Jacob J. Anton, Zisis Kozlakidis and Sabine Hazan
Int. J. Mol. Sci. 2024, 25(11), 5904; https://doi.org/10.3390/ijms25115904 (registering DOI) - 28 May 2024
Viewed by 233
Abstract
For the past 70 years, the dopamine hypothesis has been the key working model in schizophrenia. This has contributed to the development of numerous inhibitors of dopaminergic signaling and antipsychotic drugs, which led to rapid symptom resolution but only marginal outcome improvement. Over [...] Read more.
For the past 70 years, the dopamine hypothesis has been the key working model in schizophrenia. This has contributed to the development of numerous inhibitors of dopaminergic signaling and antipsychotic drugs, which led to rapid symptom resolution but only marginal outcome improvement. Over the past decades, there has been limited research on the quantifiable pathological changes in schizophrenia, including premature cellular/neuronal senescence, brain volume loss, the attenuation of gamma oscillations in electroencephalograms, and the oxidation of lipids in the plasma and mitochondrial membranes. We surmise that the aberrant activation of the aryl hydrocarbon receptor by toxins derived from gut microbes or the environment drives premature cellular and neuronal senescence, a hallmark of schizophrenia. Early brain aging promotes secondary changes, including the impairment and loss of mitochondria, gray matter depletion, decreased gamma oscillations, and a compensatory metabolic shift to lactate and lactylation. The aim of this narrative review is twofold: (1) to summarize what is known about premature cellular/neuronal senescence in schizophrenia or schizophrenia-like disorders, and (2) to discuss novel strategies for improving long-term outcomes in severe mental illness with natural senotherapeutics, membrane lipid replacement, mitochondrial transplantation, microbial phenazines, novel antioxidant phenothiazines, inhibitors of glycogen synthase kinase-3 beta, and aryl hydrocarbon receptor antagonists. Full article
(This article belongs to the Section Molecular Neurobiology)
Show Figures

Figure 1

Figure 1
<p>In the cytosol, the AhR is stabilized by two HSP90 molecules. DA, oxidized lipids (and toxic ceramide), clozapine, serotonin, melatonin, and vitamin D3 are AhR ligands [<a href="#B95-ijms-25-05904" class="html-bibr">95</a>,<a href="#B96-ijms-25-05904" class="html-bibr">96</a>]. Pollutants, such as phthalate and bisphenol A (BPA), are also AhR ligands. In contrast, aripiprazole binds to the AhR chaperone, HSP90. HSP90 prevents AhR’s entry into the nucleus where it drives the transcription of genes, including those for cellular senescence.</p>
Full article ">Figure 2
<p>Glial cells, including astrocytes, supply neurons with healthy mitochondria via tunneling nanotubules, preventing apoptosis [<a href="#B137-ijms-25-05904" class="html-bibr">137</a>,<a href="#B138-ijms-25-05904" class="html-bibr">138</a>]. In addition, astrocytes prevent neuronal ferroptosis by transferring antioxidants, including GPX-4. Astrocytes uptake cystine via the cystine/glutamate antiporter (Xc<sup>−</sup>). Cysteine can also be obtained from methionine via glutathione. Fe<sup>2+</sup> enters neurons through the transferrin receptor-1 (TRF-1), which is stored in ferritin and requires ferritinophagy to be released. Excess Fe<sup>2+</sup> exits the neurons via ferroportin (FPT) channels.</p>
Full article ">Figure 3
<p>The AhR is represented in the cytosol and mitochondria (mitoAhR). Akt negatively phosphorylates GSK-3β, inhibiting its function. Toxic ceramides and iron activate GSK-3β, resulting in excessive mitochondrial ROS (mtROS) levels, which activate the mitoAhR, triggering organelle death. mtROS can also cause mitochondrial demise by activating cytochrome-C and caspase-3. The natural compounds berberine and kaempferol inhibit GSK-3β, averting organelle death.</p>
Full article ">Figure 4
<p>The lipid bilayer of neuronal membranes is easily oxidated when intracellular iron is upregulated [<a href="#B182-ijms-25-05904" class="html-bibr">182</a>]. Oxysterols, including 7-ketocholesterol (a toxic oxide), and oxidated phospholipids alter the biophysical properties of cell membranes, disrupting neurotransmission [<a href="#B183-ijms-25-05904" class="html-bibr">183</a>]. In addition, oxidized lipids activate the AhR, triggering premature neuronal senescence. Phenazines, phenothiazines, and their derivatives intercalate themselves into the lipid bilayer, repairing the lipids in cellular and/or mitochondrial membranes.</p>
Full article ">Figure 5
<p>Phenazine vs. phenothiazine: similarities and differences.</p>
Full article ">Figure 6
<p>Membrane lipid replacement (MLR) replenishes oxidized lipids from the plasma and mitochondrial membrane, such as oxysterols, ceramide, and oxidized phospholipids (OxPLs) with natural glycerophospholipids. Oxidized lipids inhibit AKT (by serine-9 phosphorylation), activating GSK-3β, an enzyme associated with SCZ, SLDs, and cancer (by p53 inhibition). Berberine and kaempferol inhibit GSK-3β activation by different mechanisms, generating beneficial effects. Ceramide activates GSK-3β by the dephosphorylation of serine-9.</p>
Full article ">Figure 7
<p>AhR agonists and antagonists relevant for neuropsychiatry [<a href="#B74-ijms-25-05904" class="html-bibr">74</a>,<a href="#B213-ijms-25-05904" class="html-bibr">213</a>,<a href="#B214-ijms-25-05904" class="html-bibr">214</a>,<a href="#B215-ijms-25-05904" class="html-bibr">215</a>,<a href="#B216-ijms-25-05904" class="html-bibr">216</a>,<a href="#B217-ijms-25-05904" class="html-bibr">217</a>,<a href="#B218-ijms-25-05904" class="html-bibr">218</a>,<a href="#B219-ijms-25-05904" class="html-bibr">219</a>,<a href="#B220-ijms-25-05904" class="html-bibr">220</a>,<a href="#B221-ijms-25-05904" class="html-bibr">221</a>,<a href="#B222-ijms-25-05904" class="html-bibr">222</a>].</p>
Full article ">
16 pages, 325 KiB  
Review
Effects of Endocrine Interventions Targeting ERα or PR on Breast Cancer Risk in the General Population and Carriers of BRCA1/2 Pathogenic Variants
by Deborah Huber, Maria Hatzipanagiotou, Susanne Schüler-Toprak, Olaf Ortmann and Oliver Treeck
Int. J. Mol. Sci. 2024, 25(11), 5894; https://doi.org/10.3390/ijms25115894 - 28 May 2024
Viewed by 294
Abstract
There is evidence suggesting that endocrine interventions such as hormone replacement therapy and hormonal contraception can increase breast cancer (BC) risk. Sexual steroid hormones like estrogens have long been known for their adverse effects on BC development and progression via binding to estrogen [...] Read more.
There is evidence suggesting that endocrine interventions such as hormone replacement therapy and hormonal contraception can increase breast cancer (BC) risk. Sexual steroid hormones like estrogens have long been known for their adverse effects on BC development and progression via binding to estrogen receptor (ER) α. Thus, in recent years, endocrine interventions that include estrogens have been discussed more and more critically, and their impact on different BC subgroups has increasingly gained interest. Carriers of pathogenic variants in BRCA1/2 genes are known to have a high risk of developing BC and ovarian cancer. However, there remain open questions to what extent endocrine interventions targeting ERα or the progesterone receptor further increase cancer risk in this subgroup. This review article aims to provide an overview and update on the effects of endocrine interventions on breast cancer risk in the general population in comparison to BRCA1/2 mutation carriers. Finally, future directions of research are addressed, to further improve the understanding of the effects of endocrine interventions on high-risk pathogenic variant carriers. Full article
(This article belongs to the Special Issue The Role of Estrogen Receptors in Health and Diseases)
29 pages, 4467 KiB  
Article
Impact of Interleukin-17 Receptor A Gene Variants on Asthma Susceptibility and Clinical Manifestations in Children and Adolescents
by Shymaa Ahmed Maher, Nouran B. AbdAllah, Essam Al Ageeli, Eman Riad, Shahad W. Kattan, Sherouk Abdelaal, Wagdy Abdelfatah, Gehan A. Ibrahim, Eman A. Toraih, Ghada A. Awadalla, Manal S. Fawzy and Ahmed Ibrahim
Children 2024, 11(6), 657; https://doi.org/10.3390/children11060657 - 28 May 2024
Viewed by 300
Abstract
Several single nucleotide polymorphisms (SNPs) in multiple interleukin receptor genes could be associated with asthma risk and/or phenotype. Interleukin-17 (IL-17) has been implicated in tissue inflammation and autoimmune diseases. As no previous studies have uncovered the potential role of IL17 receptor A ( [...] Read more.
Several single nucleotide polymorphisms (SNPs) in multiple interleukin receptor genes could be associated with asthma risk and/or phenotype. Interleukin-17 (IL-17) has been implicated in tissue inflammation and autoimmune diseases. As no previous studies have uncovered the potential role of IL17 receptor A (RA) gene variants in asthma risk, we aimed to explore the association of four IL17RA SNPs (i.e., rs4819554A/G, rs879577C/T, rs41323645G/A, and rs4819555C/T) with asthma susceptibility/phenotype in our region. TaqMan allelic discrimination analysis was used to genotype 192 individuals. We found that the rs4819554 G/G genotype significantly reduced disease risk in the codominant (OR = 0.15, 95%CI = 0.05–0.45, p < 0.001), dominant (OR = 0.49, 95%CI = 0.26–0.93, p = 0.028), and recessive (OR = 0.18, 95%CI = 0.07–0.52, p < 0.001) models. Similarly, rs879577 showed reduced disease risk associated with the T allele across all genetic models. However, the A allele of rs41323645 was associated with increased disease risk in all models. The G/A and A/A genotypes have higher ORs of 2.47 (95%CI = 1.19–5.14) and 3.86 (95%CI = 1.62–9.18), respectively. Similar trends are observed in the dominant 2.89 (95%CI = 1.47–5.68, p = 0.002) and recessive 2.34 (95%CI = 1.10–4.98, p = 0.025) models. For the rs4819555 variant, although there was no significant association identified under any models, carriers of the rs4819554*A demonstrated an association with a positive family history of asthma (71.4% in carriers vs. 27% in non-carriers; p = 0.025) and the use of relievers for >2 weeks (52.2% of carriers vs. 28.8% of non-carriers; p = 0.047). Meanwhile, the rs4819555*C carriers displayed a significant divergence in the asthma phenotype, specifically atopic asthma (83.3% vs. 61.1%; p = 0.007), showed a higher prevalence of chest tightness (88.9% vs. 61.5%; p = 0.029), and were more likely to report comorbidities (57.7% vs. 16.7%, p = 0.003). The most frequent haplotype in the asthma group was ACAC, with a frequency of 22.87% vs. 1.36% in the controls (p < 0.001). In conclusion, the studied IL17RA variants could be essential in asthma susceptibility and phenotype in children and adolescents. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Interleukin-17 signaling pathway (Kyoto Encyclopedia of Genes and Genomes (KEGG) ID: Human (Homo sapiens), hsa04657). The IL-17 cytokine family (IL-17A-F) plays a role in acute and chronic inflammation. IL-17A, predominantly secreted by T helper 17 (TH17) cells, is essential for protecting against external pathogens and is implicated in autoimmune inflammation. IL-17F primarily protects mucosal surfaces, while IL-17E promotes Th2 immune responses. IL-17C shares similar functions with IL-17A. The IL-17 family communicates via specific receptors, such as IL17RA, which several family members use. Activation of downstream signaling pathways, including NF-kappa B, mitogen-activated protein kinases (MAPKs), and CCAAT-enhancer-binding proteins (C/EBPs), results in the production of antimicrobial peptides and the expression of cytokines and chemokines. Adopted from (<a href="https://www.genome.jp/pathway/hsa04657+23765" target="_blank">https://www.genome.jp/pathway/hsa04657+23765</a>) (last acceded 7 July 2023) [<a href="#B9-children-11-00657" class="html-bibr">9</a>,<a href="#B10-children-11-00657" class="html-bibr">10</a>].</p>
Full article ">Figure 2
<p>Molecular features of interleukin-17RA (<span class="html-italic">IL17RA)</span> gene and protein. (<b>A</b>) The <span class="html-italic">IL17RA</span> gene (Gene ID: 23765) is located on the long arm of chromosome 22-NC_000022.11:17,084,954-17,115,693 according to Homo sapiens assembly GRCh38.p14, with 30,740 bp long, on the forward strand. It consists of 13 exons. (<b>B</b>) The gene has eight transcripts: the major one (IL17RA-201 transcript; ENST00000319363.11) has 8566 nucleotides, which codes for 866aa protein; the IL17RA-204 (2499 nucleotides) codes for the 832aa protein, the IL17RA-207 (739 nucleotides) codes for the129aa nonsense-mediated decay transcript, the IL17RA-203 (651 nucleotides) codes for the 59aa nonsense-mediated decay transcript, and the IL17RA-206, 205, 208, and 202 (675, 611, 847, and 621 nucleotides, respectively) are non-protein coding transcripts. (<b>C</b>) The encoded protein is a single polypeptide chain of 866 amino acid residues, starting with the signal peptide (1–32 aa, red color) in the amino terminus, the extracellular domain (33–320aa), the transmembrane domain (321–341aa), and the cytoplasmic domain (342–866aa). The studied missense variants-related amino acid substitutions were labeled in the cytoplasmic domain and explained in the legend with the specified colors. (<b>D</b>) The subcellular localization of IL17RA can be observed, with the intensity of green coloration indicating its relative abundance. (<b>E</b>) The interacting protein–protein network demonstrates the functional partners of the IL17RA. The generated network consists of 11 nodes (each node corresponds to a protein generated from a unique gene locus) and 45 edges (each edge signifies an association between proteins, which could imply sharing functionality but does not necessarily mean the proteins are physically bound to each other), with average local clustering coefficient = 0.889, and the protein–protein interaction enrichment <span class="html-italic">p</span>-value = 4.22 × 10<sup>−15</sup>. (<b>F</b>) <span class="html-italic">IL17RA</span> gene coexpression triangle matrix. The confidence level of association is demonstrated by the color intensity, given the overall expression data in humans. Data sources: “<a href="https://www.genecards.org/" target="_blank">https://www.genecards.org/</a>, <a href="https://www.ncbi.nlm.nih.gov/gene/23765" target="_blank">https://www.ncbi.nlm.nih.gov/gene/23765</a>, <a href="https://asia.ensembl.org/Homo_sapiens/Tran-script/" target="_blank">https://asia.ensembl.org/Homo_sapiens/Tran-script/</a>, <a href="https://wlab.ethz.ch/protter/" target="_blank">https://wlab.ethz.ch/protter/</a>, <a href="https://www.uniprot.org/" target="_blank">https://www.uniprot.org/</a>, <a href="https://compartments.jensenlab.org/" target="_blank">https://compartments.jensenlab.org/</a>, and <a href="https://string-db.org/" target="_blank">https://string-db.org/</a>” (last accessed on 20 July 2023).</p>
Full article ">
Back to TopTop