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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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 = nuclear translocation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
26 pages, 7287 KiB  
Article
Mitochondrial Genome Insights into Evolution and Gene Regulation in Phragmites australis
by Jipeng Cui, Qianhui Yang, Jiyue Zhang, Chuanli Ju and Suxia Cui
Int. J. Mol. Sci. 2025, 26(2), 546; https://doi.org/10.3390/ijms26020546 - 10 Jan 2025
Viewed by 126
Abstract
As a globally distributed perennial Gramineae, Phragmites australis can adapt to harsh ecological environments and has significant economic and environmental values. Here, we performed a complete assembly and annotation of the mitogenome of P. australis using genomic data from the PacBio and BGI [...] Read more.
As a globally distributed perennial Gramineae, Phragmites australis can adapt to harsh ecological environments and has significant economic and environmental values. Here, we performed a complete assembly and annotation of the mitogenome of P. australis using genomic data from the PacBio and BGI platforms. The P. australis mitogenome is a multibranched structure of 501,134 bp, divided into two circular chromosomes of 325,493 bp and 175,641 bp, respectively. A sequence-simplified succinate dehydrogenase 4 gene was identified in this mitogenome, which is often translocated to the nuclear genome in the mitogenomes of gramineous species. We also identified tissue-specific mitochondrial differentially expressed genes using RNAseq data, providing new insights into understanding energy allocation and gene regulatory strategies in the long-term adaptive evolution of P. australis mitochondria. In addition, we studied the mitogenome features of P. australis in more detail, including repetitive sequences, gene Ka/Ks analyses, codon preferences, intracellular gene transfer, RNA editing, and multispecies phylogenetic analyses. Our results provide an essential molecular resource for understanding the genetic characterisation of the mitogenome of P. australis and provide a research basis for population genetics and species evolution in Arundiaceae. Full article
(This article belongs to the Special Issue Transcriptional Regulation in Plant Development: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">P. australis</span> mitochondrial gene assembly process and structural characterisation. (<b>a</b>) Organelle genome assembly process from genome sequencing data. (<b>b</b>) <span class="html-italic">P. australis</span> mitogenome master graphs. (<b>c</b>) Two circle graphs were obtained by hybrid assembly using Unicycler. Each circle graph represents the ring molecules of one chromosome of the mitogenome. Fragment colours indicate the same contig sequence fragments as in the master graphs. (<b>d</b>) Basic mitogenome information.</p>
Full article ">Figure 2
<p>A schematic map of the mitogenome of <span class="html-italic">P. australis</span> and the identification of the succinate dehydrogenase subunit gene. (<b>a</b>) Schematic maps of the two circular chromosomes of the <span class="html-italic">P. australis</span> mitogenome, with the colour used to distinguish genes of different functional groups. The images illustrate the sequence of events from the inside out. (1) The relationship between dispersed repeat sequences. (2) The distribution of dispersed repeat sequences on the chromosome where yellow represents direct dispersed repeat sequences and green represents inverted dispersed repeat sequences. (3) The distribution of tandem repeat sequences on the chromosome. (4) The distribution of tandem repeat sequences. (5) The distribution of GC content on the chromosome. (6) The scale coordinate axis. (7) Genes located on the negative strand. (8) Genes located on the positive strand. (9) Orange shadows represent forward dispersed repeats that exceed the selection threshold. (<b>b</b>) Phylogenetic tree of the SDH gene family in the nuclear genome of <span class="html-italic">P. australis.</span> (<b>c</b>) Sequence comparison of 3 nuclear genome Nu_SDH4 transcripts with Mt_SDH4 in the mitogenome. (<b>d</b>) Structure prediction of Mt_SDH4 protein in 3 nuclear genome Nu_SDH4 transcripts versus mitogenome.</p>
Full article ">Figure 3
<p>Repetitive sequences in the <span class="html-italic">P. australis</span> mitogenome. (<b>a</b>) Types and numbers of SSRs in the mitogenome of <span class="html-italic">P. australis</span>. The number of repeats in each category is shown at the top of the corresponding bar diagram. (<b>b</b>) The frequency of classified repeat types (considering sequence complementary) in monomeric, dimeric, and trimeric repeat types. (<b>c</b>) Statistics of repeat sequence types in the mitogenome of <span class="html-italic">P. australis</span>. (<b>d</b>) The figure of the distribution of repetitive sequences in the mitogenome of <span class="html-italic">P. australis</span>. The orange colour indicates the distribution density of SSRs, blue circles indicate the distribution density of tandem repeat sequences, inside lines indicate dispersed repeat sequences, yellow lines indicate forward repeats (F), and green lines indicate palindromic repeats (P).</p>
Full article ">Figure 4
<p>Boxplot of Ka/Ks ratios of <span class="html-italic">P. australis</span> with 32 other Graminaceous species.</p>
Full article ">Figure 5
<p>Analysis of the <span class="html-italic">P. australis</span> mitogenome relative synonymous codon usage. The coloured blocks below indicate the type of codon encoding each amino acid, and the coloured blocks above are the RSCU values for the corresponding codons.</p>
Full article ">Figure 6
<p>A schematic map of the MTPTs and NUMTs of the mitogenome of <span class="html-italic">P. australis</span>. (<b>a</b>) A schematic map of the chloroplast genome assembled in this study. (<b>b</b>) A Circos plot showing the MTPTs between the mitochondrial and chloroplast genomes. Genes labelled in the figure indicate genes contained in the MTPT, and the red text indicates genes in the MTPT with unchanged functions in the mitochondrial and chloroplast genomes. (<b>c</b>) A Circos plot showing NUMTs between 25 chromosomes of nuclear genomes and the mitogenome of <span class="html-italic">P. australis</span>. The arcs in B and C connect homologous sequence fragments between different genomes.</p>
Full article ">Figure 7
<p>Characterisation of RNA editing sites in the mitogenome of <span class="html-italic">P. australis</span>. (<b>a</b>) Statistics of the number of predicted RNA editing sites in PCGs. (<b>b</b>) Amino acid changes caused by RNA editing in PCGs. * Indicates a codon that has been changed to a stop codon by RNA editing. (<b>c</b>) Location statistics of RNA editing in codon.</p>
Full article ">Figure 8
<p>The mtDEG in the mitochondrial transcriptome of <span class="html-italic">P. australis</span>. (<b>a</b>) Principal component analysis of the transcriptome of <span class="html-italic">P. australis</span> leaf, aerial stem, and rhizome tissues. (<b>b</b>–<b>d</b>) Volcano plots showing mtDEG between different tissues, respectively, |log2FoldChange| &gt; 1, <span class="html-italic">p</span> value &lt; 0.05.</p>
Full article ">Figure 9
<p>Phylogenetic relationships between <span class="html-italic">P. australis</span> and Gramineae. Phylogenetic trees (<b>a</b>,<b>b</b>) have been constructed using <span class="html-italic">P. australis</span> mitochondrial and chloroplast genomes, respectively, based on the maximum likelihood method. Phylogenetic trees (<b>c</b>,<b>d</b>) have been constructed using <span class="html-italic">P. australis</span> mitochondrial and chloroplast genomes, respectively, based on the Bayesian inference method. The topological differences in the phylogenetic trees constructed based on mitochondrial genomes (<b>left</b>) and chloroplast genomes (<b>right</b>) are shown using phytools (v 2.3-0). Branches connected by dotted lines indicate the matching of different genomic data of the same species in the phylogenetic tree. The use of coloured backgrounds allows for the clear identification of the species belonging to the subfamily to which they belong. Further details pertaining to the phylogenetic tree can be found in <a href="#app1-ijms-26-00546" class="html-app">Supplementary Figure S6</a>.</p>
Full article ">
15 pages, 3793 KiB  
Article
Panduratin A Inhibits TNF Alpha-Stimulated Endothelial Cell Activation Through Suppressing the NF-κB Pathway
by Kriangkrai Kiatsoonthon, Nitchakarn Phimthong, Saranyapin Potikanond, Nitwara Wikan and Wutigri Nimlamool
Biomolecules 2025, 15(1), 34; https://doi.org/10.3390/biom15010034 - 30 Dec 2024
Viewed by 427
Abstract
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway [...] Read more.
Upon exposure to inflammatory stimuli including TNF-α, endothelial cells are activated leading to the adhesion of monocytes to their surface. These events are involved in the pathophysiology of atherosclerosis. Since TNF-α activates the NF-κB pathway, which contributes to atherosclerosis, targeting this signaling pathway may help prevent the risk of developing the disease. The current study elucidated the inhibitory effect of panduratin A (PA) on TNF-α-induced endothelial activation and monocyte adhesion. We discovered that PA reduced the level of pro-inflammatory cytokine IL-6 and chemokine MCP-1 in the media collected from endothelial cells stimulated with TNF-α. In addition, PA inhibited the expression of ICAM-1 and VCAM-1 on the surface of TNF-α-induced endothelial cells resulting in a decrease in the number of monocytes attached to endothelial cell surface. Mechanistically, PA prevented IκB degradation and specifically suppressed NF-κB phosphorylation and nuclear translocation in endothelial cells. However, PA had no inhibitory effect on the phosphorylation of AKT, ERK1/2, p38, and JNK. Taken together, PA blocked the production of cytokine and chemokine, adhesion molecules, and monocyte adhesion in response to TNF-α stimulation, in part, through NF-κB inhibition. Our study suggests that PA may possibly be effective in blocking the pathophysiology of atherosclerosis. Full article
(This article belongs to the Special Issue The Value of Natural Compounds as Therapeutic Agents: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Effect of PA on the viability or proliferation of endothelial cells. Cells were treated with PA (at concentrations ranging from 0 to 10 μM) or DMSO (vehicle control) for 48 h prior to the cell viability test. Data are obtained from at least 3 individual experiments and presented as mean ± SD.</p>
Full article ">Figure 2
<p>Effect of PA on secretion of IL-6 and MCP-1 in the TNF-α-induced endothelial cell culture media. The level of IL-6 (<b>A</b>) and MCP-1 (<b>B</b>) in the media of the PA-treated cells stimulated with 10 TNF-α for 24 h evaluated by ELISA. Data are presented as mean ± SD * <span class="html-italic">p</span> &lt; 0.05 (compared with the DMSO-treated group).</p>
Full article ">Figure 3
<p>PA decreased TNF-α- induced expression of vascular adhesion molecules on endothelial cell surface. Endothelial cells (untreated, TNF-α-simulated, or TNF-α-simulated with the presence of PA at 4 μM for 24 h) were subject to immunofluorescence for detecting VCAM-1 (<b>A</b>) and ICAM-1 (<b>B</b>). Nuclei (blue) were visualized by DAPI staining. The micrographs were captured at 100× magnification.</p>
Full article ">Figure 4
<p>Effects of TNF-α and PA on the morphology and cytoskeletal arrangement of endothelial cells: (<b>A</b>) Phase-contrast microscopy visualizing endothelial cells without any treatment (Untreated), induced with TNF-α, or induced with TNF-α with the presence of 4 μM of PA at 0, 12, and 24 h (Scale bar = 300 μm). Immunofluorescence study detecting α-tubulin (<b>B</b>), β-catenin (<b>C</b>), and vimentin (<b>D</b>) in endothelial cells after treatment for 24 h. Cells were also stained for F-actin (red) and nuclei (blue). The micrographs (immunofluorescence study) were captured at 100× magnification.</p>
Full article ">Figure 5
<p>The effect of PA on THP-1 cell binding to TNF-α-activated endothelial cells. Adhesion of the labeled THP-1 monocytes to endothelial cells (untreated, induced with TNF-α, induced with TNF-α with the presence of PA at 4 μM, or induced with TNF-α with the presence of DMSO (vehicle control) for 24 h) was visualized and quantified by using a live-cell imager: (<b>A</b>) Phase-contrast microscopy showing THP-1 monocytes bound to endothelial monolayer, and fluorescent microscopy showing the nuclei (blue) of THP-1 monocytes. Scale bar = 300 μm. (<b>B</b>) Quantitative analysis of THP-1 cells bound to endothelial cells. Data are expressed as mean ± SD. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>The effect of PA on TNF-α-activated phosphorylation of NF-κB (pNF-κB) and the degradation of IκB. (<b>A</b>) Western blot analysis showing the level of pNF-κB and IκB in cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO (vehicle control). (<b>B</b>) Densitometry of pNF-κB immunoreactive bands from cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO. (<b>C</b>) Densitometry of IκB immunoreactive bands from cells without any treatment, TNF-α induction, and TNF-α induction with the presence of PA, or TNF-α induction with the presence of DMSO. * <span class="html-italic">p</span> &lt; 0.05. Original images of (<b>A</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 7
<p>The effect of PA on inhibiting TNF-α-activated nuclear relocalization of NF-κB and IκB degradation: (<b>A</b>) The presence of NF-κB in the nuclear extract of TNF-α-activated endothelial cells with or without the presence of PA. (<b>B</b>) Densitometric analysis of NF-κB immunoreactive bands from the Western blot of the nuclear extracts. (<b>C</b>) Immunofluorescence study visualizing relocalization of NF-κB into the nucleus of TNF-α-activated endothelial cells with or without the presence of PA. (<b>D</b>) Western blot analysis detecting IκB in the cytoplasmic extracts of TNF-α-activated endothelial cells with or without the presence of PA. (<b>E</b>) Densitometric analysis of IκB immunoreactive bands from the Western blot of the cytoplasmic extracts. (<b>F</b>) Immunofluorescence study visualizing the existence of IκB in the cytosol of TNF-α-activated endothelial cells with or without the presence of PA. * <span class="html-italic">p</span> &lt; 0.05. The micrographs were captured at 100× magnification. Original images of (<b>A</b>,<b>D</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 8
<p>The effect of PA on the phosphorylation status of important molecular markers for growth and survival: (<b>A</b>) Western blot determining the level of pAKT. (<b>B</b>) Quantitative analysis of pAKT immunoreactive bands. (<b>C</b>) Western blot determining the level of pERK1/2. (<b>D</b>) Quantitative analysis of pERK1/2 immunoreactive bands. (<b>E</b>) Western blot analysis detecting pp38. (<b>F</b>) Quantitative analysis of pp38 immunoreactive bands. (<b>G</b>) Western blot analysis detecting pJNK. (<b>H</b>) Quantitative analysis of pJNK immunoreactive bands. * <span class="html-italic">p</span> &lt; 0.05. Original images of (<b>A</b>,<b>C</b>,<b>E</b>,<b>G</b>) can be found in <a href="#app1-biomolecules-15-00034" class="html-app">Supplementary Materials</a>.</p>
Full article ">Figure 9
<p>The proposed model for the mechanism of PA in reducing TNF-α-stimulated endothelial activation and monocyte adhesion. This figure was created with an approval from <a href="http://BioRender.com" target="_blank">BioRender.com</a> (accessed on 28 November 2024).</p>
Full article ">
15 pages, 4063 KiB  
Article
HDAC1 and HDAC2 Are Involved in Influenza A Virus-Induced Nuclear Translocation of Ectopically Expressed STAT3-GFP
by Jessica Leong and Matloob Husain
Viruses 2025, 17(1), 33; https://doi.org/10.3390/v17010033 - 29 Dec 2024
Viewed by 470
Abstract
Influenza A virus (IAV) remains a pandemic threat. Particularly, the evolution and increased interspecies and intercontinental transmission of avian IAV H5N1 subtype highlight the importance of continuously studying the IAV and identifying the determinants of its pathogenesis. Host innate antiviral response is the [...] Read more.
Influenza A virus (IAV) remains a pandemic threat. Particularly, the evolution and increased interspecies and intercontinental transmission of avian IAV H5N1 subtype highlight the importance of continuously studying the IAV and identifying the determinants of its pathogenesis. Host innate antiviral response is the first line of defense against IAV infection, and the transcription factor, the signal transducer and activator of transcription 3 (STAT3), has emerged as a critical component of this response. Also, histone deacetylase 1 (HDAC1) and HDAC2 have been identified as important components of IAV-induced host innate antiviral response. Upon IAV infection, STAT3 is activated and translocated to the nucleus to initiate the transcription of innate response genes. Also, the HDAC1 and HDAC2 are localized to the nucleus. In this study, we sought to investigate the role of HDAC1 and HDAC2 in IAV-induced STAT3 nuclear translocation. We employed a quantitative confocal microscopy approach and analyzed the nuclear translocation of plasmid-expressed STAT3-GFP in IAV-infected cells depleted with the expression of HDAC1 or HDAC2. We found that the depletion of both HDAC1 and HDAC2 expression inhibits the IAV-induced nuclear translocation of STAT3-GFP. These findings will help elucidate the significance of the emerging role of acetylation in IAV infection and disease severity. Full article
(This article belongs to the Special Issue Interplay between Influenza Virus and Host Factors)
Show Figures

Figure 1

Figure 1
<p>Plasmid-expressed STAT3-GFP translocates to the nucleus in response to interferon treatment. HeLa cells were transfected with plasmid pEGFP-N1-STAT3 for 48 h, and then treated with IFN-α, IFN-β, or IFN-γ (500 IU). After 1 h, cells were fixed and stained with DAPI (4′,6-diamidino-2-phenylindole), and then imaged using a confocal microscope under 40× magnification. Plot profiles of cells treated with IFN-α, IFN-β, and IFN-γ (next to respective rows) were generated using ImageJ 1.54g; red lines in Merged column indicate the profiled area; x-axes and y-axes on graphs represent distance and pixel intensity, respectively, along the lines; green and blue lines on graphs represent plot profiles of STAT3-GFP and DAPI, respectively.</p>
Full article ">Figure 2
<p>Plasmid-expressed STAT3-GFP translocates to the nucleus in response to IAV infection. HeLa cells were transfected with plasmid pEGFP-N1-STAT3 for 48 h, and then infected with influenza virus A/Puerto Rico/8/1934(H1N1), hereafter referred to as PR8, at the multiplicity of infection (MOI) of 1.0, 3.0, or 5.0. After 6 h, cells were fixed, permeabilized, and stained with Goat anti-IAV nucleoprotein (NP) antibody followed by Donkey anti-Goat IgG conjugated with Alexa Fluor 647. Subsequently, cells were stained with DAPI and imaged using a confocal microscope under 40× magnification. Plot profiles of uninfected (UNI) and infected (INF) cells (next to respective rows) were generated using ImageJ 1.54g; red lines in Merged column indicate the profiled area; x-axes and y-axes on graphs represent distance and pixel intensity, respectively, along the lines; green and blue lines on graphs represent plot profiles of STAT3-GFP and DAPI, respectively.</p>
Full article ">Figure 3
<p>The IAV-induced nuclear translocation of STAT3-GFP is inhibited in HDAC1-depleted cells. HeLa cells were first transfected with either control siRNA or HDAC1 siRNA for 30 h and then transfected with plasmid pEGFP-N1-STAT3 for a further 30 h. Cells were then infected with PR8 at MOI of 3.0. After 6 h, cells were fixed, permeabilized, and stained with Mouse anti-HDAC1 antibody followed by Donkey anti-Mouse IgG conjugated with Alexa Fluor 594. Subsequently, cells were stained with Goat anti-NP antibody followed by Donkey anti-Goat IgG conjugated with Alexa Fluor 647. Finally, cells were stained with DAPI and imaged using a confocal microscope under 40× magnification. UNI, uninfected; INF, infected.</p>
Full article ">Figure 4
<p>Western blots showing the siRNA-mediated depletion of HDAC1 and HDAC2 expression in uninfected and infected cells. HeLa cells were transfected with either control (CT), HDAC1 (HD1), or HDAC2 (HD2) siRNA for 30 h. Cells were then infected with PR8 at MOI of 3.0. After 6 h, HDAC1 (62 kDa), HDAC2 (60 kDa), NP (56 kDa), and Actin (42 kDa) were detected in total cell lysates by Western blotting. UNI, uninfected; INF, infected; kDa, kilodalton.</p>
Full article ">Figure 5
<p>The nuclear/cytoplasmic ratio of STAT3-GFP is similar in HDAC1-depleted uninfected cells and HDAC1-depleted infected cells. The pixel density of STAT3-GFP in the nucleus and the cytoplasm of Control siRNA-transfected and HDAC1 siRNA-transfected uninfected as well as infected cells was quantified using Arivis Vision 4D ver.4.1.2 software (Zeiss) integrated with Cellpose. Then, STAT3-GFP pixel density per µm<sup>2</sup> of nucleus was divided by STAT3-GFP pixel density per µm<sup>2</sup> of cytoplasm to calculate the STAT3-GFP nuclear/cytoplasmic (N:C) ratio. The data presented are the Mean ± SD of N:C ratio of cells imaged across three biological replicates; n = 102 (36 from each replicate). The <span class="html-italic">p</span>-value was calculated using two-way Analysis of Variance (ANOVA) employing Šídák’s multiple comparisons test. **** <span class="html-italic">p</span> = &lt;0.0001, ns, not significant.</p>
Full article ">Figure 6
<p>The IAV-induced nuclear translocation of STAT3-GFP is inhibited in HDAC2-depleted cells. HeLa cells were first transfected with either Control siRNA or HDAC2 siRNA for 30 h and then transfected with plasmid pEGFP-N1-STAT3 for a further 30 h. Cells were then infected with PR8 at MOI of 3.0. After 6 h, cells were fixed, permeabilized, and stained with Mouse anti-HDAC2 antibody followed by Donkey anti-Mouse IgG conjugated with Alexa Fluor 594. Subsequently, cells were stained with Goat anti-NP antibody followed by Donkey anti-Goat IgG conjugated with Alexa Fluor 647. Finally, cells were stained with DAPI and imaged using a confocal microscope under 40× magnification. UNI, uninfected; INF, infected.</p>
Full article ">Figure 7
<p>The nuclear/cytoplasmic ratio of STAT3-GFP is similar in HDAC2-depleted uninfected cells and HDAC2-depleted infected cells. The pixel density of STAT3-GFP in the nucleus and the cytoplasm of Control siRNA-transfected and HDAC2 siRNA-transfected uninfected as well as infected cells was quantified using Arivis Vision 4D ver.4.1.2 software (Zeiss) integrated with Cellpose. Then, STAT3-GFP pixel density per µm<sup>2</sup> of nucleus was divided by STAT3-GFP pixel density per µm<sup>2</sup> of cytoplasm to calculate the STAT3-GFP N:C ratio. The data presented are the Mean ± SD of N:C ratio of cells imaged across three biological replicates; n = 138 (46 from each replicate). The <span class="html-italic">p</span>-value was calculated using two-way ANOVA employing Šídák’s multiple comparisons test. **** <span class="html-italic">p</span> = &lt;0.0001; ns, not significant.</p>
Full article ">
17 pages, 4463 KiB  
Article
Rutin Alleviates Zearalenone-Induced Endoplasmic Reticulum Stress and Mitochondrial Pathway Apoptosis in Porcine Endometrial Stromal Cells by Promoting the Expression of Nrf2
by Chuangjiang Chen, Chenlong Wang, Hui Jiang, Mengya Wang, Sajid Ur Rahman, Changjiang Chen, Hongyan Ding, Chang Zhao, Wanyue Huang and Xichun Wang
Toxins 2025, 17(1), 7; https://doi.org/10.3390/toxins17010007 - 26 Dec 2024
Viewed by 416
Abstract
Zearalenone (ZEA) is a mycotoxin commonly found in moldy cereals and has a range of toxic effects that have seriously affected animal husbandry. Rutin, a natural flavonoid with antioxidant activities, has been studied for its potential involvement in mitigating ZEA-induced apoptosis in porcine [...] Read more.
Zearalenone (ZEA) is a mycotoxin commonly found in moldy cereals and has a range of toxic effects that have seriously affected animal husbandry. Rutin, a natural flavonoid with antioxidant activities, has been studied for its potential involvement in mitigating ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) and its potential molecular mechanism, particularly concerning the expression of Nrf2. This study investigates the molecular pathways by which rutin alleviates ZEA-induced ESC apoptosis, focusing on the role of Nrf2. Experimental data reveal that ZEA suppresses Nrf2 nuclear translocation and reduces mitochondrial membrane potential (MMP), leading to oxidative stress, endoplasmic reticulum stress (ERS), and mitochondrial pathway-driven apoptosis. Notably, rutin mitigates ZEA-induced apoptosis through Nrf2 activation. These findings highlight Nrf2 as a critical factor in rutin’s protective effects against ZEA-induced apoptosis, offering valuable insights for the clinical prevention and treatment of ZEA toxicity. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) Effect of different concentrations of ZEA on cell viability. (<b>B</b>) Prediction of the ZEA IC<sub>50</sub> concentration by GraphPad. (<b>C</b>) Effect of different concentrations of rutin on cell viability. (<b>D</b>) Effects of different concentrations of rutin combined with ZEA on cell viability. Data presented above are means ± SD. * and ** indicate significant differences compared to the control group (<span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2
<p>(<b>A</b>) The green fluorescence represents Nrf2, and the blue fluorescence represents the nucleus. (<b>B</b>) Relative luminance of Nrf2 protein fluorescence (<b>C</b>) Relative expression level of <span class="html-italic">Nrf2</span> mRNA. (<b>D</b>) Nrf2 protein expression. (<b>E</b>) Relative expression of Nrf2 protein. Data presented above are means ± SD. ** indicated significant difference compared to control group; ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R and Z+N group (<span class="html-italic">p &lt;</span> 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 3
<p>(<b>A</b>) ROS level detected by FCM and ROS horizontal bar chart. (<b>B</b>,<b>C</b>) The content of MDA and 4-HNE. (<b>D</b>,<b>E</b>) Activity of SOD and GSH-Px. Data presented above are means ± SD. ** indicated significant difference compared to the control group; ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R group and Z+N group (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>(<b>A</b>) The MMP detected by FCM (<b>B</b>) The histogram represents the MMP level. Data presented above are means ± SD. ** indicated significant difference compared to the control group; # and ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R and Z+N group (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>(<b>A</b>) The apoptosis rate detected by FCM (<b>B</b>) The histogram represents the apoptosis rate. Data presented above are means ± SD. ** indicated significant difference compared to the control group; ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R and Z+N group (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>(<b>A</b>) <span class="html-italic">Bcl-2</span> mRNA expression. (<b>B</b>) <span class="html-italic">Bax</span> mRNA expression. (<b>C</b>) <span class="html-italic">Cyt c</span> mRNA expression. (<b>D</b>) <span class="html-italic">GRP78</span> mRNA expression. (<b>E</b>) <span class="html-italic">Caspase12</span> mRNA expression. (<b>F</b>) <span class="html-italic">CHOP</span> mRNA expression. (<b>G</b>) <span class="html-italic">Nrf2</span> mRNA expression. Data presented above are means ± SD. ** indicated significant difference compared to the control group; ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R and Z+N group (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 7
<p>(<b>A</b>,<b>B</b>) Bcl-2, Bax, Cyt c, Grp78, Caspase12, and CHOP protein expression were detected by WB. (<b>C</b>–<b>H</b>) Relative expression of Bcl-2, Bax, Cyt C, Grp78, Caspase12, and CHOP protein. Data presented above are means ± SD. ** indicated significant difference compared to the control group; and ## indicated significant difference compared to the ZEA group; ++ indicated significant difference between the Z+N+R and Z+N group (<span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
14 pages, 16811 KiB  
Article
Exogenous Melatonin Boosts Heat Tolerance in Rosa hybrida via RhCOMT1 Modulation
by Chenyang Li, Zhiyin Ding, Zipeng Cai, Yongying Ruan, Peitao Lü and Yang Liu
Plants 2025, 14(1), 29; https://doi.org/10.3390/plants14010029 - 25 Dec 2024
Viewed by 327
Abstract
Rosa hybrida is one the most commonly cultivated ornamental plant of economic importance and faces major challenges under heat stress. Melatonin has been widely shown to regulate plant stress response; however, the exact mechanism involved in heat stress in R. hybrida has yet [...] Read more.
Rosa hybrida is one the most commonly cultivated ornamental plant of economic importance and faces major challenges under heat stress. Melatonin has been widely shown to regulate plant stress response; however, the exact mechanism involved in heat stress in R. hybrida has yet to be determined. Here, we observed that R. hybrida in vitro plantlets supplemented with melatonin in the culture medium exhibited higher chlorophyll content, relative ion leakage, and fresh weight after 12 d of high-temperature treatment; the optimal concentration was established at 5 mg/L. Using molecular and biochemical techniques, we explored the roles of a melatonin synthase gene RhCOMT1, which expression was influenced by heat stress and melatonin. RhCOMT1 was located in the nuclear-cytoplasmic under ambient conditions, while heat stress translocated the distribution of RhCOMT1 to chloroplasts. Overexpression of RhCOMT1 in rose petal enhanced thermotolerance, and silencing of RhCOMT1 reduced thermotolerance via affect H2O2 content and relative ion leakage. These findings collectively emphasize the pivotal role of melatonin in enhancing thermotolerance to R. hybrida by alleviation of oxidative stress, through modulation of RhCOMT1 expression and location. Full article
Show Figures

Figure 1

Figure 1
<p>Phenotypic characterization of the buds and sterilized stems under heat stress. (<b>A</b>) Phenotypes of the <span class="html-italic">Rosa hybrida</span> buds under heat stress. Scale bar = 5 cm. (<b>B</b>,<b>C</b>) Flower diameters and (<b>B</b>) petal numbers (<b>C</b>) of CK, HT, MT + HT groups. (<b>D</b>) Phenotypes of the <span class="html-italic">R. hybrida</span> in vitro sterilized stems in different concentrations of melatonin medium under high-temperature treatment. Scale bar = 1 cm. (<b>E</b>–<b>G</b>) ChlorophyII content, (<b>E</b>) relative ion leakage, and (<b>F</b>) relative fresh weight (<b>G</b>) of the <span class="html-italic">R. hybrida</span> sterilized stems grow in different concentrations of melatonin medium under 12 d of HT. CK, control group; HT, 42 °C high-temperature treatment group; MT, melatonin treatment; error bars represent ± SD (n = 3). Significant differences compared to the 0 mg/L melatonin treatment group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns, no significance, Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>Expression pattern and bioinformatics analysis of <span class="html-italic">RhCOMT1</span>. (<b>A</b>) The expression levels of <span class="html-italic">RhCOMT1</span> under high-temperature treatment. (<b>B</b>) The expression levels of <span class="html-italic">RhCOMT1</span> under melatonin treatment of different concentrations. (<b>C</b>) Phylogenetic relationships between RhCOMT1 and other COMT proteins. The data are presented as the means ± SDs (n = 9). Significant differences compared to the 0 h high-temperature treatment and 0 mg/L melatonin treatment group are indicated by asterisks (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, ns, no significance, Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>Subcellular localization assay of RhCOMT1 under RT and HT. The location of RhCOMT1 is based on visualization of GFP in tobacco leaves transformed with a fusion construct (pSuper1300:GFP-<span class="html-italic">RhCOMT1</span>) or empty vector (pSuper1300:GFP). Microscopic images were taken under bright field and fluorescence. The overlapped images are shown on the right. RT, room temperature; HT, high-temperature treatment; GFP, green fluorescent protein.</p>
Full article ">Figure 4
<p>Thermotolerance assay using <span class="html-italic">RhCOMT1</span>-overexpressing petal discs. (<b>A</b>) The phenotypes of rose petal discs under room temperature (RT, 25 °C) and post high-temperature treatment (HT, 42 °C, 36 h) with DAB staining. The experiment was repeated three times, with 27 rose petal discs per experiment. Scale bar = 1 cm. (<b>B</b>) Detection of <span class="html-italic">RhCOMT1</span> expression in <span class="html-italic">RhCOMT1</span>-overexpression petal discs. (<b>C</b>,<b>D</b>) Determination of H<sub>2</sub>O<sub>2</sub> content and (<b>C</b>) relative ion leakage (<b>D</b>) of petal discs post HT (42 °C, 36 h). The data are presented as the means ± SD for three replicates (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, Student’s <span class="html-italic">t</span>-test, the pSuper1300:<span class="html-italic">RhCOMT1</span> compared with the pSuper1300 control post HT, respectively). RT, room temperature; HT, high-temperature treatment; DAB, 3,3′diaminobenzidine.</p>
Full article ">Figure 5
<p>Thermotolerance assay using <span class="html-italic">RhCOMT1</span>-silenced petal discs. (<b>A</b>) The phenotypes of rose petal discs under room temperature (RT, 25 °C) and high-temperature treatment (HT, 42 °C, 36 h) with DAB staining. The experiment was repeated three times, with 27 rose petal discs per experiment. Scale bar = 1 cm. (<b>B</b>) Detection of <span class="html-italic">RhCOMT1</span> expression in TRV-VIGS petal discs. (<b>C</b>,<b>D</b>) Determination of H<sub>2</sub>O<sub>2</sub> content and (<b>C</b>) relative ion leakage (<b>D</b>) of petal discs post HT (42 °C, 36 h). The data are presented as the means ± SDs for three replicates (* <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, Student’s <span class="html-italic">t</span>-test, the pTRV2-<span class="html-italic">RhCOMT1</span> compared with the pTRV2-control post HT, respectively). RT, room temperature; HT, high-temperature treatment; DAB, 3,3′diaminobenzidine.</p>
Full article ">Figure 6
<p>Model for <span class="html-italic">RhCOMT</span> responding to heat stress of <span class="html-italic">R. hybrida</span>. <span class="html-italic">RhCOMT1</span> can be significantly induced by heat stress. The over-expression of RhCOMT1 has been shown to enhance heat stress tolerance by increasing the capability to scavenge ROS. Furthermore, RhCOMT1 has been observed to localise in chloroplasts at high temperatures, which has the potential to improve chloroplast chlorophyll stability and photosynthetic efficiency.</p>
Full article ">
14 pages, 2758 KiB  
Review
The Expression Regulation and Cancer-Promoting Roles of RACGAP1
by Jiacheng Lin, Yuhao Zhu, Zhaoping Lin, Jindong Yu, Xiaobing Lin, Weiyuan Lai, Beibei Tong, Liyan Xu, Enmin Li and Lin Long
Biomolecules 2025, 15(1), 3; https://doi.org/10.3390/biom15010003 - 24 Dec 2024
Viewed by 312
Abstract
RACGAP1 is a Rho-GTPase-activating protein originally discovered in male germ cells to inactivate Rac, RhoA and Cdc42 from the GTP-bound form to the GDP-bound form. GAP has traditionally been known as a tumor suppressor. However, studies increasingly suggest that overexpressed RACGAP1 activates Rac [...] Read more.
RACGAP1 is a Rho-GTPase-activating protein originally discovered in male germ cells to inactivate Rac, RhoA and Cdc42 from the GTP-bound form to the GDP-bound form. GAP has traditionally been known as a tumor suppressor. However, studies increasingly suggest that overexpressed RACGAP1 activates Rac and RhoA in multiple cancers to mediate downstream oncogene overexpression by assisting in the nuclear translocation of signaling molecules and to promote cytokinesis by regulating the cytoskeleton or serving as a component of the central spindle. Contradictorily, it was also reported that RACGAP1 in gastric cancer could inactivate Rac and RhoA. In addition, studies have revealed that RACGAP1 can be a biomarker for prognosis, and its role in reducing doxorubicin sensitivity poses difficulties for treatment, while the current drug targets mainly focus on its downstream molecule. This article mainly reviews the expression regulation of RACGAP1 and its cancer-promoting functions through oncogene expression mediation and Rho-GTPase activation. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>RACGAP1 promotes cancer through the Hippo pathway and JAK/STAT pathway. In the Hippo pathway, the phosphorylated form of MST1/2 activates LAST1/2 via phosphorylation. YAP can be deactivated by the activated form of LAST when phosphorylated. YAP in the non-phosphorylated state can enter the nucleus, bind to the promoter region of TPR with TEAD and promote its transcription. TPR, RACGAP1 and AURKB form complexes that promote spindle formation. Moreover, RACGAP1 inhibits the Hippo pathway by promoting the inactivation of LAST1/2 through F-actin. RACGAP1 forms a ternary complex together with STAT3 and Rac1 to phosphorylate STAT3, and accomplishes the nuclear transport of STAT3 as a nuclear chaperone. RACGAP1 binds JAK2 to enable the phosphorylation of STAT3 by JAK2. Rac1 and p-STAT3 activate the nuclear localization signal (NLS) in RACGAP1 so that it can promote the nuclear translocation and the transcriptional activity of p-STAT3. RACGAP1 can inactivate Rac1, but after binding with APPL2, it cannot inactivate Rac1.</p>
Full article ">Figure 2
<p>RACGAP1 can activate Rho-GTPases. (<b>a</b>) RACGAP1 inhibits Rac1, Cdc42 and RhoA. (<b>b</b>) Phosphorylated RACGAP1 can activate Rac1. (<b>c</b>) RACGAP1 can activate RhoA in hepatocellular carcinoma. (<b>d</b>) RACGAP1 can form a complex with Rac1 and STAT3, and with ECT and MKLP1, the latter complex of which can activate RhoA. (<b>e</b>) In NIH3T3 cells, Rac1 can inhibit RhoA. (<b>f</b>) In Swiss 3T3 fibroblasts, RhoA can activate Rac1.</p>
Full article ">Figure 3
<p>RACGAP1 binding ECT2 to promotes actomyosin ring formation. Plk phosphorylates RACGAP1 at 2 sites so that RACGAP1 binds to ECT2 with the help of MKLP1, and the complex activates RhoA, Myosin II and ERK. PP2A-B′ can bind to RACGAP1 to break the interaction between RACGAP1 and ECT2.</p>
Full article ">
11 pages, 1974 KiB  
Article
Nuclear Factor-κB Signaling Regulates the Nociceptin Receptor but Not Nociceptin Itself
by Lan Zhang, Ulrike M. Stamer, Robin Moolan-Vadackumchery and Frank Stüber
Cells 2024, 13(24), 2111; https://doi.org/10.3390/cells13242111 - 20 Dec 2024
Viewed by 409
Abstract
The nociceptin receptor (NOP) and nociceptin are involved in the pathways of pain and inflammation. The potent role of nuclear factor-κB (NFκB) in the modulation of tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β on the nociceptin system in human THP-1 cells under inflammatory [...] Read more.
The nociceptin receptor (NOP) and nociceptin are involved in the pathways of pain and inflammation. The potent role of nuclear factor-κB (NFκB) in the modulation of tumor necrosis factor-α (TNF-α) and interleukin (IL)-1β on the nociceptin system in human THP-1 cells under inflammatory conditions were investigated. Cells were stimulated without/with phorbol-myristate-acetate (PMA), TNF-α, IL-1β, or PMA combined with individual cytokines. To examine NFκB’s contribution to the regulation of the nociceptin system, PMA-stimulated cells were treated with NFκB inhibitor BAY 11-7082, JSH-23, or anacardic acid before culturing with TNF-α or IL-1β. NOP and prepronociceptin (ppNOC) mRNA were quantified by RT-qPCR; cell membrane NOP and intracellular nociceptin protein levels were measured by flow cytometry. Phosphorylation and localization of NFκB/p65 were determined using ImageStream. PMA + TNF-α decreased NOP mRNA compared to stimulation with PMA alone, while PMA + IL-1β did not. BAY 11-7082 and JSH-23 reversed the repression of NOP by PMA + TNF-α. TNF-α and IL-1β attenuated PMA’s upregulating effects on ppNOC. None of the inhibitors preserved the upregulation of ppNOC in PMA + TNF-α and PMA + IL-1β cultures. TNF-α strongly mediated the nuclear translocation of NFκB/p65 in PMA-treated cells, while IL-1β did not. Proinflammatory cytokines suppressed NOP and ppNOC mRNA in PMA-induced human THP-1 cells. NFκB signaling seems to be an important regulator controlling the transcription of NOP. These findings suggest that the nociceptin system may play an anti-inflammatory role during immune responses. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of PMA on NOP (<b>A</b>) and nociceptin (<b>B</b>) mRNA and protein levels in THP-1 cells. Cells were treated with PMA 5 ng/mL or without PMA (controls) for 24 h. Median with interquartile range and 10–90 percentiles; mean “+”; mRNA and protein data are representative of six and twelve independent experiments, respectively. Wilcoxon tests * <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; 0.001, compared to the respective controls.</p>
Full article ">Figure 2
<p>Dose-dependent effects of cytokines and NFκB inhibitors. THP-1 cells were cultured without/with PMA 5 ng/mL and without/with various concentrations of individual cytokines for 24 h. mRNA expression of <span class="html-italic">NOP</span> (<b>A</b>) and <span class="html-italic">ppNOC</span> (<b>B</b>) is presented as mRNA ratio related to the respective PMA samples. (<b>C</b>) Dose-dependent inhibitory effects of different NFκB inhibitors on <span class="html-italic">IL1B</span> mRNA levels induced by LPS. Cells were pre-treated without/with various concentrations of BAY 11-7082 (BAY), JSH-23 (JSH), or anarcadic acid (AA) for 1 h prior to culturing without/with LPS 100 ng/mL for 6 h. <span class="html-italic">IL1B</span> mRNA levels are presented as mRNA ratio related to an untreated group. Data are from two independent experiments and measures are expressed in mean with range. One-sample t test, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005, compared to the PMA group (<span class="html-italic">NOP</span> and <span class="html-italic">ppNOC</span>) and compared to the LPS group (<span class="html-italic">IL1B</span>).</p>
Full article ">Figure 3
<p>Effects of TNF-α and IL-1β on <span class="html-italic">NOP</span> and <span class="html-italic">ppNOC</span> mRNA expression. THP-1 cells were cultured without/with PMA 5 ng/mL in the presence or absence of TNF-α 10 ng/mL or IL-1β 10 ng/mL for 24 h. Quantitative PCR analysis of <span class="html-italic">NOP</span> (<b>A</b>) and <span class="html-italic">ppNOC</span> mRNA expression (<b>B</b>). Fold change values in mRNA levels are normalized against the respective PMA groups. Data are from six independent experiments and are presented as median with interquartile range and 10–90 percentiles; mean “+”. Wilcoxon test with post hoc test. ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 4
<p>Effects of NFκB inhibitors on <span class="html-italic">NOP</span> and <span class="html-italic">ppNOC</span> mRNA expression. THP-1 cells were stimulated without/with PMA 5 ng/mL for 24 h, followed by treatment without/with one of the NFκB inhibitors (BAY 11-7082 (BAY, 100 nM), JSH-23 (JSH, 100 nM), and anacardic acid (AA, 100 nM)) for 1 h before exposure to TNF-α 10 ng/mL or IL-1β 10 ng/mL for 6 h (BAY and JSH) and for 12 h (AA). <span class="html-italic">NOP</span> (<b>A</b>–<b>C</b>) and <span class="html-italic">ppNOC</span> mRNA levels (<b>D</b>–<b>F</b>) are fold-change related to the corresponding PMA groups (controls). Median with interquartile range and 10–90 percentiles, experiment using BAY or AA, <span class="html-italic">n</span> = 4; experiment using JSH, <span class="html-italic">n</span> = 7. Mann–Whitney U test with post hoc test. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 5
<p>NFκB/p65 phosphorylation and nuclear translocation. THP-1 cells were treated without/with PMA 5 ng/mL for 24 h, followed by incubation with TNF-α 10 ng/mL or IL-1β 10 ng/mL or without additional cytokines for 1 h. (<b>A</b>) Composite images of representative cells (60×) in real time showing level of NFκB/p65 protein and its localization. From left to right in each panel, brightfield images of each cell, followed by the nuclear (red) and NFκB/p65 (green) images, and the merged image of the nucleus with NFκB/p65. The scale bar is 7 μm. (<b>B</b>) NFκB/p65 translocation into the nucleus, measured using similarity scores (SS). Histogram displaying varying SS in the cells of each group: untreated (white), TNF-α (yellow), IL-1β (green), PMA (purple), PMA + TNF-α (red) and PMA+ IL-1β (blue). (<b>C</b>) Median SS (MSS) for NFκB/p65 of the individual groups. Data are from three independent experiments and measures are expressed in median with interquartile range.</p>
Full article ">
16 pages, 3701 KiB  
Article
Essential Role of Cortactin in Myogenic Differentiation: Regulating Actin Dynamics and Myocardin-Related Transcription Factor A-Serum Response Factor (MRTFA-SRF) Signaling
by Quoc Kiet Ly, Mai Thi Nguyen, Thanh Huu Phan Ngo and Wan Lee
Int. J. Mol. Sci. 2024, 25(24), 13564; https://doi.org/10.3390/ijms252413564 - 18 Dec 2024
Viewed by 400
Abstract
Cortactin (CTTN) is an actin-binding protein regulating actin polymerization and stabilization, which are vital processes for maintaining skeletal muscle homeostasis. Despite the established function of CTTN in actin cytoskeletal dynamics, its role in the myogenic differentiation of progenitor cells remains largely unexplored. In [...] Read more.
Cortactin (CTTN) is an actin-binding protein regulating actin polymerization and stabilization, which are vital processes for maintaining skeletal muscle homeostasis. Despite the established function of CTTN in actin cytoskeletal dynamics, its role in the myogenic differentiation of progenitor cells remains largely unexplored. In this study, we investigated the role of CTTN in the myogenic differentiation of C2C12 myoblasts by analyzing its effects on actin cytoskeletal remodeling, myocardin-related transcription factor A (MRTFA) nuclear translocation, serum response factor (SRF) activation, expression of myogenic transcription factors, and myotube formation. CTTN expression declined during myogenic differentiation, paralleling the reduction in MyoD, suggesting a potential role in the early stages of myogenesis. We also found that CTTN knockdown in C2C12 myoblasts reduced filamentous actin, enhanced globular actin levels, and inhibited the nuclear translocation of MRTFA, resulting in suppressed SRF activity. This led to the subsequent downregulation of myogenic regulatory factors, such as MyoD and MyoG. Furthermore, CTTN knockdown reduced the nuclear localization of YAP1, a mechanosensitive transcription factor, further supporting its regulatory roles in cell cycle and proliferation. Consequently, CTTN depletion impeded proliferation, differentiation, and myotube formation in C2C12 myoblasts, highlighting its dual role in the coordination of cell cycle regulation and myogenic differentiation of progenitor cells during myogenesis. This study identifies CTTN as an essential regulator of myogenic differentiation via affecting the actin remodeling-MRTFA-SRF signaling axis and cell proliferation. Full article
Show Figures

Figure 1

Figure 1
<p>Modulation of CTTN expression during myoblast differentiation. (<b>A</b>) Immunoblotting was conducted to assess CTTN expression levels in C2C12 myoblasts and various tissues from C57BL/6 mice, with α-tubulin as a loading control. (<b>B</b>) C2C12 myoblasts were harvested on specified differentiation days, and the protein expression levels of MyoD, MyoG, MyHC, and CTTN were analyzed by immunoblotting, with β-actin as a loading control. (<b>C</b>) Protein expression levels were normalized to β-actin, and relative expression ratios were calculated, setting day 0 as one for MyoD and CTTN, day 1 for MyoG, and day 2 for MyHC. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 2
<p>CTTN knockdown led to a reduction in F-actin levels and an increase in G-actin levels. C2C12 myoblasts were transfected with 200 nM of either control scRNA or siCTTN (siCTTN-1 or siCTTN-2). (<b>A</b>) CTTN expression was assessed by immunoblotting 24 h after transfection. CTTN expression levels were normalized to β-actin, and relative expression ratios were calculated with the control scRNA set to one. (<b>B</b>) After 24 h post-transfection, cells were stained with FITC-phalloidin (green) for F-actin and Hoechst 33,342 (blue) for nuclei. Scale bar: 25 μm. Phalloidin intensities were quantified using ImageJ software, version 1.5.4. (<b>C</b>) F- and G-actin levels were quantified by flow cytometry after staining with FITC-phalloidin for F-actin and DNase I for G-actin, respectively. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>CTTN depletion impaired the nuclear localization of MRTFA and YAP1. C2C12 myoblasts were transfected with either control scRNA or siCTTN and analyzed 24 h post-transfection. (<b>A</b>) Cytoplasmic and nuclear fractions were subjected to immunoblot analysis for MRTFA, SRF, YAP1, pYAP1 (phosphorylated YAP1), and CTTN expression. For MRTFA, different exposure times were used to account for its varied distribution between cytoplasmic and nuclear compartments. α-Tubulin and lamin B2 served as cytoplasmic and nuclear markers, respectively. β-Actin was used as a loading control. (<b>B</b>,<b>C</b>) The protein expression levels were normalized to β-actin, and relative expression ratios were calculated with the control scRNA set to one. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 4
<p>CTTN knockdown suppressed SRF transcriptional activity. (<b>A</b>) Diagram of the luciferase reporter construct featuring the truncated SMYD1 promoter region, including the CArG box for SRF binding. (<b>B</b>) C2C12 myoblasts were transfected with either the pGL3 vector (Vector) or pGL3 containing the SMYD1 promoter (SMYD1) along with control scRNA or siCTTN. Relative luciferase activity was measured 24 h post-transfection. (<b>C</b>) C2C12 myoblasts were transfected with either control scRNA or siCTTN, and mRNA levels of SRF, Vinculin, and SMYD1 were assessed by RT-qPCR, normalized to GAPDH expression 24 h post-transfection. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns indicates non-significance.</p>
Full article ">Figure 5
<p>CTTN depletion impeded cell proliferation and cell cycle progression. C2C12 myoblasts were transfected with either control scRNA or siCTTN and analyzed 24 h post-transfection. (<b>A</b>) Cell proliferation was evaluated by EdU incorporation (green) to label replicating cells, with Hoechst 33,342 (blue) as a nuclear counterstain. Scale bar: 50 µm. (<b>B</b>) The percentage of EdU-positive cells was quantified using ImageJ software. (<b>C</b>) Viable cell numbers were measured using a cell viability assay kit. (<b>D</b>) mRNA levels of proliferation markers (PCNA, cyclin B1, and cyclin D1) were assessed by RT-<span class="html-italic">q</span>PCR and normalized to GAPDH expression. (<b>E</b>,<b>F</b>) Cell cycle analysis was performed using flow cytometry with scatter plots. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 6
<p>CTTN knockdown suppressed the expression of myogenic regulatory factors. (<b>A</b>) C2C12 myoblasts were transfected with either control scRNA or siCTTN, allowed to differentiate, and then harvested on specified differentiation days. Protein expression levels of MyoD, MyoG, MyHC, and CTTN were analyzed by immunoblotting. (<b>B</b>) Protein expression levels for scRNA (open column) and siCTTN (blue column) were normalized to β-actin and presented as relative ratios, with scRNA expression levels on day 0 (for CTTN and MyoD) or day 3 (for MyoG and MyHC) set to one. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001); ns indicates non-significance.</p>
Full article ">Figure 7
<p>CTTN depletion impaired myogenic differentiation. C2C12 myoblasts were transfected with either control scRNA or siCTTN and then allowed to differentiate for 5 days. (<b>A</b>) Representative immunocytochemistry stained with MyHC antibody (green) and Hoechst 33,342 (blue). Scale bar: 100 μm. (<b>B</b>) MyHC-positive areas, differentiation indices, fusion indices, and myotube widths were determined as described in <a href="#sec4-ijms-25-13564" class="html-sec">Section 4</a>. Data are presented as means ± SEM (n = 3), with asterisks indicating statistical significance (*** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 8
<p>Schematic illustration of the actin-MRTFA-SRF and YAP1 signaling pathway regulated by CTTN.</p>
Full article ">
30 pages, 10385 KiB  
Article
Second-Generation Antipsychotics Induce Metabolic Disruption in Adipose Tissue-Derived Mesenchymal Stem Cells Through an aPKC-Dependent Pathway
by Marco Varalda, Jacopo Venetucci, Herald Nikaj, Chaitanya Reddy Kankara, Giulia Garro, Nazanin Keivan, Valentina Bettio, Paolo Marzullo, Annamaria Antona, Guido Valente, Sergio Gentilli and Daniela Capello
Cells 2024, 13(24), 2084; https://doi.org/10.3390/cells13242084 - 17 Dec 2024
Viewed by 573
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities, including visceral obesity, dyslipidemia, and insulin resistance. In this regard, visceral white adipose tissue (vWAT) plays a critical role, influencing energy metabolism, immunomodulation, and oxidative stress. Adipose-derived stem cells (ADSCs) are key players in [...] Read more.
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities, including visceral obesity, dyslipidemia, and insulin resistance. In this regard, visceral white adipose tissue (vWAT) plays a critical role, influencing energy metabolism, immunomodulation, and oxidative stress. Adipose-derived stem cells (ADSCs) are key players in these processes within vWAT. While second-generation antipsychotics (SGAs) have significantly improved treatments for mental health disorders, their chronic use is associated with an increased risk of MetS. In this study, we explored the impact of SGAs on ADSCs to better understand their role in MetS and identify potential therapeutic targets. Our findings reveal that olanzapine disrupts lipid droplet formation during adipogenic differentiation, impairing insulin receptor endocytosis, turnover, and signaling. SGAs also alter the endolysosomal compartment, leading to acidic vesicle accumulation and increased lysosomal biogenesis through TFEB activation. PKCζ is crucial for the SGA-induced nuclear translocation of TFEB and acidic vesicle formation. Notably, inhibiting PKCζ restored insulin receptor tyrosine phosphorylation, normalized receptor turnover, and improved downstream signaling following olanzapine treatment. This activation of PKCζ by olanzapine is driven by increased phosphatidic acid synthesis via phospholipase D (PLD), following G protein-coupled receptor (GPCR) signaling activation. Overall, olanzapine and clozapine disrupt endolysosomal homeostasis and insulin signaling in a PKCζ-dependent manner. These findings highlight SGAs as valuable tools for uncovering cellular dysfunction in vWAT during MetS and may guide the development of new therapeutic strategies to mitigate the metabolic side effects of these drugs. Full article
(This article belongs to the Special Issue Adipose Tissue, Obesity, and Metabolic Diseases)
Show Figures

Figure 1

Figure 1
<p>Assessment of olanzapine and clozapine cytotoxic activity in ADSCs. Viabilities of ADSCs treated with scalar doses of drugs for 72 h; IC50, i.e., we calculated the drug concentration reduced by 50% in terms of viability compared to the control (<b>a</b>). Bar graphs showing cell viability after 7 days of treatment with scalar doses of drugs; viability data are presented as the percentage of viable cells relative to the negative control treated with DMSO. Data are presented as mean ± SEM from three independent experiments (<b>b</b>). **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001; ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 1 Cont.
<p>Assessment of olanzapine and clozapine cytotoxic activity in ADSCs. Viabilities of ADSCs treated with scalar doses of drugs for 72 h; IC50, i.e., we calculated the drug concentration reduced by 50% in terms of viability compared to the control (<b>a</b>). Bar graphs showing cell viability after 7 days of treatment with scalar doses of drugs; viability data are presented as the percentage of viable cells relative to the negative control treated with DMSO. Data are presented as mean ± SEM from three independent experiments (<b>b</b>). **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001; ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>Olanzapine affects ADSC adipogenic differentiation. Representative images of lipid droplets in ADSC#3 treated with 5 μM olanzapine alone or in combination with WAT-differentiating medium using HCS LipidTox for neutral lipids; nuclei were stained using Hoechst 33342 (<b>a</b>). Bar graphs showing quantification of lipid droplet staining/blue nuclei staining ratio as fold change relative to control in ADSC#3 (<b>b</b>) and ADSC#5 (<b>c</b>); data are expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate. Graphs showing quantification of mean dimension and number of lipid droplets in ADSCs treated with olanzapine and in controls (<b>d</b>–<b>g</b>); data are expressed as the mean ± SD of a representative experiment out of three independent experiments. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05. **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01 ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001. ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Olanzapine downregulates insulin signaling. Representative Western blot of ADSC#3 cells after 16-h pretreatment with 5 μM olanzapine and stimulation with insulin (50 ng/mL) for 5, 30, and 60 min; lysates were analyzed for P-INSRβ Y1146, INSRβ, P-AKT T308, P-AKT S473, total AKT, P-ERK T202/Y204, and ERK (<b>a</b>). Bar graphs showing quantification of P-INSRβ Y1142 (<b>b</b>), P-AKT T308 (<b>c</b>), P-AKT S473 (<b>d</b>), and P-ERK T202/Y204 (<b>e</b>) normalized on their respective total proteins and expressed as fold change relative to control. Western blot analysis of immunoprecipitated INSRβ P-Ser in cells stimulated with insulin 50 ng/mL for 15 min (<b>f</b>). Bar graph showing quantification of P-Ser signals normalized on total INSR; densitometry expressed as fold change relative to control (<b>g</b>). Graphs are expressed as the mean ± SD of three independent experiments. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Olanzapine impairs INSR endocytosis. Western blot analysis showing internalization of biotinylated INSRβ in ADSC#5 treated for 16 h with olanzapine and stimulated with insulin for 15 min: Ts represents total biotinylated proteins on the surface, T0 the surface proteins after quenching of the membrane in unstimulated cells, and T15 the surface proteins after endocytosis (<b>a</b>). Bar graph representative of 3 independent experiments showing quantification of internalized receptor; densitometry is expressed as T15 /Ts ratio normalized on total INSRβ, as fold change relative to control (<b>b</b>). Representative images of INSRβ localization on ADSC#5 plasma membrane after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green), while actin was stained using phalloidin 546 (<b>c</b>). Bar graphs showing colocalization of INSRβ and actin on ADSC#3 (<b>d</b>) and ADSC#5 (<b>e</b>) plasma membrane expressed as Pearson coefficient; data are expressed as the mean ± SD of 3 independent experiments. Representative images of INSRβ intracellular localization in ADSC#5 treated with olanzapine and in control cells. INSRβ intracellular localization in RAB7-positive late endosomes after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); RAB7 using anti-RAB7 primary antibody and secondary Alexa Fluor 546 (red) (<b>f</b>). Bar graph showing colocalization of INSRβ and RAB7 expressed as Pearson coefficient (<b>g</b>). Representative images of INSRβ localization in CD 63-positive exocytic vesicles after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); CD63 using anti-CD63 primary antibody and secondary Alexa Fluor 546 (red) (<b>h</b>). Bar graph showing colocalization of INSRβ and CD63 expressed as Pearson coefficient (<b>i</b>). Representative images of INSR localization in lysosomes after 16 h olanzapine treatment and 15 min insulin stimulation. INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); LAMP1 using anti-LAMP1 primary antibody and secondary Alexa Fluor 546 (red) (<b>j</b>). Bar graph showing colocalization of INSRβ and LAMP1 expressed as Pearson coefficient (<b>k</b>). Results are expressed as the mean ± SD of three independent experiments. White boxes indicates zoom area, white arrows indicates colocalization spots. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4 Cont.
<p>Olanzapine impairs INSR endocytosis. Western blot analysis showing internalization of biotinylated INSRβ in ADSC#5 treated for 16 h with olanzapine and stimulated with insulin for 15 min: Ts represents total biotinylated proteins on the surface, T0 the surface proteins after quenching of the membrane in unstimulated cells, and T15 the surface proteins after endocytosis (<b>a</b>). Bar graph representative of 3 independent experiments showing quantification of internalized receptor; densitometry is expressed as T15 /Ts ratio normalized on total INSRβ, as fold change relative to control (<b>b</b>). Representative images of INSRβ localization on ADSC#5 plasma membrane after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green), while actin was stained using phalloidin 546 (<b>c</b>). Bar graphs showing colocalization of INSRβ and actin on ADSC#3 (<b>d</b>) and ADSC#5 (<b>e</b>) plasma membrane expressed as Pearson coefficient; data are expressed as the mean ± SD of 3 independent experiments. Representative images of INSRβ intracellular localization in ADSC#5 treated with olanzapine and in control cells. INSRβ intracellular localization in RAB7-positive late endosomes after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); RAB7 using anti-RAB7 primary antibody and secondary Alexa Fluor 546 (red) (<b>f</b>). Bar graph showing colocalization of INSRβ and RAB7 expressed as Pearson coefficient (<b>g</b>). Representative images of INSRβ localization in CD 63-positive exocytic vesicles after 16 h olanzapine treatment and 15 min insulin stimulation; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); CD63 using anti-CD63 primary antibody and secondary Alexa Fluor 546 (red) (<b>h</b>). Bar graph showing colocalization of INSRβ and CD63 expressed as Pearson coefficient (<b>i</b>). Representative images of INSR localization in lysosomes after 16 h olanzapine treatment and 15 min insulin stimulation. INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 488 (green); LAMP1 using anti-LAMP1 primary antibody and secondary Alexa Fluor 546 (red) (<b>j</b>). Bar graph showing colocalization of INSRβ and LAMP1 expressed as Pearson coefficient (<b>k</b>). Results are expressed as the mean ± SD of three independent experiments. White boxes indicates zoom area, white arrows indicates colocalization spots. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5
<p>Olanzapine and clozapine induce expansion of intracellular acidic compartments and lysosomal biogenesis. Effects of olanzapine and clozapine on intracellular acidic compartments were evaluated by Lysotracker red staining and fluorescence microscopy after 24 h, 72 h, and 7 days of treatment. Nuclei were stained using Hoechst 33342. Representative images of ADSC#3 treated with vehicle (DMSO, negative control), 5 µM olanzapine, or clozapine at different time points (<b>a</b>). Graphs showing quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to control; data are expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>b</b>,<b>c</b>). Representative image of WB analysis of ADSC#3 after 16 h treatment with SGAs; lysates were analyzed for LC3B, P62, and GAPDH (<b>d</b>). Bar graph showing quantification of the LC3B II/I ratio in ADSC#3 upon chloroquine treatment; densitometric analyses are expressed as the mean ± SD of three independent experiments performed in triplicate (<b>e</b>). Colocalization between LC3B (green) and LAMP1 (red) evaluated in ADSC#3 using confocal microscopy after 16 h treatment with vehicle, olanzapine, or clozapine (<b>f</b>). Histogram showing colocalization LAMP1/LC3B in ADSC#3 expressed as Pearson coefficient (<b>g</b>). Evaluation of intracellular acidic compartments, using Lysotracker red staining, in ADSC#3 cells after 16-h treatment with SGAs alone or in combination with 3-methyladenine (<b>h</b>). Bar graph showing acidic vesicle accumulation in ADSC#3 (<b>i</b>)) and ADSC#5 (<b>j</b>) treated for 16 h with olanzapine and 5 μM clozapine alone or in association with 3-methyladenine (3-MA); data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001; ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5 Cont.
<p>Olanzapine and clozapine induce expansion of intracellular acidic compartments and lysosomal biogenesis. Effects of olanzapine and clozapine on intracellular acidic compartments were evaluated by Lysotracker red staining and fluorescence microscopy after 24 h, 72 h, and 7 days of treatment. Nuclei were stained using Hoechst 33342. Representative images of ADSC#3 treated with vehicle (DMSO, negative control), 5 µM olanzapine, or clozapine at different time points (<b>a</b>). Graphs showing quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to control; data are expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>b</b>,<b>c</b>). Representative image of WB analysis of ADSC#3 after 16 h treatment with SGAs; lysates were analyzed for LC3B, P62, and GAPDH (<b>d</b>). Bar graph showing quantification of the LC3B II/I ratio in ADSC#3 upon chloroquine treatment; densitometric analyses are expressed as the mean ± SD of three independent experiments performed in triplicate (<b>e</b>). Colocalization between LC3B (green) and LAMP1 (red) evaluated in ADSC#3 using confocal microscopy after 16 h treatment with vehicle, olanzapine, or clozapine (<b>f</b>). Histogram showing colocalization LAMP1/LC3B in ADSC#3 expressed as Pearson coefficient (<b>g</b>). Evaluation of intracellular acidic compartments, using Lysotracker red staining, in ADSC#3 cells after 16-h treatment with SGAs alone or in combination with 3-methyladenine (<b>h</b>). Bar graph showing acidic vesicle accumulation in ADSC#3 (<b>i</b>)) and ADSC#5 (<b>j</b>) treated for 16 h with olanzapine and 5 μM clozapine alone or in association with 3-methyladenine (3-MA); data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001; ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>TFEB nuclear localization was investigated using confocal microscopy in ADSC#3 treated for 16 h with a vehicle or olanzapine. TFEB was stained using an anti-TFEB primary antibody and a secondary Alexa Fluor 488 (green), nuclei were stained using DAPI, and actin was stained using phalloidin 546 (<b>a</b>). Bar graph showing quantification of TFEB nuclear localization expressed as TFEB mean fluorescence in nuclear area normalized as fold change relative to control of three independent experiments (<b>b</b>). Representative images of WB analysis of ADSC#3 after 16-h treatment with olanzapine; lysates were analyzed for cathepsin B and GAPDH (<b>c</b>). Bar graph showing quantification of cathepsin B expression normalized on GAPDH. Densitometric analysis is expressed as the mean ± SD of three independent experiments (<b>d</b>). Evaluation of intracellular acidic compartments, using Lysotracker red staining, in ADSC#3 cells after 16-h treatment with SGAs alone or in combination with CHX (<b>e</b>). Bar graph showing acidic vesicle accumulation in cells treated for 16 h with olanzapine (5 µM) and clozapine (5 µM) alone or in the presence of CHX; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control (<b>f</b>,<b>g</b>). **, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.01; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001, Student’s <span class="html-italic">t</span>-test ****, <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 7
<p>PKCζ-dependent expansion of acidic vesicles mediated by SGAs. Western blot analysis of ADSCs treated with 5 μM SGAs for 16 h. Lysates were analyzed for P-(Ser)-PKC substrate, P-PKCζ T560, total PKCζ, and tubulin (<b>a</b>). Confocal microscopy experiments showing P-PKCζ T560 localization in ADSC#3 treated with 5 μM olanzapine for 16 h (<b>b</b>). Bar graph showing quantification of P-PKCζ T560 normalized to cell area; data are expressed as the mean ± SD of three independent experiments (<b>c</b>,<b>d</b>). Evaluation of intracellular acidic compartments, based on Lysotracker red staining, in ADSC#3 cells after 16-h treatment with SGAs alone or in combination with Go6850 or PKCζ inhibitory pseudosubstrate (PS-PKCζ); nuclei were stained using Hoechst 33342 (<b>e</b>). Bar graph showing acidic vesicle quantification in cells treated for 16 h with olanzapine (5 μM) or clozapine (5 μM) alone, or in combination with Go6850 or PS-PKCζ; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control and expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>f</b>). Representative images showing acidic vesicle accumulation in ADSC#3 transfected with SiRNA NT and SiRNA PKCζ and treated with olanzapine or clozapine for 16 h; nuclei were stained using Hoechst 33342 (<b>g</b>). Bar graph showing acidic vesicle quantification in ADSC#3 cells silenced for PKCζ and treated for 16 h with olanzapine or clozapine; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control and expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>h</b>). Representative images of confocal microscopy analysis of TFEB localization using anti-TFEB primary antibody and Alexa Fluor 488 secondary antibody in ADSC#3 treated with olanzapine alone or in combination with PS-PKCζ (<b>i</b>). Bar graph showing quantification of TFEB nuclear localization expressed as TFEB mean fluorescence in nuclear area normalized as fold change relative to control of three independent experiments (<b>j</b>). ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7 Cont.
<p>PKCζ-dependent expansion of acidic vesicles mediated by SGAs. Western blot analysis of ADSCs treated with 5 μM SGAs for 16 h. Lysates were analyzed for P-(Ser)-PKC substrate, P-PKCζ T560, total PKCζ, and tubulin (<b>a</b>). Confocal microscopy experiments showing P-PKCζ T560 localization in ADSC#3 treated with 5 μM olanzapine for 16 h (<b>b</b>). Bar graph showing quantification of P-PKCζ T560 normalized to cell area; data are expressed as the mean ± SD of three independent experiments (<b>c</b>,<b>d</b>). Evaluation of intracellular acidic compartments, based on Lysotracker red staining, in ADSC#3 cells after 16-h treatment with SGAs alone or in combination with Go6850 or PKCζ inhibitory pseudosubstrate (PS-PKCζ); nuclei were stained using Hoechst 33342 (<b>e</b>). Bar graph showing acidic vesicle quantification in cells treated for 16 h with olanzapine (5 μM) or clozapine (5 μM) alone, or in combination with Go6850 or PS-PKCζ; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control and expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>f</b>). Representative images showing acidic vesicle accumulation in ADSC#3 transfected with SiRNA NT and SiRNA PKCζ and treated with olanzapine or clozapine for 16 h; nuclei were stained using Hoechst 33342 (<b>g</b>). Bar graph showing acidic vesicle quantification in ADSC#3 cells silenced for PKCζ and treated for 16 h with olanzapine or clozapine; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control and expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate (<b>h</b>). Representative images of confocal microscopy analysis of TFEB localization using anti-TFEB primary antibody and Alexa Fluor 488 secondary antibody in ADSC#3 treated with olanzapine alone or in combination with PS-PKCζ (<b>i</b>). Bar graph showing quantification of TFEB nuclear localization expressed as TFEB mean fluorescence in nuclear area normalized as fold change relative to control of three independent experiments (<b>j</b>). ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Olanzapine-induced metabolic alterations in ADSCs are PKCζ-dependent. Representative Western blot of ADSC#3 cells after 16 h of pretreatment with 5 µM olanzapine, either alone or in the presence of PS-PKCζ, followed by insulin stimulation (50 ng/mL) for 30 min. Lysates were analyzed for P-INSRβ Y1146 and total INSRβ (<b>a</b>). The bar graph shows the quantification of P-INSRβ Y1146 normalized to total INSRβ, expressed as fold change relative to control; data are presented as mean ± SD from three independent experiments (<b>b</b>). Representative images of ADSC#3 stimulated with insulin (50 ng/mL) following 16-h treatment with 5 μM olanzapine alone or in combination with PKCζ inhibitory pseudosubstrate showing INSRβ localization on the plasma membrane and in late endosomes; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 546 (red); RAB7 was stained using anti-RAB7 primary antibody and secondary Alexa Fluor 488 (green); and actin was stained using phalloidin 633 (<b>c</b>). Bar graph showing colocalization of INSRβ and actin (<b>d</b>) or RAB7 (<b>e</b>) in ADSC#3 expressed as Pearson coefficient; data are expressed as the mean ± SD of three independent experiments. Bar graph showing colocalization of INSRβ and actin or RAB7 in ADSC#5 expressed as Pearson coefficient; data are expressed as the mean ± SD of three independent experiments (<b>f</b>,<b>g</b>). Representative images of ADSC#3 transfected with siRNA-targeting PKCζ and stimulated with insulin (50 ng/mL) after 16 h of treatment with 5 μM olanzapine. The images show INSRβ localization on the plasma membrane and within late endosomes; INSRβ was stained using anti-INSRβ primary antibody and secondary Alexa Fluor 546 (red); RAB7 was stained using anti-RAB7 primary antibody and secondary Alexa Fluor 488 (green); and actin was stained using phalloidin 633 (<b>h</b>). Bar graph showing colocalization of INSRβ and actin on plasma membrane expressed as Pearson coefficient in ADSC#3 and #5; data are expressed as the mean ± SD of three independent experiments (<b>i</b>,<b>k</b>). Bar graph showing quantification of colocalization of INSRβ with late endosome marker RAB7 expressed as Pearson coefficient in ADSC#3 and #5; results are expressed as the mean ± SD of three independent experiments (<b>j</b>,<b>l</b>). White arrows indicates colocalization spots. *, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.05; ***, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.001; ****, Student’s <span class="html-italic">t</span>-test <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 9
<p>Olanzapine activates PKCζ by modulating GPCR signaling. PA accumulation was analyzed in 3T3L1 cells transfected with Pii-PA (PA indicator with superior sensitivity) DOCK2 (DOCK2-Pii) [<a href="#B34-cells-13-02084" class="html-bibr">34</a>] after 16 h treatment with olanzapine alone or in combination with PLD inhibitor FIPI, EPAC inhibitor CE3F4, and Gq/11 inhibitor YM254890. Representative images of transfected cells treated with DMSO, negative control, 5 μM olanzapine alone or in combination with 750 nM FIPI. Arrows point to dots representing PA accumulation (<b>a</b>). Bar graph quantification of green dots normalized on cell area and expressed as fold change relative to control; results are expressed as the mean ± SD of three independent experiments (<b>b</b>). Bar graphs showing acidic vesicle accumulation in ADSC#3 (<b>c</b>) and ADSC#5 (<b>d</b>) treated for 16 h with olanzapine or clozapine (5 μM) alone or in combination with 750 nM Fipi, 10 μM EPAC inhibitor, 10 μM SQ22, 10 μM Suramin, or 10 μM YM254890; data are expressed as quantification of red Lysotracker staining/blue nuclei staining ratio as fold change relative to negative control and are expressed as the mean ± SD of a representative experiment out of three independent experiments performed in triplicate. Confocal microscopy assessment of P-PKCζ T560 expression in cells treated with 5 μM olanzapine/vehicle alone or in combination with 750 nM Fipi or 10 μM YM254890 for 16 h; phosphorylated PKC was evaluated using P-PKCζ T560 primary antibody and Alexa Fluor 546 secondary antibody, while actin was stained using Phalloidin 633 (<b>e</b>). Bar graph showing quantification of P-PKCζ T560 normalized to cell area; data are expressed as mean ± SD from three independent experiments (<b>f</b>). ** <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 (Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 10
<p>Proposed mechanism of olanzapine-induced metabolic disruption. Olanzapine’s effects are mediated by Gαq and Gαs, initiating signaling cascades that activate phospholipase D and PKCζ. PKCζ disrupts insulin signaling and impairs INSR turnover. Through PP2A activation, this leads to TFEB dephosphorylation and nuclear translocation, promoting lysosomal biogenesis. The combined effects of lysosomal accumulation and PKCζ-induced disruption of INSR phosphorylation further impair insulin signaling and INSR turnover. Image created in <a href="https://BioRender.com" target="_blank">https://BioRender.com</a> (accessed on 30 October 2024).</p>
Full article ">
13 pages, 2263 KiB  
Article
10(E)-Pentadecenoic Acid Inhibits Melanogenesis Partly Through Suppressing the Intracellular MITF/Tyrosinase Axis
by Min-Kyeong Lee, Kyoung Mi Moon, Su-Yeon Park, Jaeseong Seo, Ah-Reum Kim and Bonggi Lee
Antioxidants 2024, 13(12), 1547; https://doi.org/10.3390/antiox13121547 - 17 Dec 2024
Viewed by 437
Abstract
Melanogenesis, the biological process responsible for melanin synthesis, plays a crucial role in determining skin and hair color, photoprotection, and serving as a biomarker in various diseases. While various factors regulate melanogenesis, the role of fatty acids in this process remains underexplored. This [...] Read more.
Melanogenesis, the biological process responsible for melanin synthesis, plays a crucial role in determining skin and hair color, photoprotection, and serving as a biomarker in various diseases. While various factors regulate melanogenesis, the role of fatty acids in this process remains underexplored. This study investigated the anti-melanogenic properties of 10(E)-pentadecenoic acid (10E-PDA) through both in silico and in vitro analyses. SwissSimilarity was utilized to predict the functional properties of 10E-PDA by comparing it with structurally similar lipids known to exhibit anti-melanogenic effects. Subsequent in vitro experiments demonstrated that 10E-PDA significantly reduced melanin production and intracellular tyrosinase activity in α-MSH (melanocyte-stimulating hormone)-stimulated B16F10 melanoma cells without exhibiting significant cytotoxicity at concentrations up to 15 μM. Further mechanistic studies revealed that 10E-PDA inhibited the nuclear translocation of microphthalmia-associated transcription factor (MITF), consistent with the decrease observed in p-MITF protein levels. It also decreased the mRNA levels of tyrosinase-related proteins (TRP-1, TRP-2) and tyrosinase, while reducing the protein levels of TRP-1 and tyrosinase, but not TRP-2. These findings suggest that 10E-PDA exerts its anti-melanogenic effects by modulating the MITF/tyrosinase axis, presenting potential therapeutic implications for skin pigmentation disorders. Full article
(This article belongs to the Special Issue Antioxidants for Skin Health)
Show Figures

Figure 1

Figure 1
<p>Similarity analysis of 10E-PDA with other compounds. SwissSimilarity was used to compare 10E-PDA with structurally similar compounds to predict its potential functions in the skin. 10E-PDA, which contains a trans double bond between the 10th and 11th carbons, exhibited very close similarity to compounds such as palmitoleic acid, cis-vaccenic acid, oleic acid, myristoleic acid, α-linolenic acid, elaidic acid, gondoic acid, and linoleic acid, with similarity scores ranging from 0.998 to 0.999. Among these, several compounds, including palmitoleic acid, α-linolenic acid, and linoleic acid, are known to have anti-melanogenic properties.</p>
Full article ">Figure 2
<p>Effects of 10E-PDA on inhibiting melanogenesis in α-MSH-stimulated B16F10 melanoma cells. (<b>a</b>) B16F10, (<b>b</b>) HS68, and (<b>c</b>) HaCaT cells were treated with increasing concentrations of 10E-PDA (1–100 μM) for 24 h, after which cell viability was assessed (n = 4 per group). (<b>d</b>,<b>e</b>) B16F10 cells were pretreated with different concentrations of 10E-PDA (1–15 μM) or kojic acid (30 μM) for 1 h, followed by exposure to α-MSH (500 nM) for 6 days to measure melanin content (<b>d</b>) or for 3 days to examine tyrosinase activity (<b>e</b>). In both panels, the white bar represents the untreated control group without α-MSH stimulation, the black bar represents the α-MSH-stimulated control group, and the dark gray bars indicate the 10E-PDA-treated groups under α-MSH stimulation. In panel (<b>e</b>), the light gray bar represents the positive control group treated with kojic acid under α-MSH stimulation. Data are shown as mean ± SEM. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the untreated control group, and * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared with the α-MSH-treated group.</p>
Full article ">Figure 3
<p>Cell-free tyrosinase and antioxidant activities of 10E-PDA. (<b>a</b>) Tyrosinase activity of 10E-PDA was measured using a mushroom tyrosinase activity assay. (<b>b</b>) The antioxidant capacity of 10E-PDA was evaluated using DPPH radical scavenging activity. (<b>c</b>) The copper ion reduction capacity of 10E-PDA was determined using CUPRAC analysis, with results compared with the reference ascorbic acid. In all panels, the white bar represents the untreated control group, and the black bars indicate the 10E-PDA-treated groups at various concentrations (1–60 μM). Data are presented as mean ± standard error (n = 3). * <span class="html-italic">p</span> &lt; 0.05 compared with the untreated control group.</p>
Full article ">Figure 4
<p>Impact of 10E-PDA on gene and protein expression of melanogenesis-related targets. To analyze the effect of 10E-PDA on melanogenesis markers, before stimulation with α-MSH (500 nM) over six days, B16F10 melanoma cells were pre-treated with 10E-PDA at concentrations of 5 and 15 μM for one hour. Quantitative PCR (qPCR) was employed to measure mRNA expression levels for (<b>a</b>) TRP-1, (<b>b</b>) TRP-2, and (<b>c</b>) tyrosinase. Additionally, Western blot analysis was conducted to evaluate protein levels of (<b>d</b>) TRP-1, (<b>e</b>) TRP-2, (<b>f</b>) tyrosinase, and (<b>g</b>) p-MITF across groups (n = 3/group). In all panels, the white bar represents the untreated control group (no α-MSH stimulation), the black bar represents the α-MSH-stimulated control group, and the dark gray bars indicate the 10E-PDA-treated groups (α-MSH-stimulated). Results are presented as mean ± SEM. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 compared with the non-treated control group, and * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 compared with the α-MSH-treated group.</p>
Full article ">Figure 5
<p>Effects of 10E-PDA on MITF translocation. Immunofluorescence analysis was performed to evaluate the inhibitory effect of 10E-PDA on the nuclear translocation of MITF. B16F10 cells were treated with 10E-PDA (5 and 15 μM) and linolenic acid (15 μM), and fluorescence microscopy evaluation was performed. MITF was detected using anti-MITF monoclonal antibody and then detected using FSD<sup>TM</sup>-conjugated secondary antibody. This analysis revealed the localization of MITF, thereby elucidating the effect of 10E-PDA on MITF nuclear translocation (Scale bars represent 200 μm).</p>
Full article ">
13 pages, 1326 KiB  
Article
Sex and Age Differences in Glucocorticoid Signaling After an Aversive Experience in Mice
by Yun Li, Bin Zhang, Youhua Yang, Ping Su, James Nicholas Samsom, Albert H. C. Wong and Fang Liu
Cells 2024, 13(24), 2041; https://doi.org/10.3390/cells13242041 - 10 Dec 2024
Viewed by 788
Abstract
Background: glucocorticoids may play an important role in the formation of fear memory, which is relevant to the neurobiology of post-traumatic stress disorder (PTSD). In our previous study, we showed the glucocorticoid receptor (GR) forms a protein complex with FKBP51, which prevents translocation [...] Read more.
Background: glucocorticoids may play an important role in the formation of fear memory, which is relevant to the neurobiology of post-traumatic stress disorder (PTSD). In our previous study, we showed the glucocorticoid receptor (GR) forms a protein complex with FKBP51, which prevents translocation of GR into the nucleus to affect gene expression; this complex is elevated in PTSD patients and by fear-conditioned learning in mice, and disrupting this complex blocks the storage and retrieval of fear-conditioned memories. The timing of release of glucocorticoid relative to the formation of a traumatic memory could be important in this process, and remains poorly understood. Methods and Results: we mapped serum corticosterone over time after fear conditioning in cardiac blood samples from male and female mice, as well as adult and aged mice using ELISA. We show a significant alteration in serum corticosterone after conditioning; notably, levels spike after 30 min but drop lower than unconditioned controls after 24 h. We further investigate the effect of glucocorticoid on GR phosphorylation and localization in HEK 293T cells by Western blot. Hydrocortisone treatment promotes phosphorylation and nuclear translocation of GR. Conclusions: these data contribute to our understanding of the processes linking stress responses to molecular signals and fear memory, which is relevant to understanding the shared mechanisms related to PTSD. Full article
Show Figures

Figure 1

Figure 1
<p>Successful establishment of fear-conditioned mouse model. (<b>A</b>) A schematic illustration schedule for fear conditioning. The conditioned stimulus (CS) was a white light illuminated for 30 s, the unconditioned stimulus (US) was a 1 s 0.5 mA foot shock. Conditioned animals received 5 CS-US pairings, control animals received the CS alone. (<b>B</b>) Time spent freezing during the 3 min CS presentation on day 5 in adult male (8-week-old) mice. Conditioned animals showed significantly more freezing behavior compared to controls. **** <span class="html-italic">p</span> &lt; 0.0001, n = 16, permutation test.</p>
Full article ">Figure 2
<p>Fear conditioning affects the time course of corticosterone levels in mice. (<b>A</b>) Corticosterone protein-expression response curves over 24 h in unconditioned (Control) and fear-conditioned animals (Conditioned). Comparison of corticosterone levels over time in conditioned (dashed) and unconditioned (solid) adult (8-week-old) male (<b>B</b>), adult female (<b>C</b>), aged (64-week-old) male (<b>D</b>), and aged female (<b>E</b>) mice. Data are shown as mean ± SEM, 4-way ANOVA (n = 4 *, female aged control n = 3), -corrected post hoc marginal means. Significance indicators: <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, <sup>####</sup> <span class="html-italic">p</span> &lt; 0.0001 relative to 0 h; <sup><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.05, <sup><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.001, <sup><span>$</span><span>$</span><span>$</span><span>$</span></sup> <span class="html-italic">p</span> &lt; 0.0001 relative to 0.5 h; <sup>%%</sup> <span class="html-italic">p</span> &lt; 0.01 relative to 1 h; <sup>^</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>^^</sup> <span class="html-italic">p</span> &lt; 0.01 relative to 2 h, <sup>&amp;&amp;&amp;</sup> <span class="html-italic">p</span> &lt; 0.001 relative to 4 h, <sup>†</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>††</sup> <span class="html-italic">p</span> &lt; 0.005, <sup>††††</sup> <span class="html-italic">p</span> &lt; 0.0001 relative to 8 h; <sup>‡</sup> <span class="html-italic">p</span> &lt; 0.05 relative to 12 h. * Control relative to Conditioned; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>Female mice have increased corticosterone over time, relative to males, after fear conditioning. Comparison of corticosterone levels over time in male (solid) and female (dashed) adult (8-week-old) unconditioned control mice (<b>A</b>), adult conditioned (<b>B</b>), aged (64-week-old) control (<b>C</b>), and aged conditioned (<b>D</b>) mice. Data are shown as mean ± SEM, 4-way ANOVA (n = 4 *, female aged control n = 3), fdr-corrected post hoc marginal means, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Age reduces corticosterone levels and reactivity to fear conditioning. Comparison of corticosterone levels over time in adult (8-week-old) (solid) and aged (64-week-old) (dashed) male unconditioned control mice (<b>A</b>), male conditioned (<b>B</b>), female control (<b>C</b>), and female conditioned (<b>D</b>) mice. Data are shown as mean ± SEM, 4-way ANOVA (n = 4 *, female aged control n = 3), fdr-corrected post hoc marginal means, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>Hydrocortisone treatment stimulates Ser211 phosphorylation of glucocorticoid receptor (GR) and promotes nuclear translocation of GR in HEK 293T cells. (<b>A</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of phosphorylated GR-S211 (pGR<sup>Ser211</sup>) in HEK 293T whole-cell lysates stimulated by different concentrations of hydrocortisone. The level of pGR<sup>Ser211</sup> is expressed as a ratio relative to levels of unphosphorylated GR normalized to the vehicle (DMSO) alone condition. (<b>B</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the levels of pGR<sup>Ser211</sup> in HEK 293T cells stimulated by hydrocortisone (100 nM) over time. pGR<sup>Ser211</sup>/GR ratios were normalized to time 0. (<b>C</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the changes in cytoplasmic GR protein expression in HEK 293T cells stimulated by different concentrations of hydrocortisone. GR levels expressed relative to α–tubulin and normalized to vehicle (DMSO). (<b>D</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the changes in nuclear GR protein expression in HEK 293T cells stimulated by different concentrations of hydrocortisone. GR levels expressed relative to histone H3 and normalized to vehicle (DMSO). Data are shown as mean ± SEM, one-way ANOVA (n = 3), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 5 Cont.
<p>Hydrocortisone treatment stimulates Ser211 phosphorylation of glucocorticoid receptor (GR) and promotes nuclear translocation of GR in HEK 293T cells. (<b>A</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of phosphorylated GR-S211 (pGR<sup>Ser211</sup>) in HEK 293T whole-cell lysates stimulated by different concentrations of hydrocortisone. The level of pGR<sup>Ser211</sup> is expressed as a ratio relative to levels of unphosphorylated GR normalized to the vehicle (DMSO) alone condition. (<b>B</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the levels of pGR<sup>Ser211</sup> in HEK 293T cells stimulated by hydrocortisone (100 nM) over time. pGR<sup>Ser211</sup>/GR ratios were normalized to time 0. (<b>C</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the changes in cytoplasmic GR protein expression in HEK 293T cells stimulated by different concentrations of hydrocortisone. GR levels expressed relative to α–tubulin and normalized to vehicle (DMSO). (<b>D</b>) Representative Western blots (<b>top</b>) and densitometric analysis (<b>bottom</b>) of the changes in nuclear GR protein expression in HEK 293T cells stimulated by different concentrations of hydrocortisone. GR levels expressed relative to histone H3 and normalized to vehicle (DMSO). Data are shown as mean ± SEM, one-way ANOVA (n = 3), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
18 pages, 2463 KiB  
Article
Interaction Between the Matrix Protein and the Polymerase Complex of Respiratory Syncytial Virus
by Elliot B. Atchison, Sarah N. Croft, Cynthia Mathew, Daniel W. Brookes, Matthew Coates, Kazuhiro Ito and Reena Ghildyal
Viruses 2024, 16(12), 1881; https://doi.org/10.3390/v16121881 - 4 Dec 2024
Viewed by 714
Abstract
The global burden of respiratory syncytial virus (RSV) and severe associated disease is prodigious. RSV-specific vaccines have been launched recently but there is no antiviral medicine commercially available. RSV polymerase (L) protein is one of the promising antiviral targets, along with fusion and [...] Read more.
The global burden of respiratory syncytial virus (RSV) and severe associated disease is prodigious. RSV-specific vaccines have been launched recently but there is no antiviral medicine commercially available. RSV polymerase (L) protein is one of the promising antiviral targets, along with fusion and nucleocapsid proteins. During medicinal chemistry campaigns, two potent L-protein inhibitors (PC786 and PC751) were identified. Both compounds inhibited the RSV A/B-induced cytopathic effect in HEp-2 cells equally, but PC786 was more potent than PC751 in bronchial epithelial cells. Repeated treatment with escalating concentrations on RSV A2-infected HEp-2 cells revealed both inhibitors led to a Y1631H mutation in the L protein, but only PC786 induced a mutation in the M protein (V153A). By L protein fragment and M protein binding analysis, we showed that the M protein interacts with the 1392–1735 amino acid region of the L protein, where PC786 potentially binds. In addition, PC786 treatment or PC786-induced mutant RSV was found to increase M-protein nuclear localisation later in infection, concomitant with delayed fusion protein localisation at the budding viral filaments. As M protein is known to play a key role in virus assembly and budding late in infection, our data suggests that disrupting the interaction between the M and L proteins could provide a novel target for antiviral development. Full article
(This article belongs to the Section Viral Immunology, Vaccines, and Antivirals)
Show Figures

Figure 1

Figure 1
<p>Changes in viral load in vehicle, PC786- and PC751-treated ALI-cultured bronchial epithelium post RSV (0.002 MOI) apical inoculation. PC786 or PC751 was added to the apical surface, once daily from Day 0 (1 h after virus inoculation) to Day 6. The assay was conducted in triplicate, and the graph indicates geometric mean with 95% CI.</p>
Full article ">Figure 2
<p>SNP genotype analysis of the L-gene of RSV A2 treated with PC786 (<b>A</b>), PC751 (<b>B</b>) or wild-type RSV A2 (passage 6), and also of the M-gene of RSV A2 treated with PC786 (<b>C</b>). The axes show the relative fluorescence of the probes specific to either the WT or mutant sequence of the L-gene or M-gene.</p>
Full article ">Figure 3
<p>PC786 treatment results in increased nuclear retention of GFP-M. Cells transfected to express GFP-Mwt were treated with 100 nM PC751 or PC786, DMSO (vehicle), or left untreated (NT) for 12 h before imaging live at 24 h post transfection. Fn/c for M protein was calculated using the formula Fn/c = (Fn − Fb)/(Fc − Fb), where Fn = nuclear fluorescence, Fc = cytoplasmic fluorescence, Fb = background autofluorescence. Unpaired Student’s <span class="html-italic">t</span> test was used to determine statistical differences indicated on the graphs. Statistical significance was set at <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, ****—<span class="html-italic">p</span> &lt; 0.00001, ns—non significant.</p>
Full article ">Figure 4
<p>A2-786 has abnormal assembly and budding compared to wild-type A2. HEp2 cells were infected with A2 wild-type or its mutants, A2-751 (mutation in L only), A2-786 (mutation in L and M) at MOI = 1. Cells were fixed at different times, permeabilised and probed with anti-RSV antibody (recognises F, N, P, M proteins) and either anti-F protein or anti-M protein antibodies. Cells were incubated with Alexa488 (green) or Alexa547 (red) secondary antibodies, stained with Hoechst (blue nuclear stain) and mounted on slides. Slides were viewed on Nikon Ti confocal system and digital images collected as described in text. Single images representative of at least 15 images from two different experiments are shown. Colocaliation is denoted by yellow colour in the images labelled ‘merge’. (<b>A</b>,<b>B</b>) Cells fixed at 18 h post infection. (<b>C</b>,<b>D</b>) Cells fixed at 30 h (A2-751) or 36 h (A2-786) post-infection. (<b>E</b>) A2-786 infected cells fixed at 42 h post-infection. (<b>F</b>) The Fn/c for M at 18 h and 30 h post-infection was calculated (as in <a href="#viruses-16-01881-f003" class="html-fig">Figure 3</a>) for digital images such as shown in (<b>A</b>–<b>D</b>). Unpaired Student’s <span class="html-italic">t</span>-test was used to determine statistical differences indicated on the graph. Statistical significance was set at <span class="html-italic">p</span> &lt; 0.05. **—<span class="html-italic">p</span> &lt; 0.001, ns—non significant.</p>
Full article ">Figure 5
<p>M protein binds to fragment 6 (1518 aa to 1863 aa) of L protein. (<b>A</b>) Schematic showing the position of the L-protein fragments and the mutation in fragment 6. The cDNA is shown, with L protein below. Red lines indicate the length of each fragment. Arrows point to mutation. (<b>B</b>) Each of the L-gene fragments shown in A was cloned into pET30a vector for expression in bacteria. Each clone was expressed in <span class="html-italic">E. coli</span> and lysate separated into soluble (SF) and insoluble (IF) fractions using B-PER reagent (Pierce). Equivalent volumes of each fraction were separated by SDS-PAGE and stained with Coomassie Brilliant Blue. Molecular weights in kDa are shown on the left and fragment identity on the top of the gel. (<b>C</b>) Protein–protein overlay binding assay. Proteins separated on SDS-PAGE as in B were transferred to a nitrocellulose membrane. The membrane was blocked and probed with purified wild-type M protein. Bound M protein was detected with mouse antibody to M protein followed by secondary antibody and development using ECL. Luminescent bands were detected on a LiCor Odyssey Fc. Molecular weights in kDa are shown on the left and fragment identity above the blot.</p>
Full article ">Figure 6
<p>Interaction between fragment 6 of L and M protein is modulated by Y1631 and V153 in L and M proteins, respectively. (<b>A</b>) L-gene fragments 5, 6, and 7, shown in <a href="#viruses-16-01881-f005" class="html-fig">Figure 5</a>, were cloned into pmCherry-C1, with a N-terminal mCherry (red) fusion tag. Each plasmid was co-transfected into COS-7 cells with wild-type M (Mwt) cloned in frame with an N-terminal EGFP (green) tag. Images were acquired 24 h later by live cell confocal microscopy on a Nikon Ti system using NIS Elements software. Single cells for each co-transfection are shown, representative of at least 20 cells and three independent experiments. Colocalization is indicated by yellow colour in the images labelled ‘merge’. (<b>B</b>) Site-directed mutagenesis was used to mutate fragment 6 (Y1631H) and Mwt (V153A). Mutants were cloned into pmCherry-C1 and pEGFP-DestC, respectively. Plasmids were co-transfected into COS-7 cells to express the requisite L fragment with Mwt or M (V153A), followed by live cell microscopy as for A. (<b>C</b>) Images such as those in A, B were analysed for co-localisation of red and green pixels using the Coloc2 plugin within Fiji. Mean ± SEM of Pearson’s correlation coefficient is depicted. L6m—mutated fragment 6 (Y1631H). Mm—mutated M (V153A). Statistical significance was set at <span class="html-italic">p</span> &lt; 0.05. **—<span class="html-italic">p</span> &lt; 0.001, ns—non significant.</p>
Full article ">
23 pages, 7444 KiB  
Article
Monocyte/Macrophage-Specific Loss of ARNTL Suppresses Chronic Kidney Disease-Associated Cardiac Impairment
by Yuya Yoshida, Naoki Nishikawa, Kohei Fukuoka, Akito Tsuruta, Kaita Otsuki, Taiki Fukuda, Yuma Terada, Tomohito Tanihara, Taisei Kumamoto, Ryotaro Tsukamoto, Takumi Nishi, Kosuke Oyama, Kengo Hamamura, Kouta Mayanagi, Satoru Koyanagi, Shigehiro Ohdo and Naoya Matsunaga
Int. J. Mol. Sci. 2024, 25(23), 13009; https://doi.org/10.3390/ijms252313009 - 3 Dec 2024
Viewed by 710
Abstract
Defects in Aryl hydrocarbon receptor nuclear translocator-like 1 (ARNTL), a central component of the circadian clock mechanism, may promote or inhibit the induction of inflammation by monocytes/macrophages, with varying effects on different diseases. However, ARNTL’s role in monocytes/macrophages under chronic kidney disease (CKD), [...] Read more.
Defects in Aryl hydrocarbon receptor nuclear translocator-like 1 (ARNTL), a central component of the circadian clock mechanism, may promote or inhibit the induction of inflammation by monocytes/macrophages, with varying effects on different diseases. However, ARNTL’s role in monocytes/macrophages under chronic kidney disease (CKD), which presents with systemic inflammation, is unclear. Here, we report that the expression of Arntl in monocytes promoted CKD-induced cardiac damage. The expression of G-protein-coupled receptor 68 (GPR68), which exacerbates CKD-induced cardiac disease, was regulated by ARNTL. Under CKD conditions, GPR68 expression was elevated via ARNTL, particularly in the presence of PU.1, a transcription factor specific to monocytes and macrophages. In CKD mouse models lacking monocyte-specific ARNTL, GPR68 expression in monocytes was reduced, leading to decreased cardiac damage and fibrosis despite no improvement in renal excretory capacity or renal fibrosis and increased angiotensin II production. The loss of ARNTL did not affect the expression of marker molecules, indicating the origin or differentiation of cardiac macrophages, but affected GPR68 expression only in cardiac macrophages derived from mature monocytes, highlighting the significance of the interplay between GPR68 and ARNTL in monocytes/macrophages and its influence on cardiac pathology. Understanding this complex relationship between circadian clock mechanisms and disease could help uncover novel therapeutic strategies. Full article
(This article belongs to the Topic Molecular and Cellular Mechanisms of Heart Disease)
Show Figures

Figure 1

Figure 1
<p>Effect of ARNTL on induction of GPR68 expression in RAW264.7 and mouse primary monocytes by 5/6Nx-derived serum. (<b>A</b>) mRNA of <span class="html-italic">Arntl</span> in RAW264.7 incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>B</b>) Transcriptional regulation of <span class="html-italic">Gpr68</span> using serum prepared from Sham or 5/6Nx mice. Number of nucleotide residues indicates distance from transcription start site (+1). RAW264.7 cells were transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc, <span class="html-italic">Gpr68</span> (-1512)-Luc, <span class="html-italic">Gpr68</span> (-1261)-Luc, <span class="html-italic">Gpr68</span> (-27)-Luc, or pGL4.18. Values are expressed as mean ± S.D. (<span class="html-italic">n</span> = 4). (<b>C</b>) Influence of CLOCK/ARNTL on transcriptional activity of mouse <span class="html-italic">GPR68</span>. RAW264.7 cells were transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc in presence or absence of CLOCK and ARNTL-expressing vectors. Relative luciferase activity of pGL4.18-transfected cells in absence of CLOCK/ARNTL was set at 1.0. (<b>D</b>) High-ARNTL-expressing RAW264.7 was created by introducing an ARNTL expression plasmid. ARNTL expression levels were measured using Western blotting. (<b>E</b>) Protein levels of GPR68 in RAW264.7-transfected pcDNA3.1 or ARNTL-expressing vectors. (<b>F</b>) Loss of <span class="html-italic">Arntl</span> caused by CRE-LOXP system resulted in loss of ARNTL protein in monocytes. Monocytes isolated from monocytic <span class="html-italic">ARNTL</span> +/+ mice or monocytic <span class="html-italic">ARNTL</span> −/− mice. (<b>G</b>) Expression of <span class="html-italic">Gpr68</span> mRNA in primary cultured monocytes, which were isolated from monocytic <span class="html-italic">ARNTL</span> +/+ mice or monocytic <span class="html-italic">ARNTL</span> −/− mice. mRNA levels of <span class="html-italic">Gpr68</span> were assessed after treatment with serum from Sham or 5/6Nx WT mice for 24 h. Values are expressed as mean ± S.D. (<span class="html-italic">n</span> = 4–6). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between two groups (two-way ANOVA with Tukey–Kramer post hoc tests or Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>The effect of monocyte/macrophage-specific transcription factor PU.1 on the induction of GPR68 expression. (<b>A</b>) The 5/6Nx-derived serum did not increase the transcriptional activity upstream of <span class="html-italic">Gpr68</span> in NIH3T3. NIH3T3 was transfected with <span class="html-italic">Gpr68</span> (-1734)-Luc or pGL4.18 and incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>B</b>) Transcription factors binding upstream of <span class="html-italic">Gpr68</span> analyzed by previous transcriptome analyses. The blue waveform shows the sequenced tags in ChIP sequence analysis for each transcription factor. The numbers on the horizontal axis indicate the distance from the transcription start site (kbp). (<b>C</b>) The PU.1 protein in RAW264.7 incubated with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>D</b>) High-PU.1-expressing NIH3T3 was created by introducing a PU.1 expression plasmid. PU.1 expression levels were measured using Western blotting. (<b>E</b>,<b>F</b>) The mRNA levels of <span class="html-italic">Gpr68</span> (<b>E</b>) and <span class="html-italic">Arntl</span> (<b>F</b>) in NIH3T3-transfected pcDNA3.1 or PU.1-expressing vectors were measured after incubation with 10% serum from Sham and 5/6Nx mice for 24 h. (<b>G</b>) A schematic of mouse <span class="html-italic">Gpr68</span>. The numbers indicate the distance from the transcription start site (+1). Black rectangles, E-box. The arrow symbols indicate the location on the gene where the primer sets localize for the analysis of ChIP. (<b>H</b>) The binding of endogenous ARNTL to the <span class="html-italic">Gpr68</span> upstream region in NIH3T3-transfected pcDNA3.1 or PU.1-expressing vectors. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 3–5). **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests or Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>The effect of the loss of monocyte-specific ARNTL on the 5/6Nx-induced induction of GPR68 expression. (<b>A</b>) The binding of endogenous ARNTL or CLOCK to the <span class="html-italic">Gpr68</span> upstream region in Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice in the blood. The primer sets used are shown in <a href="#ijms-25-13009-f002" class="html-fig">Figure 2</a>G. (<b>B</b>) The expression of <span class="html-italic">Gpr68</span> mRNA in Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice in the blood and spleen. (<b>C</b>,<b>D</b>) Flow cytometry analysis was performed to detect high-GPR68-expressing Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells in the blood and spleen. The ratio of high-GPR68-expressing monocytes in the blood and spleen. For all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–7). **, <span class="html-italic">p</span> &lt; 0.01, ** indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 4
<p>The effect of the deficiency of monocyte-specific ARNTL on 5/6Nx-induced cardiac injury. (<b>A</b>) Serum BNP concentrations in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 6). (<b>B</b>,<b>C</b>) Mutations in <span class="html-italic">Arntl</span> in monocytes ameliorated CKD-induced cardiac fibrosis. Panel (B) shows Masson’s trichrome staining of tissue fibrosis in blue. Scale bars indicate 1 mm (upper panel) and 50 μm (lower panel). Panel (<b>C</b>) shows the quantification of the fibrosis area under light microscopy. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). (<b>D</b>) The total amount of collagen throughout the ventricle. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>E</b>) Cardiac TIMP-1 protein levels in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>F</b>) The mRNA levels of <span class="html-italic">Tnf-α</span> and <span class="html-italic">Il-6</span> and fibrosis-related factors (<span class="html-italic">Col1a1</span>, <span class="html-italic">Col1a2</span>, <span class="html-italic">Mmp1a</span>, <span class="html-italic">Timp-1</span>, and <span class="html-italic">αSma</span>) in the cardiac ventricle of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 5
<p>The effect of the loss of monocyte-specific ARNTL on renal function in 5/6Nx mice. (<b>A</b>) Masson’s trichrome staining for the kidneys prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Scale bars indicate 50 μm. (<b>B</b>) The total amount of collagen throughout the kidney. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>C</b>) Renal TIMP-1 protein levels in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. Values were corrected for total protein mass. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5–6). (<b>D</b>) The mRNA levels of <span class="html-italic">Tnf-α</span> and <span class="html-italic">Il-6</span> and fibrosis-related factors (<span class="html-italic">Col1a1</span>, <span class="html-italic">Col1a2</span>, <span class="html-italic">Mmp1a</span>, <span class="html-italic">Timp-1</span>, and <span class="html-italic">αSma</span>) in the kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. Values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 5). (<b>E</b>–<b>H</b>) The serum concentrations of creatinine (<b>E</b>), urea nitrogen (<b>F</b>), angiotensin II (<b>G</b>), and aldosterone (<b>H</b>), in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. (<b>I</b>) The mRNA levels of <span class="html-italic">Tgf-β</span> in the kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set as 1.0. (<b>J</b>) The serum concentrations of retinol in <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. In all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 4–5). **, <span class="html-italic">p</span> &lt; 0.01; *, <span class="html-italic">p</span> &lt; 0.05 significant difference between the two groups (one-way or two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">Figure 6
<p>The effect of the loss of monocyte-specific ARNTL on the 5/6Nx-induced induction of GPR68 expression. (<b>A</b>,<b>B</b>) The mRNA levels of <span class="html-italic">Vcam1</span> and <span class="html-italic">Sele</span> in the cardiac ventricle or kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. (<b>C</b>,<b>D</b>) The number of cardiac or renal F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> cells (<b>C</b>) and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells (<b>D</b>) in each organ. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group in each organ was set as 1.0. (<b>E</b>) The mRNA levels of <span class="html-italic">Gpr68</span> in the cardiac ventricle or kidney of <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice. The mean value of the Sham-operated <span class="html-italic">ARNTL</span> +/+ group was set to 1.0. (<b>F</b>) The expression of <span class="html-italic">Gpr68</span> mRNA in cardiac F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells prepared from <span class="html-italic">ARNTL</span> +/+ or <span class="html-italic">ARNTL</span> −/− Sham and 5/6Nx mice ventricles. (<b>G</b>) The expression levels in the cardiac F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>+</sup> and F4/80<sup>+</sup>/Ly6G<sup>−</sup>/CD11b<sup>+</sup>/Ly6C<sup>−</sup> cells of markers indicative of a subset of macrophages. Histograms showing the expression of each marker were obtained by flow cytometric analysis. For all panels, values are expressed as the mean ± S.D. (<span class="html-italic">n</span> = 4–7). *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01 indicates significant differences between the two groups (two-way ANOVA with Tukey–Kramer post hoc tests).</p>
Full article ">
24 pages, 2915 KiB  
Article
Detection of Hepatitis C Virus Infection from Patient Sera in Cell Culture Using Semi-Automated Image Analysis
by Noemi Schäfer, Paul Rothhaar, Christian Heuss, Christoph Neumann-Haefelin, Robert Thimme, Julia Dietz, Christoph Sarrazin, Paul Schnitzler, Uta Merle, Sofía Pérez-del-Pulgar, Vibor Laketa and Volker Lohmann
Viruses 2024, 16(12), 1871; https://doi.org/10.3390/v16121871 - 30 Nov 2024
Viewed by 674
Abstract
The study of hepatitis C virus (HCV) replication in cell culture is mainly based on cloned viral isolates requiring adaptation for efficient replication in Huh7 hepatoma cells. The analysis of wild-type (WT) isolates was enabled by the expression of SEC14L2 and by inhibitors [...] Read more.
The study of hepatitis C virus (HCV) replication in cell culture is mainly based on cloned viral isolates requiring adaptation for efficient replication in Huh7 hepatoma cells. The analysis of wild-type (WT) isolates was enabled by the expression of SEC14L2 and by inhibitors targeting deleterious host factors. Here, we aimed to optimize cell culture models to allow infection with HCV from patient sera. We used Huh7-Lunet cells ectopically expressing SEC14L2, CD81, and a GFP reporter with nuclear translocation upon cleavage by the HCV protease to study HCV replication, combined with a drug-based regimen for stimulation of non-modified wild-type isolates. RT-qPCR-based quantification of HCV infections using patient sera suffered from a high background in the daclatasvir-treated controls. We therefore established an automated image analysis pipeline based on imaging of whole wells and iterative training of a machine learning tool, using nuclear GFP localization as a readout for HCV infection. Upon visual validation of hits assigned by the automated image analysis, the method revealed no background in daclatasvir-treated samples. Thereby, infection events were found for 15 of 34 high titer HCV genotype (gt) 1b sera, revealing a significant correlation between serum titer and successful infection. We further show that transfection of viral RNA extracted from sera can be used in this model as well, albeit with so far limited efficiency. Overall, we generated a robust serum infection assay for gt1b isolates using semi-automated image analysis, which was superior to conventional RT-qPCR-based quantification of viral genomes. Full article
(This article belongs to the Special Issue Hepatitis C Virus 2024)
Show Figures

Figure 1

Figure 1
<p>Establishment of a cell culture model to assess HCV WT replication: (<b>A</b>,<b>B</b>,<b>D</b>,<b>F</b>) Subgenomic reporter replicons based on Con1 WT and GLT1 WT were electroporated into Huh7-Lunet C/M/S cells and treated with either DMSO or the indicated concentrations of (<b>A</b>) H479 combined with PI4KA-F1 (PCi), or with PCi and the indicated concentrations of DCV (<b>B</b>)/Legalon SIL (<b>D</b>) or Alisporivir (<b>F</b>) at 4 h after electroporation. Cells were harvested 72 h later and firefly luciferase activity from cell lysates (RLU) was quantified as a correlate of RNA replication efficiency and normalized to 4 h. A replication-deficient Con1 variant (ΔGDD) served as negative control and is indicated with a dashed grey line. Values are shown as means from two (<b>A</b>,<b>B</b>) or three (<b>D</b>,<b>F</b>) independent experiments with two technical replicates each. (<b>C</b>,<b>E</b>,<b>G</b>) Huh7-Lunet C/M/S cells were seeded into 96-well plates and treated with the indicated concentrations of DCV (<b>C</b>)/Legalon SIL (<b>E</b>) or Alisporivir (<b>G</b>). After 72 h, cell viability in cell lysates was measured via Cell-titer-Glow and normalized to the DMSO control. Values are shown as means from at least three independent experiments. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>Comparison of luciferase activity and RT-qPCR-based readouts in persistent replicon cells and HCV infected cells: (<b>A</b>) Cell lines harboring the indicated replicons were treated with 250 µM SIL, 1 µM Alisporivir, 1 nM DCV, or were left untreated, and harvested at the indicated time points. Upper panels: Firefly luciferase activity from cell lysates (RLU) was quantified as a correlate of RNA replication efficiency and normalized to 4 h. Values are shown as means from two independent experiments with two technical replicates each. Lower panels: After RNA extraction, HCV RNA copies were quantified using RT-qPCR. HCV RNA copy numbers (CN) were normalized on GAPDH and are shown as CN per well. Values are shown as means from two independent experiments with three technical replicates each. (<b>B</b>) Huh7-Lunet C/M/S cells were infected with either GLT1 patient serum or JC1cc infectious particles (MOI of 10 at the first repetition, MOI of 1 at the second and third repetition). After 72 h, samples were harvested, and HCV RNA replication was measured via RT-qPCR. HCV RNA copy numbers (CN) were normalized on GAPDH and are shown as CN per well. Values are shown as means of four (GLT1: untr. + DCV), three (JC1cc: untr. + DCV), or two (GLT1 and JC1cc: SIL and Alisporivir treatment) independent experiments with three technical replicates each. <span class="html-italic">p</span>-values from unpaired two-tailed Student’s <span class="html-italic">t</span>-test are indicated for all experiments with 3 or more biological replicates.</p>
Full article ">Figure 3
<p>Workflow for serum infection and establishment of semi-automated image analysis: (<b>A</b>) Workflow for serum infection. (<b>B</b>) Principle of automated image analysis by machine learning. Infected cells are identified by nuclear GFP due to co-localization with DAPI. Positive cells were marked in red by ilastik, facilitating visual validation. (<b>C</b>) Huh7-Lunet C/M/S cells were infected with JC1cc (MOI = 1) alone or additionally treated with DCV, fixed, and stained with DAPI 72 h post-infection. A group of infected cells is indicated by a white box and additionally displayed with a higher magnification. Percentage of positive cells (right panel) was obtained by automated image analysis. *** <span class="html-italic">p</span> &lt; 0.001 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test). (<b>B</b>,<b>C</b>) Scale bar indicates a length of 100 µm. (<b>D</b>) Optimization of the automated image analysis. False-positive background in images of either uninfected or DCV-treated cells was reduced by iterative training of the machine learning tool, including adjustment of cell count per well and refocusing strategy. (<b>E</b>) Workflow of automated image analysis (AIA) followed by visual validation.</p>
Full article ">Figure 4
<p>Comparison of HCV infection using high-titer sera by RT-qPCR or microscopic analysis: (<b>A</b>–<b>C</b>) Huh7-Lunet C/M/S cells treated with PCi and infected with patient sera in presence or absence of DCV; 72 h after infection, cells were fixed, stained with DAPI. (<b>A</b>) Total RNA was extracted 72 h after infection and HCV RNA was quantified. (<b>B</b>) 72 h after infection, quantification by automated image analysis was performed. (<b>C</b>) Truly infected cells detected after visual validation of hits generated by the automated image analysis. Note that no bona fide infected cells were detected in the DCV-treated samples. All values are means and SD of three or two (serum 44) independent biological replicates. Note that RT-qPCR data from the GLT1 serum were taken from <a href="#viruses-16-01871-f002" class="html-fig">Figure 2</a>B to allow a direct comparison (n = 4). n.d.: not detectable. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test). (<b>D</b>) Results of the semi-automated image-based analysis of all data shown in panel (<b>C</b>) and in Figure 6C were analyzed regarding the impact of RNA titer and post-transplant status on infection outcome. The anonymized abbreviation of each serum is given next to the respective dot (see <a href="#viruses-16-01871-t001" class="html-table">Table 1</a> and <a href="#viruses-16-01871-t002" class="html-table">Table 2</a>). Samples were judged positive if at least one infected cell was found via the automated image analysis combined with visual validation. * <span class="html-italic">p</span> &lt; 0.05 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>Establishment of an HCV WT RNA transfection protocol. In vitro transcribed RNA of GLT1 WT (gt1b), TN WT (gt1a), and DBN WT (gt3a) was transfected into Huh7-Lunet C/M/S cells with different amounts of input RNA, ranging from 10<sup>7</sup> to 10<sup>10</sup> RNA copies per transfection, either by electroporation (<b>A</b>,<b>C</b>) or by lipid-based transfection (<b>B</b>,<b>D</b>). RT-qPCR (<b>A</b>,<b>B</b>) and the automated image analysis served as readouts (<b>C</b>,<b>D</b>). Samples were measured in technical triplicates. In the image-based analysis (<b>C</b>,<b>D</b>), the samples transfected with 10<sup>8</sup> RNA copies of TN WT and DBN WT were set as false-positive backgrounds, since no infected cells could be detected upon visual validation, depicted as a dashed grey line. Numbers of cells visually validated represent only DCV- samples. (<b>A</b>,<b>C</b>) Electroporation: Samples were harvested after 72 h. Values are taken from two (TN WT, DBN WT) or three (GLT1 WT) independent experiments. (<b>B</b>,<b>D</b>) Lipid-based transfection: Cells were split 24 h post-transfection, at this time point DCV was added. Samples were harvested after 2 (to stick to the total incubation time of 72 h) or 3 (to ensure DCV treatment for 72 h) days. Since there were no relevant differences in the results with varying incubation times observed, the data of the two experiments are shown in the same graph. * <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 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p><b>Results of RNA transfection in comparison to serum infection</b>. Ten patient sera were tested using RNA transfection and serum infection in parallel. All samples were harvested after 72 h, treatment with DCV served as negative control. Samples were analyzed via RT-qPCR (<b>A</b>), AIA (<b>B</b>), and with AIA followed by visual validation (<b>C</b>). Values are shown as means of three biological replicates and were measured in technical triplicates in RT-qPCR. Data of the serum infection assay of sera 2130, 2213, AU, and GLT1 were taken from <a href="#viruses-16-01871-f004" class="html-fig">Figure 4</a> for better comparison. n.d.: not detectable. ** <span class="html-italic">p</span> &lt; 0.01 (unpaired two-tailed Student’s <span class="html-italic">t</span>-test).</p>
Full article ">
30 pages, 16811 KiB  
Article
Unveiling Smyd-2’s Role in Cytoplasmic Nrf-2 Sequestration and Ferroptosis Induction in Hippocampal Neurons After Cerebral Ischemia/Reperfusion
by Daohang Liu and Yizhun Zhu
Cells 2024, 13(23), 1969; https://doi.org/10.3390/cells13231969 - 28 Nov 2024
Viewed by 784
Abstract
SET and MYND Domain-Containing 2 (Smyd-2), a specific protein lysine methyltransferase (PKMT), influences both histones and non-histones. Its role in cerebral ischemia/reperfusion (CIR), particularly in ferroptosis—a regulated form of cell death driven by lipid peroxidation—remains poorly understood. This study identifies the expression of [...] Read more.
SET and MYND Domain-Containing 2 (Smyd-2), a specific protein lysine methyltransferase (PKMT), influences both histones and non-histones. Its role in cerebral ischemia/reperfusion (CIR), particularly in ferroptosis—a regulated form of cell death driven by lipid peroxidation—remains poorly understood. This study identifies the expression of Smyd-2 in the brain and investigates its relationship with neuronal programmed cell death (PCD). We specifically investigated how Smyd-2 regulates ferroptosis in CIR through its interaction with the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway. Smyd-2 knockout protects HT-22 cells from Erastin-induced ferroptosis but not TNF-α + Smac-mimetic-induced apoptosis/necroptosis. This neuroprotective effect of Smyd-2 knockout in HT-22 cells after Oxygen–Glucose Deprivation/Reperfusion (OGD/R) was reversed by Erastin. Smyd-2 knockout in HT-22 cells shows neuroprotection primarily via the Nuclear Factor Erythroid-2-related Factor-2 (Nrf-2)/Kelch-like ECH-associated protein (Keap-1) pathway, despite the concurrent upregulation of Smyd-2 and Nrf-2 observed in both the middle cerebral artery occlusion (MCAO) and OGD/R models. Interestingly, vivo experiments demonstrated that Smyd-2 knockout significantly reduced ferroptosis and lipid peroxidation in hippocampal neurons following CIR. Moreover, the Nrf-2 inhibitor ML-385 abolished the neuroprotective effects of Smyd-2 knockout, confirming the pivotal role of Nrf-2 in ferroptosis regulation. Cycloheximide (CHX) fails to reduce Nrf-2 expression in Smyd-2 knockout HT-22 cells. Smyd-2 knockout suppresses Nrf-2 lysine methylation, thereby promoting the Nrf-2/Keap-1 pathway without affecting the PKC-δ/Nrf-2 pathway. Conversely, Smyd-2 overexpression disrupts Nrf-2 nuclear translocation, exacerbating ferroptosis and oxidative stress, highlighting its dual regulatory role. This study underscores Smyd-2’s potential for ischemic stroke treatment by disrupting the Smyd-2/Nrf-2-driven antioxidant capacity, leading to hippocampal neuronal ferroptosis. By clarifying the intricate interplay between ferroptosis and oxidative stress via the Nrf-2/Keap-1 pathway, our findings provide new insights into the molecular mechanisms of CIR and identify Smyd-2 as a promising therapeutic target. Full article
Show Figures

Figure 1

Figure 1
<p>MCAO and OGD/R conduce to Smyd-2 activation in the hippocampus and HT-22 cells. (<b>A</b>). Representative images of TTC staining, quantitative analysis of infarct volume, asymmetrical test scores, and the adhesive removal scores in MCAO mice (<span class="html-italic">n</span> = 5). (<b>B</b>). Smyd-2 expression in mouse brain after CIR (<span class="html-italic">n</span> = 5). (<b>C</b>). Smyd-2 expression in Ht-22 cells challenged with OGD/R (<span class="html-italic">n</span> = 5). * <span class="html-italic">p</span> &lt; 0.05 vs. sham group, *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group.</p>
Full article ">Figure 2
<p>The inhibition of Smyd-2 expression delays the progression of CIR impairment. (<b>A</b>–<b>C</b>) Effects of Smyd-2 on cerebral infarction volume and neurobehavioral function in MCAO mice (<span class="html-italic">n</span> = 5). (<b>D</b>) Representative images of FJB staining slices of the hippocampus and cortex; scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>E</b>) Representative images of LFB staining slices of the hippocampus and cortex and quantitative analysis of the breakdown products of myelin sheathes in the hippocampus and cortex; scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>F</b>) The transfection effects of Smyd-2-overexpressing adenovirus and siRNA in Ht-22 cells (<span class="html-italic">n</span> = 5). (<b>G</b>) The neuronal viability, LDH release, SOD level, and MDA level of Ht-22 cells challenged with Smyd-2 siRNA and Smyd-2-overexpressing adenovirus after OGD/R (<span class="html-italic">n</span> = 5). ** <span class="html-italic">p</span> &lt; 0.01 vs. sham group, *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001 vs. CIR group and CIR + IVC-NC group; *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-NC group.</p>
Full article ">Figure 3
<p>Smyd-2 knockout counteracts the effect of OGD/R on lipid peroxidation in Ht-22 cells. (<b>A</b>) BODIPY-581/591-C11 staining was applied to analyze and quantify the effect of Smyd-2 siRNA and adenovirus-mediated Smyd-2 on HT-22 cell ferroptosis challenged with OGD/R. The representative images were obtained with an optical microscope at 400× magnification; scale bars = 50 μm. The GSH level of HT-22 cells challenged with AD-Smyd-2 and Si-Smyd-2 after OGD/R. (<b>B</b>) DCFH-DA staining was applied to analyze and quantify the effect of Smyd-2 siRNA and adenovirus-mediated Smyd-2 on HT-22 cell ferroptosis challenged with OGD/R. The representative images were obtained with a confocal microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 6). (<b>C</b>) Immunofluorescence method was applied to investigate Smyd-2 and GPX-4 protein expression and localization in HT-22 cells and their relation to neuronal ferroptosis after OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-NC group.</p>
Full article ">Figure 4
<p>Smyd-2 regulates the abnormality in neuronal ferroptosis caused by CIR. (<b>A</b>) Representative images of Perls staining slices of the hippocampus and cortex. Scale bars = 40 μm (<span class="html-italic">n</span> = 5). (<b>B</b>–<b>D</b>) Representative images of Western blot and quantitative analysis of the expression of Smyd-2, GPX-4, FTH-1, SLC7A11, ACSL-4, 15-LOX, COX-2, NQO-1, Keap-1, HO-1, nucleus Nrf-2, <span class="html-italic">p</span>-Nrf-2, and Nrf-2 (TP) in the hippocampus. *** <span class="html-italic">p</span> &lt; 0.001 vs. sham group; ### <span class="html-italic">p</span> &lt; 0.001 vs. CIR group and CIR + ICV-NC group.</p>
Full article ">Figure 5
<p>The effect of Smyd-2 overexpression on programmed cell death of HT-22 cells induced by Erastin and Smac mimetic + TNF-α. (<b>A</b>) The neuronal viability and LDH release of HT-22 cells challenged with Erastin. (<b>B</b>,<b>C</b>) The neuronal viability and LDH release of HT-22 cells challenged with Erastin. (c,d,e,f,g,h,i,j) The neuronal viability, LDH release, SOD level, and MDA level of Si-Smyd-2 HT-22 cells challenged with Erastin and Smac mimetic (<span class="html-italic">n</span> = 6). (<b>D</b>) Annexin V/PI staining was applied to observe and analyze the effect of Erastin and Smac mimetic on different types of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 6). *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group, * <span class="html-italic">p</span> &lt; 0.05 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group, # <span class="html-italic">p</span> &lt; 0.05 vs. OGD/R + Si-NC group.</p>
Full article ">Figure 6
<p>The effect of Si-Smyd-2 combined with Erastin on the ferroptosis of HT-22 cells induced by OGD/R. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells challenged with Si-Smyd-2 and Erastin after OGD/R. (<b>B</b>) GSH kit was applied to analyze and quantify the effect of the Si-RNA-mediated Smyd-2 and Erastin on HT-22 cell ferroptosis challenged with OGD/R (<span class="html-italic">n</span> = 5). (<b>C</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by transmission electron microscopy. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The enlarged region bounded by a rectangular dotted box conduces to obtaining a more detailed view of the mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm. The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm (<span class="html-italic">n</span> = 5). (<b>D</b>–<b>F</b>) Representative Western blots and quantitative evaluation of Smyd-2, SLC7A11, ACSL-4, FTH-1, GPX-4, Nrf-2, Keap-1, p-Nrf-2, and nucleus Nrf-2 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + Si-Smyd-2 group.</p>
Full article ">Figure 7
<p>Effects of Smyd-2 (KO) combined with Nrf-2 inhibitor ML-385 on ferroptosis and lipid peroxidation in MCAO mice and OGD/R-induced HT-22 cells. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells were challenged with Si-Smyd-2 and ML-385 after OGD/R (<span class="html-italic">n</span> = 5). (<b>B</b>) BODIPY-581/591-C11 staining, GSH kit, and DCFH-DA staining were used to analyze and quantify the effect of Si-RNA-mediated Smyd-2 and ML-385 on lipid peroxidation of HT-22 cells after OGD/R. The representative images of BODIPY-581/591-C11 staining were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). The representative images of DCFH-DA staining were obtained with an optical microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Annexin V/PI double fluorescence staining was used to study the effect of Si-RNA-mediated Smyd-2 and ML-385 on a different form of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). (<b>D</b>) The effect of Si-RNA-mediated Smyd-2 combined with ML-385 on ferroptosis-related proteins in HT-22 cells induced by OGD/R. Representative Western blots and quantitative evaluation of ACSL-4, Keap-1, SLC7A11, Smyd-2, FTH-1, GPX-4, p-Nrf-2, Nrf-2 (TP), nucleus Nrf-2, PGC-1α, COX-2, 15-LOX, NQO-1, and HO-1 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>E</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by TEM. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The zoom region bounded by a rectangular dotted box allows a more detailed view of mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm (<span class="html-italic">n</span> = 5). The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm. *** <span class="html-italic">p</span> &lt; 0.001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.001 vs. OGD/R group and Si-NC + OGD/R group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001 vs. Si-Smyd-2 + OGD/R group.</p>
Full article ">Figure 7 Cont.
<p>Effects of Smyd-2 (KO) combined with Nrf-2 inhibitor ML-385 on ferroptosis and lipid peroxidation in MCAO mice and OGD/R-induced HT-22 cells. (<b>A</b>) The neuronal viability, LDH release, SOD, and MDA of HT-22 cells were challenged with Si-Smyd-2 and ML-385 after OGD/R (<span class="html-italic">n</span> = 5). (<b>B</b>) BODIPY-581/591-C11 staining, GSH kit, and DCFH-DA staining were used to analyze and quantify the effect of Si-RNA-mediated Smyd-2 and ML-385 on lipid peroxidation of HT-22 cells after OGD/R. The representative images of BODIPY-581/591-C11 staining were obtained with an optical microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). The representative images of DCFH-DA staining were obtained with an optical microscope at 200× magnification; scale bars = 100 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Annexin V/PI double fluorescence staining was used to study the effect of Si-RNA-mediated Smyd-2 and ML-385 on a different form of programmed cell death in HT-22 cells induced by OGD/R. The representative images were obtained with a confocal microscope at 400× magnification; scale bars = 50 μm (<span class="html-italic">n</span> = 5). (<b>D</b>) The effect of Si-RNA-mediated Smyd-2 combined with ML-385 on ferroptosis-related proteins in HT-22 cells induced by OGD/R. Representative Western blots and quantitative evaluation of ACSL-4, Keap-1, SLC7A11, Smyd-2, FTH-1, GPX-4, p-Nrf-2, Nrf-2 (TP), nucleus Nrf-2, PGC-1α, COX-2, 15-LOX, NQO-1, and HO-1 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>E</b>) Pathophysiological and physiological morphologies of mitochondria in each HT-22 cell group were observed by TEM. The red arrows mark the increased electron density of the matrix and fractured and vague cristae. The blue arrows mark vacuoles in mitochondria. The zoom region bounded by a rectangular dotted box allows a more detailed view of mitochondria for each experimental condition. The representative images were obtained with an optical microscope at 8k× magnification; scale bars = 1 μm (<span class="html-italic">n</span> = 5). The representative enlarged images were obtained with an optical microscope at 20k× magnification; scale bars = 500 nm. *** <span class="html-italic">p</span> &lt; 0.001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.001 vs. OGD/R group and Si-NC + OGD/R group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.001 vs. Si-Smyd-2 + OGD/R group.</p>
Full article ">Figure 8
<p>Smyd-2 methylates Nrf-2 (Lys-508) to inhibit OGD/R-induced Nrf-2 (Ser-40) phosphorylation and nuclear translocation. (<b>A</b>) Representative Western blot and quantitative evaluation of p-Nrf-2, Nrf-2 (TP), and nucleus Nrf-2 expression levels in each HT-22 cell group. Data normalized to the loading control GAPDH are expressed as % of control. Data normalized to the loading control GAPDH and histone H-3 are expressed as % of control (<span class="html-italic">n</span> = 5). (<b>B</b>) Confocal images of the localization with immunofluorescence-stained Smyd-2 (green) and immunofluorescence-stained Nrf-2 (red) in various HT-22 cell groups after OGD/R. The zoom region bounded by rectangular boxes represents Smyd-2-Nrf-2 binding in the cytoplasm of HT-22 cells, and the Nrf-2 was transported to the cell nucleus. Scale bars = 10 μm (<span class="html-italic">n</span> = 5). (<b>C</b>) Representative Western blots and quantitative evaluation of Nrf-2 (TP), p-Nrf-2, nucleus Nrf-2, PKC-δ, p-PKC-δ, Smyd-2, and Keap-1 expression levels in each HT-22 cell group. Data normalized to the loading control histone H-3 are expressed as % of GAPDH (<span class="html-italic">n</span> = 5). (<b>D</b>) Quantitative analysis of the expression of the methylation level of Nrf-2 (<span class="html-italic">n</span> = 5). (<b>E</b>) The possible docking sites of two target proteins, Smyd-2/Nrf-2. The binding mode of the complex Nrf-2 with Smyd-2. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + Si-NC group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group; ^^^ <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + CHX group. *** <span class="html-italic">p</span> &lt; 0.0001 vs. control group; ### <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R group and OGD/R + AD-Smyd-2 group; &amp;&amp;&amp; <span class="html-italic">p</span> &lt; 0.0001 vs. OGD/R + AD-Smyd-2 group.</p>
Full article ">
Back to TopTop