[go: up one dir, main page]

 
 
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (563)

Search Parameters:
Keywords = ectopic function

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 3546 KiB  
Article
Impact of Sex on the Therapeutic Efficacy of Rosiglitazone in Modulating White Adipose Tissue Function and Insulin Sensitivity
by Marco Bauzá-Thorbrügge, Emilia Amengual-Cladera, Bel Maria Galmés-Pascual, Andrea Morán-Costoya, Magdalena Gianotti, Adamo Valle, Ana Maria Proenza and Isabel Lladó
Nutrients 2024, 16(18), 3063; https://doi.org/10.3390/nu16183063 - 11 Sep 2024
Viewed by 363
Abstract
Obesity and type 2 diabetes mellitus are global public health issues. Although males show higher obesity and insulin resistance prevalence, current treatments often neglect sex-specific differences. White adipose tissue (WAT) is crucial in preventing lipotoxicity and inflammation and has become a key therapeutic [...] Read more.
Obesity and type 2 diabetes mellitus are global public health issues. Although males show higher obesity and insulin resistance prevalence, current treatments often neglect sex-specific differences. White adipose tissue (WAT) is crucial in preventing lipotoxicity and inflammation and has become a key therapeutic target. Rosiglitazone (RSG), a potent PPARγ agonist, promotes healthy WAT growth and mitochondrial function through MitoNEET modulation. Recent RSG-based strategies specifically target white adipocytes, avoiding side effects. Our aim was to investigate whether sex-specific differences in the insulin-sensitizing effects of RSG exist on WAT during obesity and inflammation. We used Wistar rats of both sexes fed a high-fat diet (HFD, 22.5% fat content) for 16 weeks. Two weeks before sacrifice, a group of HFD-fed rats received RSG treatment (4 mg/kg of body weight per day) within the diet. HFD male rats showed greater insulin resistance, inflammation, mitochondrial dysfunction, and dyslipidemia than females. RSG had more pronounced effects in males, significantly improving insulin sensitivity, fat storage, mitochondrial function, and lipid handling in WAT while reducing ectopic fat deposition and enhancing adiponectin signaling in the liver. Our study suggests a significant sexual dimorphism in the anti-diabetic effects of RSG on WAT, correlating with the severity of metabolic dysfunction. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

Figure 1
<p>RSG and sex effects on body weight and adiposity. (<b>A</b>) Body weight. (<b>B</b>) White adipose tissue (WAT) weight refers to 100 g of body weight. (<b>C</b>) Adipocyte volume. (<b>D</b>) Adiposity index calculated as the sum of gonadal, retroperitoneal, and mesenteric depot weights relative to 100 g of body weight. Values are presented as the mean ± SEM (5–9 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats. WAT—white adipose tissue.</p>
Full article ">Figure 2
<p>Glucose Tolerance Test. (<b>A</b>) Glucose tolerance curves. (<b>B</b>) AUC—area under the curve. Values are presented as the mean ± SEM (5–7 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Insulin sensitivity and lipid mobilization in WAT. Gene expression analysis of (<b>A</b>) <span class="html-italic">Adipoq</span>, adiponectin; (<b>B</b>) <span class="html-italic">Pparg</span>—peroxisome proliferator-activated receptor gamma; (<b>C</b>) <span class="html-italic">Cisd1</span>—mitoNEET; (<b>D</b>) <span class="html-italic">Plin5</span>—perilipin 5; and (<b>E</b>) <span class="html-italic">Hsl</span>—lipase, hormone-sensitive. (<b>F</b>) pAKT/AKT ratio analyzed by Western blot. AKT serine-threonine kinase activation: insulin-treated animals received an intraperitoneal injection of insulin (5 U/kg) 20 min before sacrifice, while untreated animals (saline group) were injected with 0.9% sodium chloride solution (<span class="html-italic">w</span>/<span class="html-italic">v</span>). Values are presented as the mean ± SEM (4–7 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">Figure 4
<p>Mitochondrial biogenesis and dynamics in WAT. Gene expression analysis of (<b>A</b>) <span class="html-italic">Ppargc1a</span>—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; (<b>B</b>) <span class="html-italic">Ppargc1b</span>—peroxisome proliferator-activated receptor gamma coactivator 1-beta; (<b>C</b>) mtDNA—mitochondrial DNA; (<b>D</b>) <span class="html-italic">Mfn1</span>—mitofusin 1; (<b>E</b>) <span class="html-italic">Mfn2</span>—mitofusin 2; (<b>F</b>) <span class="html-italic">Fis1</span>—fission, mitochondrial 1. Values are presented as the mean ± SEM (4–7 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">Figure 5
<p>Mitochondrial biogenesis and dynamics in 3T3-L1 adipocytes. Adipocytes were treated for 24 h with IL−6 (20 ng/mL) combined with E2 (10 µM), T (10 µM), or RSG (15 µM), respectively. Gene expression analysis was measured of (<b>A</b>) <span class="html-italic">Ppargc1a</span>—peroxisome proliferator-activated receptor gamma coactivator 1-alpha; (<b>B</b>) <span class="html-italic">Ppargc1b</span>—peroxisome proliferator-activated receptor gamma coactivator 1-beta; (<b>C</b>) <span class="html-italic">Cs</span>—citrate synthase; (<b>D</b>) Cox4—cytochrome C oxidase subunit 4I1; (<b>E</b>) Cisd1—mitoNEET; (<b>F</b>) <span class="html-italic">Mfn1</span>—mitofusin 1; (<b>G</b>) <span class="html-italic">Mfn2</span>—mitofusin 2; (<b>H</b>) <span class="html-italic">Fis1</span>—fission, mitochondrial 1; (<b>I</b>) <span class="html-italic">Pparg</span>—peroxisome proliferator-activated receptor gamma; and (<b>J</b>) <span class="html-italic">Adipoq</span>—adiponectin. Values are presented as the mean ± SEM (n = 6). Differences between groups were analyzed by two-way ANOVA to detect a differential response to E2 and RSG (ANOVA1) and to T and RSG (ANOVA2) (<span class="html-italic">p</span> &lt; 0.05), respectively: E—E2 effect, T—testosterone effect, E*R and T*R—interactive effect, and NS—non-significant. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. control; (b) vs. control + RSG; (c) vs. E2; (d) vs. T.</p>
Full article ">Figure 6
<p>Markers of inflammation, hypoxia and apoptosis in WAT. Gene expression analysis of (<b>A</b>) <span class="html-italic">Cd68</span> antigen; (<b>B</b>) <span class="html-italic">Bad</span>/<span class="html-italic">Bcl2</span>—BCL2 associated agonist of cell death/BCL2 apoptosis regulator. (<b>C</b>) <span class="html-italic">Serpine1</span>, PAI-1—plasminogen activator inhibitor-1; (<b>D</b>) <span class="html-italic">Tnf</span>—tumor necrosis factor alpha; and (<b>E</b>) <span class="html-italic">Hif1a</span>—hypoxia inducible factor 1 alpha. Values are presented as the mean ± SEM (4–6 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">Figure 7
<p>Markers of insulin sensitivity in the liver. Western blot analysis of (<b>A</b>) AdipoR2—adiponectin receptor 2; (<b>B</b>) APPL1—adaptor protein, phosphotyrosine interacting with PH domain and leucine zipper 1; (<b>C</b>) pAMPK/AMPK—phospho- and total protein kinase AMP-Activated Catalytic Subunit Alpha 1; (<b>D</b>) pAKT/AKT—pAKT/AKT, phospho- and total AKT serine-threonine kinase; and (<b>E</b>) PEPCK activity—phosphoenolpyruvate carboxykinase. To measure this enzymatic activity, insulin-treated animals received an intraperitoneal injection of insulin (5 U/kg) 20 min before sacrifice, while untreated animals (saline group) were injected with 0.9% sodium chloride solution (<span class="html-italic">w</span>/<span class="html-italic">v</span>). Values are presented as the mean ± SEM (5–7 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">Figure 8
<p>Lipid metabolism in the liver. Gene expression analysis of (<b>A</b>) <span class="html-italic">Cd36</span>—Cd36 antigen. (<b>B</b>) <span class="html-italic">Mttp</span>—microsomal triglyceride transfer protein. (<b>C</b>) <span class="html-italic">Srebp1c</span>—sterol regulatory element-binding protein 1. (<b>D</b>) <span class="html-italic">Ppara</span>—peroxisome proliferator-activated receptor alpha. (<b>E</b>) <span class="html-italic">Fabpl</span>—fatty acid binding protein 1, liver. Values are presented as the mean ± SEM (4–7 animals per group). Differences between sexes within groups and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">Figure 9
<p>RSG and sex effects on hepatic mitochondrial biogenesis. (<b>A</b>) mtDNA—mitochondrial DNA, gene expression analysis of (<b>B</b>) <span class="html-italic">Ppargc1α</span>—peroxisome proliferator-activated receptor gamma coactivator 1-alpha and (<b>C</b>) <span class="html-italic">Ppargc1β</span>—peroxisome proliferator-activated receptor gamma coactivator 1-beta; and Western blot analysis of (<b>D</b>) TFAM protein levels; transcription factor A, mitochondrial; and (<b>E</b>) pJNK/JNK, phospho- and total c-Jun <span class="html-italic">N</span>-terminal kinase. Values are presented as the mean ± SEM (6–7 animals per group). Sex and RSG effects were analyzed by two-way ANOVA (<span class="html-italic">p</span> &lt; 0.05): S—sex effect; R—RSG effect; and S*R—interactive effect. Fisher’s LSD post-hoc test (<span class="html-italic">p</span> &lt; 0.05): (a) vs. HFD rats; (b) vs. male rats.</p>
Full article ">
22 pages, 10840 KiB  
Article
Murine Retina Outer Plexiform Layer Development and Transcriptome Analysis of Pre-Synapses in Photoreceptors
by Soo-Young Kim, Christine Haewon Park, Bo-Hyun Moon and Gail K. Seabold
Life 2024, 14(9), 1103; https://doi.org/10.3390/life14091103 - 2 Sep 2024
Viewed by 486
Abstract
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited [...] Read more.
Photoreceptors in the mammalian retina convert light signals into electrical and molecular signals through phototransduction and transfer the visual inputs to second-order neurons via specialized ribbon synapses. Two kinds of photoreceptors, rods and cones, possess distinct morphology and function. Currently, we have limited knowledge about rod versus (vs.) cone synapse development and the associated genes. The transcription factor neural retina leucine zipper (NRL) determines the rod vs. cone photoreceptor cell fate and is critical for rod differentiation. Nrl knockout mice fail to form rods, generating all cone or S-cone-like (SCL) photoreceptors in the retina, whereas ectopic expression of Nrl using a cone-rod homeobox (Crx) promoter (CrxpNrl) forms all rods. Here, we examined rod and cone pre-synapse development, including axonal elongation, terminal shaping, and synaptic lamination in the outer plexiform layer (OPL) in the presence or absence of Nrl. We show that NRL loss and knockdown result in delayed OPL maturation and plasticity with aberrant dendrites of bipolar neurons. The integrated analyses of the transcriptome in developing rods and SCLs with NRL CUT&RUN and synaptic gene ontology analyses identified G protein subunit beta (Gnb) 1 and p21 (RAC1) activated kinase 5 (Pak5 or Pak7) transcripts were upregulated in developing rods and down-regulated in developing SCLs. Notably, Gnb1 and Gnb5 are rod dominant, and Gnb3 is enriched in cones. NRL binds to the genes of Gnb1, Gnb3, and Gnb5. NRL also regulates pre-synapse ribbon genes, and their expression is altered in rods and SCLs. Our study of histological and gene analyses provides new insights into the morphogenesis of photoreceptor pre-synapse development and regulation of associated genes in the developing retina. Full article
(This article belongs to the Special Issue Retinal Diseases: From Molecular Mechanisms to Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Wild-type outer plexiform layer (OPL) development. (<b>A</b>) Developing OPL stained by anti-Ribeye (synaptic ribbons, green) and anti-Calbindin (horizontal neurons, red). Nuclei stained with DAPI. The boxed areas are shown in insets with higher magnification. OPL (arrowheads) and separate fields of dendrites and axon branches from horizontal neurons (arrows) are shown. (<b>B</b>) Developing OPL stained by anti-Ribeye (green) and anti-PKCα (rod bipolar neurons, red). Clusters of pedicle ribbons (arrows) are shown. (<b>C</b>) Developing rod photoreceptors. Three-dimensional Volocity converted confocal images labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> plasmids. Insert at P6 is shown with higher magnification. (<b>D</b>) Growth cone-like structure at P6 terminals of rod photoreceptors. Confocal images displayed at z-thickness of 0.5 μm from synaptic terminal to axon stalk, labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>. (<b>E</b>) Schematic summary of OPL and photoreceptor synapse development. Abbreviations: ONL, outer nuclear layer; INL, inner nuclear layer; OPL, outer plexiform layer; DAPI, 4′,6-diamidino-2-phenylindole; P, postnatal day; PKCα, protein kinase C alpha. Scale bars, 10 μm in (<b>A</b>,<b>B</b>) and 5 μm in (<b>D</b>).</p>
Full article ">Figure 2
<p>Wild-type spherule and pedicle. (<b>A</b>) P14 vertical retina sections stained by anti-Reep6 (spherules; green) and PNA (pedicles; red) (upper panel) or anti-CAR (M-cone, green) and PNA (S-cone, red) (lower panel). (<b>B</b>) Horizontal OPL images of retina whole mounts stained by anti-CAR and PNA (upper panels). Pure M-cone (arrows) and S-cone (arrowheads) pedicles are observed. The graph shows the distribution (%, average ± SEM) of CAR pedicle areas in OPL of P14, 21, and 28 whole mount retinas. Over 180 CAR positive pedicles were measured from 3 wild-type C57BL/6J retinas. The area distribution after 30 μm<sup>2</sup> is magnified on the left side. (<b>C</b>) Spherules (green) and a pedicle (red) in CD1 retina whole mount labeled by in vivo electroporation of <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> and <span class="html-italic">S-opsinp-tdT</span>. Telodendria (yellow arrows) are observed. (<b>D</b>) Representative images of M/S-, S-cone pedicles and spherules and their area size comparison. Telodendria (yellow arrows) are observed in cones. M-cone and pure S-cone pedicles are segregated by anti-M-opsin staining in the retina whole mounts labeled by <span class="html-italic">S-opsin</span>p-<span class="html-italic">tdT</span> electroporation. M/S-pedicles (n = 54), S-pedicles (n = 14), and Rod spherules (n = 64) from 3 to 5 wild-type CD1 retinas were measured. The graph displays the average ± SD of each: 70.79 ± 21.48 μm<sup>2</sup> for M pedicles, 45.91 ± 18.97 μm<sup>2</sup> for S pedicles, and 2.64 ± 0.81 μm<sup>2</sup> for rod spherules. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. Abbreviations: CAR, cone arrestin; PNA, peanut agglutin lectin; <span class="html-italic">S-opsin</span> promoter-driven <span class="html-italic">tdTomato (S-opsinp-tdT</span>); P, postnatal day; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 10 μm in (<b>A</b>)<b>,</b> and 5 μm in (<b>B</b>–<b>D</b>).</p>
Full article ">Figure 3
<p>Photoreceptor pre-synapse terminals in <span class="html-italic">Nrl</span><sup>-/-</sup> retina. (<b>A</b>) Vertical retina sections of P18 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-Ribeye (red). (<b>B</b>) Horizontal OPL of retina whole mounts in P18 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-Ribeye (red). (<b>C</b>) Horizontal OPL of retina whole mounts in P14 and P28 <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> and <span class="html-italic">Nrl</span><sup>-/-</sup>/<span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span> mice, stained by anti-CAR (red). Arrows indicate pre-synaptic terminals in small size. Abbreviations: <span class="html-italic">Nrl</span>, neural retina leucine zipper; <span class="html-italic">GFP</span>, green fluorescent protein; <span class="html-italic">Nrl</span>p-<span class="html-italic">GFP</span>, <span class="html-italic">Nrl</span> promoter-driven <span class="html-italic">GFP</span>; P, postnatal day; CAR, cone arrestin. Scale bars, 1 μm in magnified box of (<b>A</b>), 5 μm in (<b>A</b>,<b>C</b>), and 2.5 μm in (<b>B</b>).</p>
Full article ">Figure 4
<p>Pre-synapse comparison in wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> or <span class="html-italic">Nrl</span> knockdown S-cone-like (SCL) photoreceptors. (<b>A</b>) Representative Volocity 3D images of wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL photoreceptors, taken from wild-type or <span class="html-italic">Nrl</span><sup>-/-</sup> retina whole mounts labeled by <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>, <span class="html-italic">S-opsinp-tdT</span>. Rods (green only), cones (red only) and SCLs (mixed green and red) were imaged. M/S- and pure S-cones were differentiated by staining with an anti-M-opsin antibody. (<b>B</b>) Representative confocal images of pre-synapse terminals of wild-type rod, M/S-cone, pure S-cone, and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL photoreceptors. (<b>C</b>) Size distribution of pre-synapses in wild-type rod (n = 64), M/S-cone (n = 54), pure S-cone (n = 14) and <span class="html-italic">Nrl</span><sup>-/-</sup> SCL (P14, n = 25; P21, n = 38) photoreceptors. (<b>D</b>) Representative Volocity 3D images of P28 retina whole mounts labeled by electroporation of scrambled or <span class="html-italic">Nrl</span> shRNA plasmid (sh<span class="html-italic">Nrl</span>) with <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span> (2:1 ratio). (<b>E</b>) Representative confocal images of pre-synaptic terminals expressing scrambled or <span class="html-italic">Nrl</span> shRNA. (<b>F</b>) Size distribution of pre-synapses in control (P14, n = 248; P21, n = 64; P28, n = 98) and developing <span class="html-italic">Nrl</span> shRNA SCL photoreceptors (P14, n = 246; P21, n = 31; P28, n = 124; P35, n = 36) labeled with <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>. Data of area in measurement were analyzed by one-way ANOVA (Tukey or Kriskal–Wallis test) and T-test (two-tailed) in Prism. *, statistically meaningful in one-way ANOVA and T-test; *t, statistically meaningful in T-test. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; 3D, three-dimensional; <span class="html-italic">Nrl</span>p-<span class="html-italic">EGFP</span>, <span class="html-italic">Nrl</span> promoter-driven enhanced <span class="html-italic">GFP</span>; <span class="html-italic">S-opsin</span> promoter-driven <span class="html-italic">tdTomato</span> (<span class="html-italic">S-opsinp-tdT</span>); P, postnatal day; SCL, S-cone-like; shRNA, short hairpin ribonucleic acid. Scale bars, 5 μm in (<b>B</b>,<b>E</b>).</p>
Full article ">Figure 5
<p>Outer plexiform layer development and synaptic connection in <span class="html-italic">Nrl</span><sup>-/-</sup> retina. (<b>A</b>) Developing (P10 to P17) retinas of wild-type and <span class="html-italic">Nrl</span><sup>-/-</sup> mice stained by anti-Ribeye (green) and anti-PKCα (red). Clustered pedicle ribbons (white dotted lines) and dendritic tips of rod bipolar neurons without synaptic ribbons (yellow arrows) are observed. (<b>B</b>) Comparison of OPL thickness in developing wild-type and <span class="html-italic">Nrl</span><sup>-/-</sup> retinas. Measurement was quantified on five images of the middle retina (with optic nerve head) from each of three to four animals in different developing stages. Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. (<b>C</b>) Comparison of the ribbon distribution in OPL. Distance of ribbon location from the ONL bottom when the OPL thickness is considered 1.0. The location of individual ribbons was measured with each OPL thickness in over two images from each of three to four animals. Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. (<b>D</b>) Number comparison (%) of rod bipolar neuron dendritic tips with or without ribbons aligned at their tops. Dendritic tips of rod bipolar neurons were measured at P10 (<span class="html-italic">WT</span>, n = 363; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 627), P12 (<span class="html-italic">WT</span>, n = 445; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 953), P14 (<span class="html-italic">WT</span>, n = 433; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 691), and P17 (<span class="html-italic">WT</span>, n = 197; <span class="html-italic">Nrl</span><sup>-/-</sup>, n = 269). Values represent mean ± SD. * <span class="html-italic">p</span> ≤ 0.05, two-tailed T-test. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; P, postnatal day; PKCα, Protein Kinase C alpha; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 5 μm in (<b>A</b>).</p>
Full article ">Figure 6
<p>Synaptic plasticity in <span class="html-italic">Nrl</span> transgenic retinas. (<b>A</b>) 1.5-month retinas of wild-type, <span class="html-italic">Nrl</span><sup>-/-</sup> (cone-only) and <span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span> (rod-only) mice stained by anti-PKCα (rod bipolar neurons, green) and anti-Goα (all ON bipolar neurons, red). (<b>B</b>) Retinas from 1.5-month-old <span class="html-italic">Clm-GFP</span> (type 9 cone bipolar neurons, green), <span class="html-italic">Clm-GFP</span>/<span class="html-italic">Nrl</span><sup>-/-</sup>, and <span class="html-italic">Clm-GFP</span>/<span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span> mice stained with anti-Ribeye (red). Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; <span class="html-italic">Crx</span>p<span class="html-italic">Nrl</span>, <span class="html-italic">Cone-rod homeobox</span> promoter-driven <span class="html-italic">Nrl</span>; Clm, clomeleon; GFP, green fluorescent protein; PKCα, Protein Kinase C alpha; Goα, guanine nucleotide-binding protein alpha subunit; ONL, outer nuclear layer; OPL, outer plexiform layer; INL, inner nuclear layer. Scale bars, 10 μm in (<b>A</b>,<b>B</b>).</p>
Full article ">Figure 7
<p>RNA-seq and CUT&amp;RUN-seq analyses of photoreceptor pre-synapse genes. (<b>A</b>) Schematic drawing of NRL gene regulation in rod photoreceptors. NRL activates rod genes and suppresses cone genes. (<b>B</b>,<b>C</b>) Venn diagrams displaying differently expressed genes in rods, SCLs, and NRL-binding genes. (<b>D</b>–<b>F</b>) SynGO visualizations of upRod-NRL, upRod-downSCL, and upSCL-NRL. (<b>G</b>) SynGO synapse genes, in order from P2 to P28 in transcript per million (TPM) heatmaps: SynGO upRod-NRL (left), SynGO upRod-downSCL (upper right), SynGO upSCL-NRL (down right). Genes in which transcripts are upregulated in developing rods and down-regulated in developing SCLs up to P28 are indicated with a green asterisk, and genes in which transcripts are down-regulated in rods and upregulated in developing SCLs are indicated with a purple asterisk. Known ribbon-associated genes are indicated with a yellow dot. (<b>H</b>) <span class="html-italic">Gnb1</span>, <span class="html-italic">Gnb3</span>, and <span class="html-italic">Gnb5</span> expression in developing rods and SCLs measured by RNAseq dataset (TPM). The line plots (average ± SD) from all relevant transcripts in 2 to 4 bio-replicates were generated using ggplot2 from R studio. (<b>I</b>) Ribbon synapse genes, in order from P2 to P29 in the TPM heatmap. Gene names in magenta correspond to genes showing higher expression in SCLs than rods at P28. Green gene names correspond to genes showing higher expression in rods than SCLs at P28. Gene names in bold font indicate that NRL binds to the genes. Abbreviations: WT, wild-type; <span class="html-italic">Nrl</span>, neural retina leucine zipper; upRod-NRL, upregulated NRL binding genes in developing WT rods; upSCL-NRL, upregulated NRL binding genes in developing S-cone-like photoreceptors; downSCL, down-regulated genes in developing S-cone-like photoreceptors; adj. <span class="html-italic">p</span>, adjusted <span class="html-italic">p</span>-value; TPM, transcript per million; SynGO, synaptic Gene Ontologies and annotations.</p>
Full article ">
Case Report
The First Patient with Tibial Hemimelia-Polysyndactyly-Triphalangeal Thumb Syndrome Caused by De Novo c.423+4916 T>C ZRS Variant: A Case Report
by Paola Montserrat Zepeda-Olmos, Kiabeth Robles-Espinoza, Eduardo Esparza-García and María Teresa Magaña-Torres
Int. J. Mol. Sci. 2024, 25(17), 9348; https://doi.org/10.3390/ijms25179348 - 29 Aug 2024
Viewed by 422
Abstract
Genetic variants in the zone of polarizing activity regulatory sequence (ZRS) that induce ectopic expression of the SHH gene have been associated with different ZRS-related phenotypes. We report the first patient with a de novo variant, c.423+4916 T>C, in ZRS (previously classified as [...] Read more.
Genetic variants in the zone of polarizing activity regulatory sequence (ZRS) that induce ectopic expression of the SHH gene have been associated with different ZRS-related phenotypes. We report the first patient with a de novo variant, c.423+4916 T>C, in ZRS (previously classified as a variant of uncertain significance) that causes tibial hemimelia-polysyndactyly-triphalangeal thumb syndrome (THPTTS). A two-month-old male patient presented with bilateral preaxial polydactyly, triphalangeal thumb, and tibial agenesis and was heterozygous for the variant c.423+4916T>C (neither of his parents was a carrier). The findings obtained from the family study were sufficient to reclassify the variant from “uncertain significance” to “likely pathogenic” according to three criteria from the American College of Medical Genetics and Genomics guidelines, as follows: (1) absence of gnomAD, (2) confirmation of paternity and maternity, and (3) strong phenotype–genotype association. In ZRS-associated syndromes, a wide clinical spectrum has been observed, ranging from polydactyly to THPTTS; our patient has the most severe and rare phenotype. We did not perform functional assays. However, the c.423+4916T>C variant is flanked by three variants, which have been proven not only to cause the phenotype but also to increase the expression of SHH. Through all this data gathering, we consider the c.423+4916T>C variant to be causative of THPTTS. Full article
(This article belongs to the Special Issue Genes and Human Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>Lower limbs. Photograph (<b>A</b>) and X-ray bone radiographic imaging (<b>B</b>) showing bilateral tibial agenesis of our patient. Hand X-rays (<b>C</b>) showing bilateral triphalangeal thumb. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>(<b>A</b>) Index case electropherogram. The arrow shows the site of variant c.423+4916T&gt;C. (<b>B</b>,<b>C</b>) show electropherograms of the mother and father. The arrow demonstrates the absence of variation. (<b>D</b>) ZRS, region comprising from c.423+4 to c.423+29. The arrowhead shows the c.423+4916T&gt;C variant, surrounded by previously reported pathogenic variants associated with the THPTTS phenotype (arrows). Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">
25 pages, 1287 KiB  
Review
The Impact of Adenomyosis on Pregnancy
by Panagiotis Tsikouras, Nektaria Kritsotaki, Konstantinos Nikolettos, Sonia Kotanidou, Efthymios Oikonomou, Anastasia Bothou, Sotiris Andreou, Theopi Nalmpanti, Kyriaki Chalkia, Vlasios Spanakis, Nikolaos Tsikouras, Melda Chalil, Nikolaos Machairiotis, George Iatrakis and Nikolaos Nikolettos
Biomedicines 2024, 12(8), 1925; https://doi.org/10.3390/biomedicines12081925 - 22 Aug 2024
Viewed by 572
Abstract
Adenomyosis is characterized by ectopic proliferation of endometrial tissue within the myometrium. Histologically, this condition is marked by the presence of islands of benign endometrial glands surrounded by stromal cells. The myometrium appears thinner, and cross-sectional analysis often reveals signs of recent or [...] Read more.
Adenomyosis is characterized by ectopic proliferation of endometrial tissue within the myometrium. Histologically, this condition is marked by the presence of islands of benign endometrial glands surrounded by stromal cells. The myometrium appears thinner, and cross-sectional analysis often reveals signs of recent or chronic hemorrhage. The ectopic endometrial tissue may respond to ovarian hormonal stimulation, exhibiting proliferative or secretory changes during the menstrual cycle, potentially leading to bleeding, uterine swelling, and pain. Adenomyosis can appear as either a diffuse or focal condition. It is crucial to understand that adenomyosis involves the infiltration of the endometrium into the myometrium, rather than its displacement. The surgical management of adenomyosis is contingent upon its anatomical extent. The high incidence of the disease and the myths that develop around it increase the need to study its characteristics and its association with pregnancy and potential obstetric complications. These complications often require quick decisions, appropriate diagnosis, and proper counseling. Therefore, knowing the possible risks associated with adenomyosis is key to decision making. Pregnancy has a positive effect on adenomyosis and its painful symptoms. This improvement is not only due to the inhibition of ovulation, which inhibits the bleeding of adenomyotic tissue, but also to the metabolic, hormonal, immunological, and angiogenic changes associated with pregnancy. Adenomyosis affects pregnancy through disturbances of the endocrine system and the body’s immune response at both local and systemic levels. It leads to bleeding from the adenomyotic tissue, molecular and functional abnormalities of the ectopic endometrium, abnormal placentation, and destruction of the adenomyotic tissue due to changes in the hormonal environment that characterizes pregnancy. Some of the obstetric complications that occur in women with adenomyosis in pregnancy include miscarriage, preterm delivery, placenta previa, low birth weight for gestational age, obstetric hemorrhage, and the need for cesarean section. These complications are an understudied field and remain unknown to the majority of obstetricians. These pathological conditions pose challenges to both the typical progression of pregnancy and the smooth conduct of labor in affected women. Further multicenter studies are imperative to validate the most suitable method for concluding labor following surgical intervention for adenomyosis. Full article
(This article belongs to the Special Issue Advanced Research in Endometriosis 4.0)
Show Figures

Figure 1

Figure 1
<p>KRAS signaling pathways. KRAS pathway is activated by a receptor tyrosine kinase and regulates the intracellular pathways of mTOR, ERK and NF-kB. GAP: GTPase activating proteins; GEF: guanine-nucleotide exchange factors.</p>
Full article ">Figure 2
<p>Theories explaining the origin of adenomyosis.</p>
Full article ">Figure 3
<p>Mechanisms that redound adenomyosis in negative pregnancy outcomes.</p>
Full article ">
25 pages, 4496 KiB  
Article
FaNPR3 Members of the NPR1-like Gene Family Negatively Modulate Strawberry Fruit Resistance against Colletotrichum acutatum
by Victoria Súnico, José Javier Higuera, Francisco Amil-Ruiz, Isabel Arjona-Girona, Carlos J. López-Herrera, Juan Muñoz-Blanco, Ana María Maldonado-Alconada and José L. Caballero
Plants 2024, 13(16), 2261; https://doi.org/10.3390/plants13162261 - 14 Aug 2024
Viewed by 495
Abstract
Strawberry fruit is highly appreciated worldwide for its organoleptic and healthy properties. However, this plant is attacked by many pathogenic fungi, which significantly affect fruit production and quality at pre- and post-harvest stages, making chemical applications the most effective but undesirable strategy to [...] Read more.
Strawberry fruit is highly appreciated worldwide for its organoleptic and healthy properties. However, this plant is attacked by many pathogenic fungi, which significantly affect fruit production and quality at pre- and post-harvest stages, making chemical applications the most effective but undesirable strategy to control diseases that has been found so far. Alternatively, genetic manipulation, employing plant key genes involved in defense, such as members of the NPR-like gene family, has been successful in many crops to improve resistance. The identification and use of the endogenous counterpart genes in the plant of interest (as it is the case of strawberry) is desirable as it would increase the favorable outcome and requires prior knowledge of their defense-related function. Using RNAi technology in strawberry, transient silencing of Fragaria ananassa NPR3 members in fruit significantly reduced tissue damage after Colletotrichum acutatum infection, whereas the ectopic expression of either FaNPR3.1 or FaNPR3.2 did not have an apparent effect. Furthermore, the ectopic expression of FaNPR3.2 in Arabidopsis thaliana double-mutant npr3npr4 reverted the disease resistance phenotype to Pseudomonas syringe to wild-type levels. Therefore, the results revealed that members of the strawberry FaNPR3 clade negatively regulate the defense response to pathogens, as do their Arabidopsis AtNPR3/AtNPR4 orthologs. Also, evidence was found showing that FaNPR3 members act in strawberry (F. ananassa) as positive regulators of WRKY genes, FaWRKY19 and FaWRKY24; additionally, in Arabidopsis, FaNPR3.2 negatively regulates its orthologous genes AtNPR3/AtNPR4. We report for the first time the functional characterization of FaNPR3 members in F. ananassa, which provides a relevant molecular basis for the improvement of resistance in this species through new breeding technologies. Full article
(This article belongs to the Section Plant Genetics, Genomics and Biotechnology)
Show Figures

Figure 1

Figure 1
<p>Expression pattern of <span class="html-italic">FaNPR3</span> genes in strawberry fruit after agroinfiltration with pFRN::FaNPR3<span class="html-italic">all</span>.RNAi construct. (<b>A</b>) Gene expression of <span class="html-italic">FaNPR3.1</span>. (<b>B</b>) Gene expression of <span class="html-italic">FaNPR3.2</span>. (<b>C</b>) Gene expression of <span class="html-italic">FaNPR3.3</span>. qRT-PCR analysis was accomplished in agroinfiltrated strawberry fruit before (0 and 2 days) and after (3, 5, and 7 days) <span class="html-italic">C. acutatum</span> inoculation (<span class="html-italic">Ca</span>). The time scale refers to days after agroinfiltration (dai). For all the genes, the arrow and horizontal line indicate the time of <span class="html-italic">Ca</span> inoculation. The black and grey columns show pFRN (control) and pFRN::FaNPR3<span class="html-italic">all</span>.RNAi (silencing) agroinfiltrations, respectively. Data from all time points are referred to data in time zero, represented as 1 (white column). Bars, mean ± standard error. Note the different scales in the relative-expression-level axis. Statistical significance was determined by one-way ANOVA. Letters indicate significant differences (<span class="html-italic">p</span>  &lt;  0.05) in HSD Tukey’s post hoc test.</p>
Full article ">Figure 2
<p>Expression pattern of <span class="html-italic">FaNPR3</span> genes in strawberry fruit after agroinfiltration with pB7GWIWG2::FaNPR32.RNAi. (<b>A</b>) Gene expression of <span class="html-italic">FaNPR3.1</span>. (<b>B</b>) Gene expression of <span class="html-italic">FaNPR3.2</span>. (<b>C</b>) Gene expression of <span class="html-italic">FaNPR3.3</span>. qRT-PCR analysis was accomplished in agroinfiltrated strawberry fruit before (0 and 2 days) and after (3, 5, and 7 days) <span class="html-italic">C. acutatum</span> inoculation (<span class="html-italic">Ca</span>). The time scale refers to days after agroinfiltration (dai). For all the genes, the arrow and the horizontal line indicate the time of <span class="html-italic">Ca</span> inoculation. The black and grey columns show pB7GWIWG2 and pB7GWIWG2::FaNPR32.RNAi agroinfiltrations, respectively. Data from all time points are referred to data in time zero, represented as 1 (white column). Bars, mean ± standard error. Note the different scales in the relative-expression-level axis. Statistical significance was determined by one-way ANOVA. Letters indicate significant differences (<span class="html-italic">p</span>  &lt;  0.05) in HSD Tukey’s post hoc test.</p>
Full article ">Figure 3
<p>Silencing effect of pFRN::FaNPR3<span class="html-italic">all</span>.RNAi and ectopic overexpression of <span class="html-italic">FaNPR3.1</span> in strawberry fruit after <span class="html-italic">C. acutatum</span> infection. (<b>A</b>,<b>B</b>) Upper panels: external surface disease symptoms on the two agroinfiltrated opposite halves of the same fruit, after silencing and overexpression, respectively. (<b>A</b>,<b>B</b>) Lower panels: internal tissue damage of the same fruit shown in the corresponding upper panels. pFRN::FaNPR3<span class="html-italic">all</span>.RNAi and pFRN, silencing construct and its corresponding empty vector, as control. pK7::FaNPR31.OE and pK7, overexpression construct and its corresponding empty vector, as control. A relevant fruit is shown for each condition, as an example. (<b>C</b>) Statistical analysis of internal tissue damage ratio of the two opposite halves of the same fruit, according to the 1 to 5 severity scale; striped and plain blue bars, pFRN/pFRN and pFRN/pFRN::FaNPR31-RNAi agroinfiltrated values, respectively; striped and plain red bars, pK7/pK7 and pK7/pK7::FaNPR31-OE agroinfiltrated values, respectively. Data correspond to mean ± SD. Within each bar, means with different letters are significantly different by LSD test at <span class="html-italic">p</span> &lt; 0.05. A ratio value of 1 indicates no differences between the opposite halves of the same fruit.</p>
Full article ">Figure 4
<p>Silencing effect of pB7GWIWG2::FaNPR32.RNAi and ectopic overexpression of <span class="html-italic">FaNPR3.2</span> in strawberry fruit after <span class="html-italic">C. acutatum</span> infection. (<b>A</b>,<b>B</b>) Upper panels: external surface disease symptoms on the two agroinfiltrated opposite halves of the same fruit, after silencing and overexpression, respectively. (<b>A</b>,<b>B</b>) Lower panels: internal tissue damage of the same fruit shown in the corresponding upper panels. pB7GWIWG2::FaNPR32.RNAi and pB7GWIWG2, silencing construct and its corresponding empty vector, as control. pB7WG2::FaNPR32.OE and pB7WG2, overexpression construct and its corresponding empty vector, as control. A relevant fruit is shown for each condition, as an example. (<b>C</b>) Statistical analysis of internal tissue damage ratio of the two opposite halves of the same fruit, according to the 1 to 5 severity scale; striped and plain blue bars, pB7GWIWG2/pB7GWIWG2 and pB7GWIWG2/pB7GWIWG2::FaNPR32-RNAi agroinfiltrated values, respectively; striped and plain red bars, pB7WG2/pB7WG2 and pB7WG2/pB7WG2::FaNPR32-OE agroinfiltrated values, respectively. Data correspond to mean ± SD. Within each bar, means with different letters are significantly different by LSD test at <span class="html-italic">p</span> &lt; 0.05. A ratio value of 1 indicates no differences between opposite halves of the same fruit.</p>
Full article ">Figure 5
<p>Silencing effect of <span class="html-italic">FaNPR3</span> members on the expression of <span class="html-italic">FaWRKY19</span> and <span class="html-italic">FaWRKY24</span> genes in strawberry fruit after <span class="html-italic">C. acutatum</span> infection. (<b>A</b>) Silencing effect of pFRN::FaNPR3<span class="html-italic">all</span>.RNAi. (<b>B</b>) Silencing effect pB7GWIWG2::FaNPR32.RNAi. Black columns, the expression of <span class="html-italic">FaWRKY19</span> and <span class="html-italic">FaWRKY24</span> genes in half fruit agroinfiltrated with pFRN (<b>A</b>) or pB7GWIWG2 (<b>B</b>) control vectors. Grey columns, the expression of <span class="html-italic">FaWRKY19</span> and <span class="html-italic">FaWRKY24</span> genes in half fruit agroinfiltrated with pFRN::FaNPR3<span class="html-italic">all</span>.RNAi (<b>A</b>) or pB7GWIWG2::FaNPR32.RNAi (<b>B</b>). qRT-PCR analysis was accomplished in agroinfiltrated strawberry fruit before (0 and 2 days) and after (3, 5, and 7 days) <span class="html-italic">C. acutatum</span> inoculation (<span class="html-italic">Ca</span>). The time scale refers to days after agroinfiltration (dai). Arrow and horizontal line indicate the time of <span class="html-italic">Ca</span> inoculation. Data from all time points are referred to data in time zero, represented as 1 (white column). Bars, mean ± standard error. Note the different scales in the relative-expression-level axis. Statistical significance was determined by one-way ANOVA. Letters indicate significant differences (<span class="html-italic">p</span>  &lt;  0.05) in HSD Tukey’s post hoc test.</p>
Full article ">Figure 6
<p>Characterization of Arabidopsis lines. (<b>A</b>) <span class="html-italic">FaNPR3.2</span> expression in Arabidopsis WT-, <span class="html-italic">npr3npr4</span> mutant-, and <span class="html-italic">FaNPR3.2</span>-overexpressing lines <span class="html-italic">npr3npr4</span>::FaNPR32 and WT::FaNPR32 (black, grey, white and striped bars, respectively). Transcript accumulation was monitored by qRT-PCR in non-infected plants as described in the <a href="#sec4-plants-13-02261" class="html-sec">Section 4</a>. Expression levels were normalized with respect to the internal control <span class="html-italic">ACTIN2</span> and displayed relatively to the threshold value of the wild-type (no expression of <span class="html-italic">FaNPR3.2</span>) that was given a value of 1 for convenience. Note that no amplification of <span class="html-italic">FaNPR3.2</span> strawberry gene is detected in neither WT nor double mutant <span class="html-italic">npr3npr4</span> using the specific primers (<a href="#app1-plants-13-02261" class="html-app">Supplementary Materials Table S1</a>). Bars represent the mean levels of transcript quantified from three independent biological experiments (±SD). (<b>B</b>–<b>D</b>) Disease resistance phenotype of Arabidopsis lines upon <span class="html-italic">Pst</span> inoculation. (<b>B</b>) Symptoms development on leaves 3, 5, and 7 days post-inoculation (dpi) (10<sup>6</sup> CFU mL<sup>−1</sup>); (<b>C</b>) trypan blue staining for the detection of cell death 1 dpi (10<sup>5</sup> CFU mL<sup>−1</sup>); (<b>D</b>) in planta bacterial growth monitored 3, 5, and 7 days post-inoculation (10<sup>6</sup> CFU mL<sup>−1</sup>). CFU, colony-forming units. Statistically significant differences are labelled by letters (one-way ANOVA, Tukey’s multiple comparisons test, <span class="html-italic">p</span> &lt; 0.05). <span class="html-italic">Pst</span> was pressure infiltrated into fully expanded mature leaves of 4–5-week-old Arabidopsis plants. As control, leaves were infiltrated with 10 mM MgCl<sub>2</sub>. The whole experiment was performed three times with similar results.</p>
Full article ">Figure 7
<p>Expression of the defense-related genes <span class="html-italic">PR1</span>, <span class="html-italic">PR2</span>, and <span class="html-italic">PR5</span> in Arabidopsis lines. Relative expression level was monitored by qRT-PCR in control (<b>A</b>) and infected plants 1 and 2 days post-inoculation (dpi) with <span class="html-italic">Pst</span> (10<sup>6</sup> CFU mL<sup>−1</sup>) (<b>B</b>), in WT, WT::FaNPR32, double mutant <span class="html-italic">npr3npr4</span>, and <span class="html-italic">npr3npr4</span>::FaNPR32 (black, grey, white and striped bars, respectively). Expression levels were normalized with respect to the internal control <span class="html-italic">ACTIN2</span> and displayed relative to the expression in mock-treated wild-type samples (<b>A</b>) or to the expression in mock-treated samples of each line (<b>B</b>) that were given a value of 1. Bars refer to mean ± standard error. Note the different scales in the relative-expression-level axis. Statistical significance was determined by one-way ANOVA. Letters indicate significant differences (<span class="html-italic">p</span>  &lt;  0.05) in HSD Tukey’s post hoc test.</p>
Full article ">Figure 8
<p>Expression of the endogenous <span class="html-italic">AtNPR1</span>, <span class="html-italic">AtNPR3</span>, and <span class="html-italic">AtNPR4</span> genes in Arabidopsis. Relative expression level was monitored by qRT-PCR in control (<b>A</b>) and infected plants 1 and 2 days post-inoculation (dpi) with <span class="html-italic">Pst</span> (10<sup>6</sup> CFU mL<sup>−1</sup>) (<b>B</b>), in WT, WT::FaNPR32, double mutant <span class="html-italic">npr3npr4</span>, and <span class="html-italic">npr3npr4</span>::FaNPR32 (black, grey, white and striped bars, respectively). Expression levels were normalized with respect to the internal control <span class="html-italic">ACTIN2</span> and displayed relative to the expression in mock-treated wild-type samples (<b>A</b>) or to the expression in mock-treated samples of each line (<b>B</b>) that were given a value of 1. Bars refer to mean ± standard error. Note the different scales in the relative-expression-level axis. Statistical significance was determined by one-way ANOVA. Letters indicate significant differences (<span class="html-italic">p</span>  &lt;  0.05) in HSD Tukey’s post hoc test. Note that the absence of transcript for <span class="html-italic">AtNPR3</span> and <span class="html-italic">AtNPR4</span> in the overexpressing lines in the double mutant <span class="html-italic">npr3npr4</span> background. It proves that Arabidopsis primers do not amplify strawberry orthologous genes.</p>
Full article ">
11 pages, 1435 KiB  
Article
Ectopic PU.1 Expression Provides Chimeric Antigen Receptor (CAR) T Cells with Innate Cell Capacities Including IFN-β Release
by Dennis Christoph Harrer, Matthias Eder, Markus Barden, Hong Pan, Wolfgang Herr and Hinrich Abken
Cancers 2024, 16(15), 2737; https://doi.org/10.3390/cancers16152737 - 1 Aug 2024
Viewed by 688
Abstract
Chimeric antigen receptor (CAR) T cell therapy has achieved extraordinary success in eliminating B cell malignancies; however, so far, it has shown limited efficacy in the treatment of solid tumors, which is thought to be due to insufficient CAR T cell activation. We [...] Read more.
Chimeric antigen receptor (CAR) T cell therapy has achieved extraordinary success in eliminating B cell malignancies; however, so far, it has shown limited efficacy in the treatment of solid tumors, which is thought to be due to insufficient CAR T cell activation. We hypothesized that the transcription factor PU.1, a master regulator of innate cell functionality, may augment pro-inflammatory CAR T cell activation. T cells were engineered with a CEA-specific CAR together with the constitutive expression of PU.1. CAR-redirected T cell activation was recorded for canonical functionality in vitro under conditions of prolonged repetitive antigen exposure. Ectopic PU.1 expression in CAR T cells upregulated the costimulatory receptors CD40, CD80, CD86, and CD70, which, unexpectedly, did not augment effector functions but hampered the upregulation of 4-1BB, decreased IL-2 production, reduced CAR T cell proliferation, and impaired their cytotoxic capacities. Under “stress” conditions of repetitive engagement of cognate tumor cells, CAR T cells with ectopic PU.1 showed reduced persistence, and finally failed to control the growth of cancer cells. Mechanistically, PU.1 caused CAR T cells to secrete IFN-β, a cytokine known to promote CAR T cell attrition and apoptosis. Collectively, PU.1 can polarize the functional capacities of CAR T cells towards innate cells. Full article
(This article belongs to the Special Issue CAR T Therapy of Solid Cancers: Problems and Progress)
Show Figures

Figure 1

Figure 1
<p>Ectopic expression of PU.1 upregulates costimulatory receptors on CAR T cells. (<b>A</b>) Schematic depiction of CAR and PU.1 expression constructs. (<b>B</b>) CAR expression by aCEA-28ζ-GFP CAR T cells (Ctrl) and aCEA-28ζ-PU.1 CAR T cells (PU.1) was detected by staining with a phycoerythrin (PE)-labeled goat anti-IgG antibody before (upper panels) and after (lower panels) magnetic cell separation (MACS). The anti-IgG antibody detects the common IgG1 CH2-CH3 spacer domain of the CAR. One representative donor out of six is shown. Data represent means ± SEM of six donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test, ns: not significant. (<b>C</b>) Western blot showing PU.1 protein expression in aCEA-28ζ-GFP CAR T cells (Ctrl) and aCEA-28ζ-PU.1 CAR T cells (PU.1) five days after retroviral transduction. One representative donor out of six donors is shown. (<b>D</b>) Flow cytometric analysis of PU.1 target genes MHC-I and MHC-II, costimulatory receptors, adhesion molecules, and the myeloid marker CD33 in CD8<sup>+</sup> (upper panels) and CD4<sup>+</sup> (lower panels) CAR T cells at baseline (d0) and 48 h after stimulation with CEA+ BxPC-3 pancreatic carcinoma cells (d2). Data represent geometric means ± SEM of four donors; <span class="html-italic">p</span> values were calculated by paired <span class="html-italic">t</span> test. * indicates <span class="html-italic">p</span> ≤ 0.05, ** indicates <span class="html-italic">p</span> ≤ 0.01, *** indicates <span class="html-italic">p</span> ≤ 0.001 and ns indicates not significant.</p>
Full article ">Figure 2
<p>Ectopic expression of PU.1 impairs IL-2 secretion and proliferation of CAR T cells. (<b>A</b>) CAR-triggered CD25 upregulation in CD8<sup>+</sup> and CD4<sup>+</sup> T cells 48 h after co-incubation with CEA<sup>+</sup> BxPC-3 cells compared with CEA<sup>−</sup> 293 T cells. (<b>B</b>) CAR-triggered secretion of IFN-γ and IL-2 by CAR T cells incubated with BxPC-3 cells and 293T cells, respectively, after 48 h as determined by ELISA. (<b>C</b>) Expansion index of CD8<sup>+</sup> and CD4<sup>+</sup> CAR T cells was determined by staining with the “Cell Proliferation Dye eFluor<sup>®</sup> 450” and co-incubation with BxPC-3 cells for five days. (<b>A</b>–<b>C</b>) Data represent means ± SEM of four donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05; *** <span class="html-italic">p</span> ≤ 0.001; ns, not significant.</p>
Full article ">Figure 3
<p>Ectopic PU.1 expression diminishes cytotoxicity of CAR T cells. (<b>A</b>) CAR-triggered CD137 upregulation in CD8<sup>+</sup> and CD4<sup>+</sup> T cells 48 h after co-incubation with BxPC-3 cells and 293 T cells. (<b>B</b>) Cytotoxicity of CAR T cells after a 24-h co-culture with CEA<sup>+</sup> BxPC-3 cells (left panel) and CEA- 293T cells (right panel) was determined by co-incubation at the indicated effector to target ratios and cytotoxicity recorded by an XTT-based colorimetric assay. (<b>C</b>) Flow cytometric analysis of granzyme B and perforin levels in CD8<sup>+</sup> CAR T cells at baseline after retroviral transduction. (<b>A</b>–<b>C</b>) Data represent means ± SEM of four donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01; **** <span class="html-italic">p</span> ≤ 0.0001; ns, not significant.</p>
Full article ">Figure 4
<p>Ectopic expression of PU.1 reduces functional persistence of CAR T cells. (<b>A</b>) CAR T cells (1 × 10<sup>5</sup> CAR T cells) were stimulated twice (R1 and R2) with GFP-labeled CEA<sup>+</sup> BxPC-3 cells (1 × 10<sup>5</sup> tumor cells). At the end of each round, live CD3<sup>+</sup> CAR<sup>+</sup> T cells (left panel) and BxPC-3 cells (right panel) were counted by flow cytometry. Data represent means ± SEM of five donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05, ** <span class="html-italic">p</span> ≤ 0.01; **** <span class="html-italic">p</span> ≤ 0.0001; ns, not significant. (<b>B</b>) CD8<sup>+</sup> and CD4<sup>+</sup> CAR T cells with PU.1 expression were co-incubated with BxPC-3 cells (0.5 × 10<sup>5</sup> tumor cells) for 72 h and Annexin V staining recorded by flow cytometry. Data represent means ± SEM of four donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05. (<b>C</b>) CD8<sup>+</sup> and CD4<sup>+</sup> CAR T cells with and without PU.1 were co-incubated with BxPC-3 cells for 72 h and Ki-67 expression determined by flow cytometry. Data represent means ± SEM of three donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05; ** <span class="html-italic">p</span> ≤ 0.01. (<b>D</b>) IFNβ released into the supernatant by CAR T cells incubated with BxPC-3 cells after 48 h as determined by ELISA. Data represent means ± SEM of three donors; <span class="html-italic">p</span> values were calculated by Student’s <span class="html-italic">t</span> test; * <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">
16 pages, 3219 KiB  
Article
Vitamin D Receptor Regulates the Expression of the Grainyhead-Like 1 Gene
by Agnieszka Taracha-Wisniewska, Emma G. C. Parks, Michal Miller, Lidia Lipinska-Zubrycka, Sebastian Dworkin and Tomasz Wilanowski
Int. J. Mol. Sci. 2024, 25(14), 7913; https://doi.org/10.3390/ijms25147913 - 19 Jul 2024
Viewed by 562
Abstract
Vitamin D plays an important pleiotropic role in maintaining global homeostasis of the human body. Its functions go far beyond skeletal health, playing a crucial role in a plethora of cellular functions, as well as in extraskeletal health, ensuring the proper functioning of [...] Read more.
Vitamin D plays an important pleiotropic role in maintaining global homeostasis of the human body. Its functions go far beyond skeletal health, playing a crucial role in a plethora of cellular functions, as well as in extraskeletal health, ensuring the proper functioning of multiple human organs, including the skin. Genes from the Grainyhead-like (GRHL) family code for transcription factors necessary for the development and maintenance of various epithelia. Even though they are involved in many processes regulated by vitamin D, a direct link between vitamin D-mediated cellular pathways and GRHL genes has never been described. We employed various bioinformatic methods, quantitative real-time PCR, chromatin immunoprecipitation, reporter gene assays, and calcitriol treatments to investigate this issue. We report that the vitamin D receptor (VDR) binds to a regulatory region of the Grainyhead-like 1 (GRHL1) gene and regulates its expression. Ectopic expression of VDR and treatment with calcitriol alters the expression of the GRHL1 gene. The evidence presented here indicates a role of VDR in the regulation of expression of GRHL1 and correspondingly a role of GRHL1 in mediating the actions of vitamin D. Full article
Show Figures

Figure 1

Figure 1
<p>Bioinformatic predictions of transcription factor binding site (TFBS) motifs for VDR in the human <span class="html-italic">GRHL1</span> gene. (<b>A</b>) Genomic location of regulatory regions of the <span class="html-italic">GRHL1</span> gene according to the Ensembl database. (<b>B</b>) VDR binding sites identified in the <span class="html-italic">GRHL1</span> gene using GTRD modules: meta-clusters, motifs, and GEM–PICS–MACS2–SISSRs clusters. (<b>C</b>) Genomic location of CpG Islands, markers of open chromatin (H3K4Me1, H3K4Me3, H3K27Ac), and transcription factor binding sites (ChIP-seq). (<b>D</b>) Multiple sequence alignment of a TFBS example across different species using T-Coffee (Pro-Coffee mode). Asterisks indicate conserved nucleotides.</p>
Full article ">Figure 2
<p>Baseline expression of <span class="html-italic">VDR</span> and <span class="html-italic">GRHL1–3</span> genes in various tissues. Based on <a href="https://www.proteinatlas.org/" target="_blank">https://www.proteinatlas.org/</a>, last accessed 11 June 2024. TPM, transcripts per million. Database version: Genotype-Tissue Expression (GTEx) RNA-seq data v8, available from the above portal.</p>
Full article ">Figure 3
<p>Overexpression of VDR alters mRNA level of <span class="html-italic">GRHL1</span> gene. (<b>A</b>) Genomic coordinates of the binding site for VDR in the promoter region of the <span class="html-italic">GRHL1</span> gene, obtained from the MotEvo database. (<b>B</b>) The mRNA expression levels of the <span class="html-italic">GRHL1–3</span> genes in HaCaT cells (1) transiently overexpressing VDR treated with ethanol, (2) transiently overexpressing VDR treated with 100 nM calcitriol, or (3) transfected with an empty vector treated with 100 nM calcitriol. The results represent relative expression of the respective target gene vs. <span class="html-italic">HPRT</span> genes. Data are shown as means ± SEM of experiments independently performed in triplicate, * significantly different at <span class="html-italic">p</span> ≤ 0.05.</p>
Full article ">Figure 4
<p>(<b>A</b>) Quantitative ChIP-PCR analysis of VDR occupancy of the <span class="html-italic">GRHL1</span> regulatory region was performed in HaCaT cells transfected with pcDNA3.1-K-DYK-VDR. Chromatin was immunoprecipitated with anti-DYK (FLAG) antibody or nonspecific antibody. The amount of DNA amplified from immunoprecipitated DNA was normalized to that amplified from input DNA. Data are shown as means ± SEM from experiments independently performed in triplicate, * significantly different at <span class="html-italic">p</span>≤ 0.05. (<b>B1</b>,<b>B2</b>) HaCaT cells were transfected with (<b>B1</b>) pcDNA3.1-K-DYK-VDR or pcDNA3.1-empty plasmid, 500 ng of the firefly luciferase vector with VDR binding site derived from the regulatory region of the <span class="html-italic">GRHL1</span> gene, and 25 ng pRL-CMV Renilla luciferase control reporter vector and treated with 100 nM calcitriol or (B2) pcDNA3.1-K-DYK-VDR, 500 ng of the firefly luciferase vector with VDR binding site derived from the regulatory region of the <span class="html-italic">GRHL1</span> gene, and 25 ng pRL-CMV Renilla luciferase control reporter vector and treated with 100 nM calcitriol or ethanol vehicle. Data are shown as means ± SEM of experiments independently performed in triplicate, * significantly different at <span class="html-italic">p</span>≤ 0.05.</p>
Full article ">
12 pages, 482 KiB  
Review
Adaptive and Maladaptive DNA Breaks in Neuronal Physiology and Alzheimer’s Disease
by Anysja Roberts, Russell H. Swerdlow and Ning Wang
Int. J. Mol. Sci. 2024, 25(14), 7774; https://doi.org/10.3390/ijms25147774 - 16 Jul 2024
Viewed by 617
Abstract
DNA strand breaks excessively accumulate in the brains of patients with Alzheimer’s disease (AD). While traditionally considered random, deleterious events, neuron activity itself induces DNA breaks, and these “adaptive” breaks help mediate synaptic plasticity and memory formation. Recent studies mapping the brain DNA [...] Read more.
DNA strand breaks excessively accumulate in the brains of patients with Alzheimer’s disease (AD). While traditionally considered random, deleterious events, neuron activity itself induces DNA breaks, and these “adaptive” breaks help mediate synaptic plasticity and memory formation. Recent studies mapping the brain DNA break landscape reveal that despite a net increase in DNA breaks in ectopic genomic hotspots, adaptive DNA breaks around synaptic genes are lost in AD brains, and this is associated with transcriptomic dysregulation. Additionally, relationships exist between mitochondrial dysfunction, a hallmark of AD, and DNA damage, such that mitochondrial dysfunction may perturb adaptive DNA break formation, while DNA breaks may conversely impair mitochondrial function. A failure of DNA break physiology could, therefore, potentially contribute to AD pathogenesis. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic of DNA Break Dynamics in AD Brains. Despite a net increase in DNA breaks at ectopic genomic hotspots, there is a significant loss of TOP2B-catalyzed adaptive DNA breaks around synaptic genes. This loss of adaptive DNA breaks is correlated with transcriptomic dysregulation, indicating potential disruptions in gene expression related to synaptic function.</p>
Full article ">
24 pages, 43777 KiB  
Article
Socket Array Irregularities and Wing Membrane Distortions at the Eyespot Foci of Butterfly Wings Suggest Mechanical Signals for Color Pattern Determination
by Yugo Nakazato and Joji M. Otaki
Insects 2024, 15(7), 535; https://doi.org/10.3390/insects15070535 - 16 Jul 2024
Viewed by 742
Abstract
Eyespot foci on butterfly wings function as organizers of eyespot color patterns during development. Despite their importance, focal structures have not been examined in detail. Here, we microscopically examined scales, sockets, and the wing membrane in the butterfly eyespot foci of both expanded [...] Read more.
Eyespot foci on butterfly wings function as organizers of eyespot color patterns during development. Despite their importance, focal structures have not been examined in detail. Here, we microscopically examined scales, sockets, and the wing membrane in the butterfly eyespot foci of both expanded and unexpanded wings using the Blue Pansy butterfly Junonia orithya. Images from a high-resolution light microscope revealed that, although not always, eyespot foci had scales with disordered planar polarity. Scanning electron microscopy (SEM) images after scale removal revealed that the sockets were irregularly positioned and that the wing membrane was physically distorted as if the focal site were mechanically squeezed from the surroundings. Focal areas without eyespots also had socket array irregularities, but less frequently and less severely. Physical damage in the background area induced ectopic patterns with socket array irregularities and wing membrane distortions, similar to natural eyespot foci. These results suggest that either the process of determining an eyespot focus or the function of an eyespot organizer may be associated with wing-wide mechanics that physically disrupt socket cells, scale cells, and the wing membrane, supporting the physical distortion hypothesis of the induction model for color pattern determination in butterfly wings. Full article
(This article belongs to the Section Insect Physiology, Reproduction and Development)
Show Figures

Figure 1

Figure 1
<p>Wing color patterns of the Blue Pansy butterfly <span class="html-italic">J. orithya</span>. (<b>a</b>) Dorsal wings. Males have a single blue form (right), and females have blue and brown forms (middle and left, respectively). In each wing, there are two eyespots, the anterior and posterior ones. Potential eyespot foci are named the first, second, third, fourth, fifth, and sixth focal areas from the anterior to posterior sides. An asterisk indicates the background area where physical damage was made to induce ectopic color patterns. (<b>b</b>) Dorsal cover scale image. Dorsal cover scales were removed at once from the wings with transparent adhesive tape for the purpose of illustrating wing veins and compartments. Scales along the wing veins are not present in this image, highlighting the wing veins. The anterior and posterior eyespot foci are located in the compartments M<sub>1</sub> and CuA<sub>1</sub>, respectively.</p>
Full article ">Figure 2
<p>Forewing eyespots. (<b>a</b>) The anterior eyespot (an arrow). (<b>b</b>) Magnification of the anterior eyespot shown in (<b>a</b>). (<b>c</b>) Focal area of (<b>b</b>). (<b>d</b>) Focal area without scales. An arrow indicates the location of the focal area, nearly at the end of the midline. This is a contralateral wing from the wing shown in (<b>a</b>–<b>c</b>). The following e and f are also contralateral wings. Thus, the wing margin is to the right. (<b>e</b>) Magnification of (<b>d</b>). (<b>f</b>) Magnification of (<b>e</b>). An arrow indicates a relatively large socket interval in the focal area. (<b>g</b>) The posterior eyespot (an arrow). (<b>h</b>) Magnification of the posterior eyespot shown in (<b>g</b>). (<b>i</b>) Focal area of (<b>h</b>). Scale directions appeared to be somewhat irregular. (<b>j</b>) Focal area without scales. An arrow indicates the location of the focal area, nearly at the end of the midline. This is a contralateral wing from the wing shown in (<b>g</b>–<b>i</b>). The following (<b>k</b>,<b>l</b>) are also contralateral wings. Thus, the wing margin is to the right. (<b>k</b>) Magnification of (<b>j</b>). (<b>l</b>) Magnification of (<b>k</b>). An arrow indicates a relatively large socket interval in the focal area.</p>
Full article ">Figure 3
<p>Anterior eyespot in the hindwing (female). (<b>a</b>) The anterior eyespot of an individual. (<b>b</b>) Magnification of the focal area of the posterior eyespot shown in (<b>a</b>). White focal scales are positioned without disorganization. (<b>c</b>) The eyespot area without scales. An arrow indicates the focal area at the end of the midline. This eyespot is identical to the one shown in (<b>a</b>,<b>b</b>). (<b>d</b>) Magnification of (<b>c</b>). (<b>e</b>) Magnification of (<b>d</b>). An arrow indicates an area of socket array irregularities. (<b>f</b>) Magnification of (<b>e</b>). An arrow indicates an area of socket array irregularities. (<b>g</b>) The anterior eyespot of another individual. (<b>h</b>) Magnification of the focal area of the posterior eyespot shown in (<b>g</b>). Blue and white focal scales are positioned without disorganization. (<b>i</b>) The eyespot without scales. An arrow indicates the focal area at the end of the midline. This eyespot is identical to the one shown in (<b>g</b>,<b>h</b>). (<b>j</b>) Magnification of (<b>i</b>). Arrows indicate distortions of the wing membrane. (<b>k</b>) Magnification of (<b>j</b>). Arrows indicate distortions of the wing membrane. (<b>l</b>) Magnification of (<b>k</b>).</p>
Full article ">Figure 4
<p>Posterior eyespot in the hindwing (male). (<b>a</b>) The posterior eyespot. (<b>b</b>) Magnification of the focal area of the posterior eyespot shown in (<b>a</b>). White focal scales are disorganized in direction. (<b>c</b>) The eyespot area without scales. The arrow indicates the focal area at the end of the midline. This eyespot is identical to the one shown in (<b>a</b>,<b>b</b>). (<b>d</b>) Magnification of (<b>c</b>). An arrow indicates an area of disorder. (<b>e</b>) Magnification of (<b>d</b>). Arrows indicate striking distortions of the wing membrane, one of which is associated with the unusual arrangement of socket arrays. (<b>f</b>) Magnification of (<b>e</b>). Arrows indicate striking distortions of the basal membrane. (<b>g</b>) Magnification of (<b>f</b>). (<b>h</b>) Another magnification of (<b>f</b>).</p>
Full article ">Figure 5
<p>Anterior and posterior eyespots in the unexpanded forewing (female). (<b>a</b>) The whole unexpanded forewing. (<b>b</b>) Magnification of the anterior eyespot shown in (<b>a</b>). (<b>c</b>) Magnification of the posterior eyespot shown in (<b>a</b>). (<b>d</b>) The anterior eyespot without scales. This is a contralateral wing from the wing shown in (<b>a</b>–<b>c</b>). Panels (<b>e</b>–<b>i</b>) also show images of the contralateral wing. Thus, the wing margin is to the left. (<b>e</b>) Magnification of (<b>d</b>). An arrow indicates the focal area, which is nearly at the end of the midline. (<b>f</b>) Magnification of (<b>e</b>). An arrow indicates a socket interval disorder. (<b>g</b>) Posterior eyespot without scales. (<b>h</b>) Magnification of the focal area of the posterior eyespot shown in (<b>g</b>). An arrow indicates the focal area, which is nearly at the end of the midline. (<b>i</b>) Magnification of (<b>h</b>). Arrows indicate irregularities in socket intervals.</p>
Full article ">Figure 6
<p>Anterior and posterior eyespots in the unexpanded hindwing (female). (<b>a</b>) The whole unexpanded hindwing. (<b>b</b>) Magnification of the anterior eyespot shown in (<b>a</b>). (<b>c</b>) Magnification of the posterior eyespot shown in (<b>a</b>). (<b>d</b>) The anterior eyespot area without scales. This is a contralateral wing from the wing shown in (<b>a</b>–<b>c</b>). (<b>e</b>) Magnification of (<b>d</b>). An arrow indicates the focal area. (<b>f</b>) Magnification of (<b>e</b>). An arrow indicates a socket interval disorder. (<b>g</b>) The posterior eyespot area without scales. This wing is obtained from an individual different from that of (<b>a</b>–<b>f</b>). (<b>h</b>) Magnification of the focal area of the posterior eyespot shown in (<b>g</b>). An arrow indicates the focal area, which is nearly at the end of the midline. (<b>i</b>) Magnification of (<b>h</b>). An arrow indicates disorders of socket intervals. (<b>j</b>) The posterior (fifth) eyespot area without scales from yet another individual. (<b>k</b>) Magnification of the focal area of the posterior eyespot shown in (<b>j</b>). An arrow indicates the focal area. (<b>l</b>) Magnification of (<b>k</b>). An arrow indicates irregularities in socket intervals.</p>
Full article ">Figure 7
<p>Potential focal areas in the unexpanded forewing. (<b>a</b>) The first focal area. (<b>b</b>) Magnification of the first focal area shown in (<b>a</b>). (<b>c</b>) Magnification of (<b>b</b>). (<b>d</b>) The third focal area. (<b>e</b>) Magnification of (<b>d</b>). (<b>f</b>) Magnification of (<b>e</b>). (<b>g</b>) The fourth focal area. (<b>h</b>) Magnification of the focal area shown in (<b>g</b>). (<b>i</b>) Magnification of (<b>h</b>).</p>
Full article ">Figure 8
<p>Potential focal areas in the unexpanded hindwing. (<b>a</b>) The first focal area. (<b>b</b>) Magnification of the first focal area shown in (<b>a</b>). (<b>c</b>) Magnification of (<b>b</b>). (<b>d</b>) The third focal area. (<b>e</b>) Magnification of (<b>d</b>). (<b>f</b>) Magnification of (<b>e</b>). (<b>g</b>) The fourth focal area. (<b>h</b>) Magnification of the focal area shown in (<b>g</b>). (<b>i</b>) Magnification of (<b>h</b>).</p>
Full article ">Figure 9
<p>Damage-induced hindwing color patterns. (<b>a</b>) A whole male wing with a damage-induced black spot. An arrow indicates the induced spot. (<b>b</b>) Magnification of the damage-induced black spot shown in (<b>a</b>). An arrow indicates the damaged point. (<b>c</b>) Magnification of (<b>b</b>). Scales are disordered in direction and density. (<b>d</b>) Damaged area without scales. The damaged area is indicated by an arrow. (<b>e</b>) Magnification of (<b>d</b>). Arrows indicate socket irregularities in the direction and substantial distortions of the wing membrane. (<b>f</b>) Magnification of (<b>e</b>). (<b>g</b>) A wing with a damage-induced orange area (an arrow). (<b>h</b>) Magnification of (<b>g</b>). There are black scales at the center (an arrow). (<b>i</b>) Magnification of (<b>h</b>). (<b>j</b>) A damaged area without scales. The arrow indicates the area of socket array irregularities. (<b>k</b>) Magnification of (<b>j</b>). An arrow indicates the area of socket array irregularity. (<b>l</b>) Magnification of (<b>k</b>). An arrow indicates an isolated socket with large intervals.</p>
Full article ">Figure 10
<p>Damage-induced color patterns in the unexpanded hindwing. (<b>a</b>) A whole male wing with a damage-induced black spot. An arrow indicates the induced spot. (<b>b</b>) Magnification of the damage-induced black spot shown in (<b>a</b>). (<b>c</b>) Magnification of (<b>b</b>). (<b>d</b>) Damaged area without scales. The damaged area is indicated by an arrow. (<b>e</b>) Magnification of (<b>d</b>). An arrow indicates the damaged area of the wing membrane. (<b>f</b>) Magnification of (<b>e</b>). There are many socket array irregularities (an arrow). (<b>g</b>) Wing with a damage-induced orange area (an arrow). (<b>h</b>) Magnification of (<b>g</b>). An arrow indicates the damaged area. (<b>i</b>) Magnification of (<b>h</b>). An arrow indicates the damaged area. (<b>j</b>) A damaged area without scales. An arrow indicates the area of socket array irregularities. (<b>k</b>) Magnification of (<b>j</b>). An arrow indicates the area of socket array irregularities. (<b>l</b>) Magnification of (<b>k</b>). An arrow indicates the irregular socket intervals.</p>
Full article ">Figure 11
<p>Female anterior eyespot focal areas of the nonrandom sampling group via light microscopy and SEM. Four representative samples are shown. Each line of panels contains images of a single individual. (<b>a</b>) Eyespot focal area of individual No. 30. (<b>b</b>,<b>c</b>) Wing membrane distortions at a branching point of socket arrays (arrows). (<b>d</b>) Magnification of (<b>b</b>). (<b>e</b>) Magnification of (<b>c</b>). (<b>f</b>) Eyespot focal area of individual No. 36. (<b>g</b>) Socket arrays with many branching points. Arrows indicate ridges or furrows parallel to socket arrays. (<b>h</b>) Socket arrays with irregularities and wing membrane distortion (an arrow). (<b>i</b>) Socket arrays with displaced sockets (arrows). (<b>j</b>) Magnification of (<b>h</b>). (<b>k</b>) Eyespot focal area of individual No. 39. (<b>l</b>) Socket arrays with many branches. (<b>m</b>) A displaced socket with wing membrane distortion (an arrow). (<b>n</b>) Magnifiction of (<b>m</b>). (<b>o</b>) A socket with the wing membrane distortion at a branching point of socket arrays. (<b>p</b>) Eyespot focal area of individual No. 42. (<b>q</b>) Socket arrays with an irregular socket at a branching point (an arrowhead). Ridges perpendicular to socket arrays are indicated (arrows). (<b>r</b>) Socket arrays with perpendicular ridges (arrows) and a parallel furrow (an arrowhead). (<b>s</b>,<b>t</b>) Magnification of q.</p>
Full article ">Figure 12
<p>Female anterior eyespot focal areas of the random sampling group via light microscopy and SEM. Four representative samples are shown. Each line of panels contains images of a single individual. (<b>a</b>) Eyespot focal area of individual No. 12. (<b>b</b>) Socket arrays with irregularities. Long (an arrow) and short (an arrowhead) socket intervals are indicated. (<b>c</b>) Socket arrays with an extra socket (an arrow) and wing membrane distortion (an arrowhead). (<b>d</b>,<b>e</b>) Magnification of (<b>c</b>). (<b>f</b>) Eyespot focal area of individual No. 17. (<b>g</b>) Socket arrays with many branching points. (<b>h</b>) Socket arrays with a long socket interval (an arrow) and a displaced socket at a branching point (an arrowhead). (<b>i</b>) Magnification of (<b>h</b>) (left side). (<b>j</b>) Magnification of (<b>h</b>). (<b>k</b>) Eyespot focal area of individual No. 23. (<b>l</b>) Socket arrays with a relatively small number of branching points. (<b>m</b>) Magnification of (<b>l</b>). (<b>n</b>) Socket array with an irregular socket (an arrow). (<b>o</b>) Magnification of (<b>n</b>). (<b>p</b>) Eyespot focal area of individual No. 43. (<b>q</b>) Socket arrays with branching points. (<b>r</b>) Magnification of (<b>q</b>). (<b>s</b>) Magnification of (<b>q</b>). A displaced socket at a branching point is indicated by an arrow. (<b>t</b>) Magnification of (<b>s</b>). An isolated large socket is shown.</p>
Full article ">Figure 13
<p>Physical distortions and mechanical signals. (<b>a</b>) Illustration of the physical distortions of the wing membrane detected in this study. The distortion is located at the tip of the midline. (<b>b</b>) Possible mechanical forces from the wing veins, the marginal band organizer, and the discal organizer (left). The prospective eyespot organizer then pushes back to balance forces (right). (<b>c</b>) Feedback and reinforcement of the mechanical signals from the prospective eyespot organizer. Reinforcement indicates further secretion of the cuticle to bind to the pupal cuticle focal spot more tightly and the polyploidization of organizing cells.</p>
Full article ">
13 pages, 2491 KiB  
Article
Excess Dally-like Induces Malformation of Drosophila Legs
by Xubo Zhang, Yi Wang, Wenting Zhao, Shumin Yang, Bernard Moussian, Zhangwu Zhao, Jianzhen Zhang and Wei Dong
Cells 2024, 13(14), 1199; https://doi.org/10.3390/cells13141199 - 15 Jul 2024
Viewed by 542
Abstract
Glypicans are closely associated with organ development and tumorigenesis in animals. Dally-like (Dlp), a membrane-bound glypican, plays pivotal roles in various biological processes in Drosophila. In this study, we observed that an excess of Dlp led to the malformation of legs, particularly [...] Read more.
Glypicans are closely associated with organ development and tumorigenesis in animals. Dally-like (Dlp), a membrane-bound glypican, plays pivotal roles in various biological processes in Drosophila. In this study, we observed that an excess of Dlp led to the malformation of legs, particularly affecting the distal part. Accordingly, the leg disc was shrunken and frequently exhibited aberrant morphology. In addition, elevated Dlp levels induced ectopic cell death with no apparent cell proliferation changes. Furthermore, Dlp overexpression in the posterior compartment significantly altered Wingless (Wg) distribution. We observed a marked expansion of Wg distribution within the posterior compartment, accompanied by a corresponding decrease in the anterior compartment. It appears that excess Dlp guides Wg to diffuse to cells with higher Dlp levels. In addition, the distal-less (dll) gene, which is crucial for leg patterning, was up-regulated significantly. Notably, dachshund (dac) and homothorax (hth) expression, also essential for leg patterning and development, only appeared to be negligibly affected. Based on these findings, we speculate that excess Dlp may contribute to malformations of the distal leg region of Drosophila, possibly through its influence on Wg distribution, dll expression and induced cell death. Our research advances the understanding of Dlp function in Drosophila leg development. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Overexpression but not repression of Dlp-induced defects of <span class="html-italic">Drosophila</span> legs. (<b>A</b>). Normal adult leg segmentation in control <span class="html-italic">en<sup>ts</sup></span> &gt; GFP flies. (<b>B</b>). Adult legs of <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>-RNAi flies show no visible defects. (<b>C</b>). Overexpression of Dlp in the <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> leg disc results in a characteristic shrinkage of the distal portion of the adult leg. The scale bar is 0.3 mm. (<b>D</b>). Statistical analysis of distal part length of adult legs shows the tarsal segments of adult legs are abbreviated significantly in <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> flies, while they are normal in <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>-RNAi flies (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 22; <span class="html-italic">en<sup>ts</sup></span>&gt; <span class="html-italic">dlp</span>-RNAi <span class="html-italic">n</span> = 20; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 23). Bars with different letters indicate significant statistical differences between the groups (<span class="html-italic">p</span> &lt; 0.01). (<b>E</b>). Anti-Dlp staining (red) shows that Dlp is roughly uniform in the control <span class="html-italic">en<sup>ts</sup></span> &gt; GFP leg discs; Dlp is effectively repressed in the P compartment of <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>-RNAi leg discs; Dlp is up-regulated in <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> leg discs. The scale bar is 50 μm. (<b>F</b>). Statistical analysis of the proportion of the P compartment area to the whole leg disc (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 16; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 16). The white dashed lines indicate the A–P compartment boundary. Asterisks indicate significant differences (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 2
<p>Excess Dlp induced shrinkage of leg discs with no apparent cell proliferation alteration. (<b>A</b>). Phalloidin staining reveals normal leg disc morphology in the control group (<span class="html-italic">en<sup>ts</sup></span> &gt; GFP), while overexpression of Dlp results in morphological deformities in the P compartment. (<b>B</b>). Staining with anti-PH3 reveals no apparent change in cell proliferation rate between the control and Dlp-overexpressing leg discs. (<b>C</b>). Statistical analysis of the PH3-positive puncta density (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 22; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 24). It reveals no apparent change in the cell proliferation rate between the control and Dlp-overexpressing leg discs. The white dashed lines indicate the A–P compartment boundary. ns means no statistically significant differences. The scale bar is 50 μm.</p>
Full article ">Figure 3
<p>Dlp overexpression induces cell apoptosis both autonomously and non-autonomously. (<b>A</b>). No apparent cell death is detected in the control leg disc. (<b>B</b>). Overexpression of Dlp induces marked cell apoptosis within the P compartment (autonomous) and in regions outside of Gal4 expression (non-autonomous). (<b>C</b>). Expression of p35 blocks the cell apoptosis in the P compartment, while non-autonomous cell death still persists in the central region of leg disc. (<b>D</b>). The size of P compartment is partially rescued by co-expression of <span class="html-italic">p35</span> (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 16; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 16; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> + <span class="html-italic">p35</span>, <span class="html-italic">n</span> = 16) (<b>E</b>). Cell death in P compartment is inhibited totally by <span class="html-italic">p35</span>, while the non-autonomous cell death in central region is still severe (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 23; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 25; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> + <span class="html-italic">p35</span>, <span class="html-italic">n</span> = 16). The white dashed lines indicate the A–P compartment boundary. Asterisks indicate significant differences (<span class="html-italic">p</span> &lt; 0.01). The scale bar is 50 μm.</p>
Full article ">Figure 4
<p>Excess Dlp changes Wg distribution in the leg disc. (<b>A</b>). In the control leg disc, the expression of <span class="html-italic">wg</span>-lacZ and the majority of Wg distribution are confined to a wedge-shaped region within the A compartment. (<b>B</b>). Overexpression of Dlp expands the Wg distribution domain within the P compartment, while concurrently causing a reduction in the distribution domain within the A compartment. <span class="html-italic">wg</span>-lacZ expression domain is enlarged. The white dashed lines indicate the A–P compartment boundary. (<b>C</b>). In the <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> leg disc, the Wg distribution domain in the P compartment is expanded; it is decreased in the A compartment (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 24; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 22). (<b>D</b>). The <span class="html-italic">wg</span>-lacZ expression domain is enlarged in the <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span> leg disc (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 24; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 21). Asterisks indicate significant differences (<span class="html-italic">p</span> &lt; 0.01). The scale bar is 50 μm.</p>
Full article ">Figure 5
<p>Excess Dlp causes mis-expression of <span class="html-italic">dll</span> but not notable alteration of <span class="html-italic">hth</span> and <span class="html-italic">dac</span>. (<b>A</b>). Excess Dlp has no apparent effect on the <span class="html-italic">hth</span> expression pattern. (<b>B</b>). Excess Dlp has no apparent effect on the Dac expression pattern. (<b>C</b>). Excess Dlp causes up-regulated <span class="html-italic">dll</span> in the edge region of the P compartment and the region adjacent to the <span class="html-italic">en</span>-Gal4 domain. (<b>D</b>). The statistical analysis shows that the <span class="html-italic">dll</span> expression is elevated significantly (mean ± SEM; <span class="html-italic">en<sup>ts</sup></span> &gt; GFP, <span class="html-italic">n</span> = 23; <span class="html-italic">en<sup>ts</sup></span> &gt; <span class="html-italic">dlp</span>, <span class="html-italic">n</span> = 21). The white dashed lines indicate the A–P compartment boundary. Asterisks indicate significant differences (<span class="html-italic">p</span> &lt; 0.01). The scale bar is 50 μm.</p>
Full article ">Figure 6
<p>Schematic diagram illustrating the effects of excess Dlp in the P compartment of the leg disc. It leads to abnormal Wg distribution, up-regulated <span class="html-italic">dll</span> expression and noticeable cell death, which potentially contribute to the abbreviated distal part of adult legs.</p>
Full article ">
15 pages, 4051 KiB  
Article
Effect of Trace Element Selenium on the Intestinal Microbial Community in Nude Mice with Colorectal Cancer
by Yintong Su, Xiaohua Cai, Xingxing Fan, Jiayu Ning and Mei Shen
Microorganisms 2024, 12(7), 1336; https://doi.org/10.3390/microorganisms12071336 - 29 Jun 2024
Viewed by 818
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as [...] Read more.
Colorectal cancer (CRC) is the third most common cancer worldwide. The role of intestinal microbiota in carcinogenesis has also become an important research topic, and CRC is closely related to the intestinal microbiota. Selenium-containing compounds have attracted more attention as anticancer drugs as they can have minimal side effects. The purpose of this study was to determine and compare the effect of sodium selenite and selenomethionine on the microbial communities of nude mice with CRC. A CRC ectopic tumorigenesis model was established by subcutaneously injecting HCT116 cells into nude mice. The mice were then intraperitoneally injected with sodium selenite and selenomethionine for 24 days to regulate their intestinal microbiota. Compared with sodium selenite, selenomethionine resulted in a greater reduction in the richness and diversity of intestinal microbiota in nude mice with CRC, and the richness and diversity were closer to healthy levels. Selenomethionine also regulated a wider variety of flora. Additionally, sodium selenite and selenomethionine produced different microorganisms, changed function and metabolic pathways in the intestinal microbiota. Both sodium selenite and selenomethionine have certain effects on restoring the intestinal microbial diversity in nude mice with CRC, and the effect of selenomethionine is better than that of sodium selenite. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of sodium selenite and selenomethionine.</p>
Full article ">Figure 2
<p>Schematic diagram of the experimental design.</p>
Full article ">Figure 3
<p>Inhibitory effects of sodium selenite and selenomethionine on nude mice with CRC. (<b>A</b>) Trend chart of body weight in each group of mice. (<b>B</b>) Tumor weight of mice in Control group, SSe group and SeMet group. (<b>C</b>) H&amp;E staining results of tumor tissues (×200) and the expression of Ki67 in tumor tissues analyzed by IHC staining (×400) of mice in Control group, SSe group and SeMet group. (* indicating <span class="html-italic">p</span> &lt; 0.05, ** indicating <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 4
<p>Alpha diversity analysis. (<b>A</b>) Rarefaction curves were constructed using Sobs indices. (<b>B</b>) Rank–abundance curves on OTU level. (<b>C</b>) Measures of richness using Sobs index. (<b>D</b>) Measures of diversity using Shannon index. (* indicating <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Partial least squares discriminant analysis (PLS-DA) analysis on the OTU level.</p>
Full article ">Figure 6
<p>The effect of sodium selenite and selenomethionine on intestinal microbiota composition. (<b>A</b>) Relative abundance at the phylum level in fecal microbiota of each group. (<b>B</b>) Relative abundance at the genus level in fecal microbiota of each group. (<b>C</b>) Difference analysis between Blank group and Control group. (<b>D</b>) Difference analysis between Control group and SSe group. (<b>E</b>) Difference analysis between Control group and SeMet group. (* indicating <span class="html-italic">p</span> &lt; 0.05, ** indicating <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 7
<p>Cladogram of significant difference between groups. (<b>A</b>) Cladogram constructed using the linear discriminant analysis effect size (LefSe) method to indicate the phylogenetic distribution of bacteria that were remarkably enriched between each group. Yellow circles indicate species with no significant differences. (<b>B</b>) Linear discriminant analysis (LDA) scores represent the gut bacteria which were of important biological significance in each group.</p>
Full article ">Figure 8
<p>Bar chart of the Clusters of Orthologous Genes (COG) function classification in (<b>A</b>) Blank group, (<b>B</b>) Control group, (<b>C</b>) SSe group and (<b>D</b>) SeMet group. (<b>E</b>) Bar chart and (<b>F</b>) box plot of COG functions with differences in each group. N: cell motility; I: lipid transport and metabolism; K: transcription; J: translation, ribosomal structure and biogenesis. (* indicating <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 9
<p>Functional analysis of (<b>A</b>) enzyme and (<b>B</b>) Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway level3 based on KEGG database.</p>
Full article ">
17 pages, 1154 KiB  
Article
Integrative Bioinformatics Analysis Reveals a Transcription Factor EB-Driven MicroRNA Regulatory Network in Endothelial Cells
by Teresa Gravina, Francesco Favero, Stefania Rosano, Sushant Parab, Alejandra Diaz Alcalde, Federico Bussolino, Gabriella Doronzo and Davide Corà
Int. J. Mol. Sci. 2024, 25(13), 7123; https://doi.org/10.3390/ijms25137123 - 28 Jun 2024
Viewed by 861
Abstract
Various human diseases are triggered by molecular alterations influencing the fine-tuned expression and activity of transcription factors, usually due to imbalances in targets including protein-coding genes and non-coding RNAs, such as microRNAs (miRNAs). The transcription factor EB (TFEB) modulates human cellular networks, overseeing [...] Read more.
Various human diseases are triggered by molecular alterations influencing the fine-tuned expression and activity of transcription factors, usually due to imbalances in targets including protein-coding genes and non-coding RNAs, such as microRNAs (miRNAs). The transcription factor EB (TFEB) modulates human cellular networks, overseeing lysosomal biogenesis and function, plasma–membrane trafficking, autophagic flux, and cell cycle progression. In endothelial cells (ECs), TFEB is essential for the maintenance of endothelial integrity and function, ensuring vascular health. However, the comprehensive regulatory network orchestrated by TFEB remains poorly understood. Here, we provide novel mechanistic insights into how TFEB regulates the transcriptional landscape in primary human umbilical vein ECs (HUVECs), using an integrated approach combining high-throughput experimental data with dedicated bioinformatics analysis. By analyzing HUVECs ectopically expressing TFEB using ChIP-seq and examining both polyadenylated mRNA and small RNA sequencing data from TFEB-silenced HUVECs, we have developed a bioinformatics pipeline mapping the different gene regulatory interactions driven by TFEB. We show that TFEB directly regulates multiple miRNAs, which in turn post-transcriptionally modulate a broad network of target genes, significantly expanding the repertoire of gene programs influenced by this transcription factor. These insights may have significant implications for vascular biology and the development of novel therapeutics for vascular disease. Full article
(This article belongs to the Special Issue Genes and Human Diseases 2.0)
Show Figures

Figure 1

Figure 1
<p>Overview of the bioinformatics analysis pipeline for defining the TFEB-driven regulatory network in the HUVECs. The blue rectangles represent the databases used to obtain annotations of transcription start sites (TSSs) on the human genome, depicted in green, for mRNAs, ncRNAs, and pri-miRNAs. The orange rectangles indicate the omics datasets used to identify the differentially expressed genes (DEGs) and the differentially expressed miRNAs (DEMs) showing a TFEB binding peak in their promoter regions. The integration of these data sets was conducted to identify a single comprehensive network of regulatory interactions, with TFEB as the master regulator (De Rie et al., 2017, [<a href="#B25-ijms-25-07123" class="html-bibr">25</a>]).</p>
Full article ">Figure 2
<p>mRNA-Seq data analysis reveals a TFEB role in regulating protein-coding and ncRNA genes in HUVECs. (<b>a</b>) Volcano plot showing differentially expressed genes (DEGs) in TFEB-silenced (shRNA-TFEB) HUVECs compared to control cells (WT). The green dots represent the downregulated genes, while the red dots represent the upregulated ones. The blue dot indicates the TFEB gene itself. (<b>b</b>) Heatmap showing unsupervised hierarchical clustering of DEGs between the TFEB-silenced HUVECs (orange) and WT (cyan). Each DEG is annotated to show the presence (green) or absence (red) of a significant TFEB binding event in its promoter region. (<b>c</b>) Venn diagram depicting the overlap between DEGs in TFEB-silenced HUVECs and WT cells, along with the total number of genes bound by TFEB in their promoter regions. (<b>d</b>) Selected GO terms obtained from the ToppGene functional analysis of the DEGs. The GO terms associated with downregulated genes are shown in blue, while those associated with upregulated genes are red. The number of genes associated with each term is indicated in parentheses. GO terms are ranked by –log10(p-value), adjusted using the Benjamini–Hochberg method.</p>
Full article ">Figure 3
<p>TFEB-mediated regulation of miRNAs in HUVECs. (<b>a</b>) Heatmap showing the unsupervised hierarchical clustering of differentially expressed miRNAs (DEMs) between the TFEB-silenced (shRNA-TFEB) (orange) and control (WT) HUVECs (cyan). Each miRNA is annotated to indicate the presence (green) or absence (red) of a TFEB significant binding event in its promoter region. In addition, each miRNA is annotated to show whether the H3K27ac and H3K4me3 marks are present (light blue) or absent (light grey) in its associated promoter region, according to ENCODE data from HUVECs. (<b>b</b>) Venn diagram showing the overlap between miRNAs among TFEB-silenced HUVECs and WT groups and the total number of miRNAs bound by TFEB in their promoter regions. (<b>c</b>) Heatmap showing the unsupervised hierarchical clustering of differentially expressed miRNAs (annotated in pink) characterized by TFEB in their promoter region (annotated in green) between the TFEB-silenced HUVECs (orange) and WT conditions (cyan). Each miRNA is also annotated to display the presence (yellow) or absence (purple) of the TFEB motif in its associated promoter region, based on data from the JASPAR database. (<b>d</b>) Graphical representation of the miRNA-222 locus on the human genome hg38, sourced from the UCSC Genome Browser. The graphic includes annotations for the miR-222 host gene (green) and its putative promoter region (−2500, +2500 DNA bases around the TSS, blue). At the top, the TFEB binding signal in HUVECs is shown in black. At the bottom, the data for H3K27ac and H3K4me3 from ENCODE HUVECs are shown.</p>
Full article ">Figure 4
<p>TFEB/miRNAs-mediated regulatory networks (<b>a</b>) Venn diagram representing the overlap between the number of anti-correlated target genes of the 7 DEMs between TFEB-silenced (shRNA-TFEB) and control (WT) HUVECs, with respect to the DEGs identified when comparing TFEB-silenced to WT HUVECs. These target genes are further characterized by the presence of TFEB binding sites in their promoter regions. (<b>b</b>) TFEB-mediated network featuring the 7 DEMs with TFEB presence in their promoter regions, along with their 152 differentially expressed target genes. Specifically, TFEB is indicated by green triangles, miRNAs by magenta diamonds, and differentially expressed target genes by blue squares. Notably, some of these target genes exhibit TFEB binding in their promoter regions, while others do not. (<b>c</b>) A focused subset of the broader regulatory network that specifically includes TFEB, the 7 DEMs, and their 44 differentially expressed target genes, where each node is marked by the presence of TFEB binding in its associated promoter region. This subset underscores the specific interactions within the network, where TFEB more likely directly influences gene expression through its binding activity.</p>
Full article ">Figure 5
<p>TFEB-miR-222-E2F1/CDK1 network in HUVECs (<b>a</b>,<b>b</b>): quantitative PCR (qPCR) analysis of hsa-pri-miR-222 and mature hsa-miR-222-5p or 3p (<b>a</b>), alongside E2F1 and CDK1 (<b>b</b>), in TFEB-silenced (shRNA-TFEB) or WT HUVECs. Data are expressed as relative fold-change compared to the expression in WT cells, normalized to the housekeeping gene TBP (n = 4, mean ± SEM; * <span class="html-italic">p</span>-value &lt; 0.01, ** <span class="html-italic">p</span>-value &lt; 0.001 and *** <span class="html-italic">p</span>-value &lt; 0.0001, determined by Student’s <span class="html-italic">t</span>-test).</p>
Full article ">
21 pages, 2729 KiB  
Article
Anti-Adenoviral Effect of Human Argonaute 2 Alone and in Combination with Artificial microRNAs
by Philipp Ausserhofer, Izabella Kiss, Angela Witte and Reinhard Klein
Cells 2024, 13(13), 1117; https://doi.org/10.3390/cells13131117 - 28 Jun 2024
Viewed by 683
Abstract
During infection, adenoviruses inhibit the cellular RNA interference (RNAi) machinery by saturating the RNA-induced silencing complex (RISC) of the host cells with large amounts of virus-derived microRNAs (mivaRNAs) that bind to the key component of the complex, Argonaute 2 (AGO2). In the present [...] Read more.
During infection, adenoviruses inhibit the cellular RNA interference (RNAi) machinery by saturating the RNA-induced silencing complex (RISC) of the host cells with large amounts of virus-derived microRNAs (mivaRNAs) that bind to the key component of the complex, Argonaute 2 (AGO2). In the present study, we investigated AGO2 as a prominent player at the intersection between human adenovirus 5 (HAdV-5) and host cells because of its ability to interfere with the HAdV-5 life cycle. First, the ectopic expression of AGO2 had a detrimental effect on the ability of the virus to replicate. In addition, in silico and in vitro analyses suggested that endogenous microRNAs (miRNAs), particularly hsa-miR-7-5p, have similar effects. This miRNA was found to be able to target the HAdV-5 DNA polymerase mRNA. The inhibitory effect became more pronounced upon overexpression of AGO2, likely due to elevated AGO2 levels, which abolished the competition between cellular miRNAs and mivaRNAs for RISC incorporation. Collectively, our data suggest that endogenous miRNAs would be capable of significantly inhibiting viral replication if adenoviruses had not developed a mechanism to counteract this function. Eventually, AGO2 overexpression-mediated relief of the RISC-saturating action of mivaRNAs strongly enhanced the effectiveness of artificial miRNAs (amiRNAs) directed against the HAdV-5 preterminal protein (pTP) mRNA, suggesting a substantial benefit of co-expressing amiRNAs and AGO2 in RNAi-based strategies for the therapeutic inhibition of adenoviruses. Full article
(This article belongs to the Section Cell and Gene Therapy)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Schematic representation of the adenoviral vectors used in this study. All vectors were based on the HAd5V-derived vectors pAd/PL-DEST ™ (ThermoFisher Scientific, Vienna, Austria) lacking the E1 and E3 genes. Expression cassettes were inserted into the deleted E1 region in antisense orientation with respect to the left inverted terminal repeat (ITR). The expression cassettes contain EGFP [<a href="#B34-cells-13-01117" class="html-bibr">34</a>] or AGO2 (this study) open reading frames, either alone or in conjunction with six tandemly repeated, either targeting (pTP-mi5) or non-targeting (NT), amiRNA hairpins incorporated into the 3′ UTR of the EGFP and AGO2 transcripts, respectively. Expression is driven by a CMV promoter (pCMV).</p>
Full article ">Figure 2
<p>AGO2 negatively affects the HAdV-5 life cycle. (<b>A</b>) Overexpression of AGO2 decreases the wt HAdV-5 genome copy numbers. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with 250 ng plasmid and infected with wt HAdV-5 at an MOI of 0.1, respectively. Concentrations of HAdV-5 genome copy numbers were measured at 96 h post-transfection/infection using an E3-probe qPCR. The data display means and standard deviations from a total of 3 independent experiments. Each independent experiment’s EGFP control preparation measurement was set as a reference at 100%. ** (<span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) Overexpression of AGO2 has no effect on the adenovirus mutant dl-sub720. Same as in (<b>A</b>), except that cells were infected with the HAdV-5 mutant dl-sub720 instead of wt HAdV-5. ns (not significant). (<b>C</b>) Compared to wt AGO2 mutants defective for miRNA binding, they differ in their ability to decrease wt HAdV-5 genome copy numbers. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with 250 ng plasmid expressing either AGO2 or mutants thereof and were infected with wt HAdV-5 at an MOI of 0.1. Concentrations of HAdV-5 genome copy numbers were measured at 96 h post-transfection/infection using an E3-probe qPCR. The data represent the means of 3 representative experiments, including standard deviations. The mean value for the AGO2 measurements was set as a reference at 100%. * (<span class="html-italic">p</span> &lt; 0.05); ** (<span class="html-italic">p</span> &lt; 0.01). (<b>D</b>) The inhibition of HAdV-5 replication by AGO2 is comparable in A549 and HeLa cells. 1.5 × 10<sup>4</sup> A549 or HeLa cells were transduced with AGO2-expressing rAdV vectors at an MOI of 100. 12 h after transduction wt HAdV-5 was added at an MOI of 0.1. Concentrations of HAdV-5 genome copy numbers were measured at 48 h post-infection using an E3-probe qPCR. The data were derived from a total of 3 representative experiments and display mean ± standard deviations. Each experiment’s EGFP control preparation measurement was set as a reference at 100%. * (<span class="html-italic">p</span> &lt; 0.05); ** (<span class="html-italic">p</span> &lt; 0.01); ns (not significant).</p>
Full article ">Figure 3
<p>Putative target sites of cellular miRNAs within HAdV-5 early mRNA transcripts Pol and pTP as per in silico TargetScan analysis. In addition to putative target sites of cellular miRNAs (red arrows), the target sites of the previously described siRNAs Pol-si2 (blue arrow) and pTP-si8 [<a href="#B42-cells-13-01117" class="html-bibr">42</a>] with their corresponding amiRNA pTPmi5 (green arrow) [<a href="#B34-cells-13-01117" class="html-bibr">34</a>] are indicated.</p>
Full article ">Figure 4
<p>Targeting of HAdV-5 DNA polymerase and pTP sequences by cellular miRNAs and their impact on viral replication. (<b>A</b>) Schematic representation of the dual-luciferase reporter vector system employed in this study. Reporter vectors contain sequences of the HAdV-5 DNA polymerase and pTP genes (Pol; pTP) inserted into the 3′UTR of a Renilla luciferase reporter gene (RLuc). MiRNAs capable of recognizing the respective target mRNAs are expected to knock down Renilla luciferase expression relative to the expression of a non-targeted firefly luciferase gene (FLuc) present on the same vector. (<b>B</b>) Targeting of the HAdV-5 DNA polymerase mRNA by miRNA mimics in reporter assays. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with a HAdV-5 DNA polymerase reporter vector carrying the DNA polymerase sequence inserted into the 3′UTR of a Renilla luciferase reporter gene and endogenous miRNA mimics, respectively. A non-targeting (NT) miRNA mimic was used as a control. Readout as per manufacturers’ instructions was conducted at 48 h post-transfection and relative light units (RLUs) for the Renilla luciferase reporter gene were normalized to those of the firefly luciferase reporter gene. Each experiment’s NT control preparation measurement was set as a reference at 100%. The data represent means ± standard deviation of 3 experiments. * (<span class="html-italic">p</span> &lt; 0.05). (<b>C</b>) Targeting of the HAdV-5 pTP mRNA by miRNA mimics in reporter assays. Same as in (<b>B</b>) except that a reporter vector carrying the HAdV-5 pTP sequence was used. (<b>D</b>) Effect of the hsa-miR-7 mimic on HAdV-5 genome copy numbers. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with an hsa-miR-7 mimic at a concentration of 10 nM and infected with HAdV-5 at an MOI of 0.1. Concentrations of HAdV-5 genome copy numbers were measured at 48 h post-transfection/infection using an E3-probe qPCR. Data represent mean ± standard deviations of a representative experiment carried out in triplicate. The non-targeting (NT) control preparation measurement was set as a reference at 100%. ** (<span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 5
<p>Inhibition of miRNA function with miRNA inhibitors and enhancement of miRNA-mediated effects on HAdV-5 by overexpression of AGO2. (<b>A</b>) Inhibition of the action of miRNA mimics with miRNA inhibitors in reporter assays. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with (i) a HAdV-5 DNA polymerase reporter vector carrying the DNA polymerase sequence inserted into the 3′UTR of a Renilla luciferase reporter gene, (ii) miRNA mimics, and (iii) corresponding miRNA Power Inhibitors (PI). Readout as per manufacturers’ instructions was conducted at 48 h post-transfection. Data (relative light units; RLU) derives from a total of 3 experiments and displays means ± standard deviations. Each experiment’s non-targeting (NT) control preparation measurement was set as a reference at 100%. ** (<span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) Effect of miRNA inhibitors in the absence of miRNA mimics in reporter assays. Same as in (<b>A</b>) except that no miRNA mimics directed against the reporter transcript were employed. ** (<span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Effect of miRNA mimics on HAdV-5 replication. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transfected with 5 nM miRNA mimics and transduced with an AGO2- or EGFP-expressing adenoviral vector at an MOI of 100. 24 h later, wt HAdV-5 was added at an MOI of 0.1. Concentrations of wt HAdV-5 genome copy numbers were measured at 48 h post-infection using an E3-probe qPCR. Data represent means ± standard deviations of a representative experiment carried out in triplicate. * (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Improved amiRNA-mediated inhibition of wt HAdV-5 replication upon co-transduction with an AGO2-expressing rAdV vector. (<b>A</b>) The potency of an amiRNA targeting the adenoviral pTP mRNA is significantly improved upon overexpression of AGO2. 1.5 × 10<sup>4</sup> cells were simultaneously transduced with (i) a rAdV expressing HAdV-5-targeting (pTP-mi5) or non-targeting (NT) amiRNAs and (ii) a rAdV expressing AGO2 or EGFP at an MOI of 250, before being exposed to wt HAdV-5 at an MOI of 1. Concentrations of wt HAdV-5 genome copy numbers were measured at 72 h post-infection using an E3-probe qPCR. Each experiment’s EGFP control preparation measurement was set as a reference at 100%. The inhibition by AGO2 alone in the absence of any targeting or non-targeting amiRNA is shown for comparison. Data were derived from a total of 3 representative experiments and display mean ± standard deviations. * (<span class="html-italic">p</span> &lt; 0.05); ns (not significant). AGO2-mediated differences between 6xNT and 6xpTP-mi5 were significant in all instances. (<b>B</b>) Evaluation of the impact of adenoviral vector MOIs in relation to wt HAdV-5 MOIs on viral replication and vector mobilization. 1.5 × 10<sup>4</sup> HeLa cells were simultaneously transduced with rAdVs at MOIs as per the X-axis and infected with wt HAdV-5 as per the Y-axis. Concentrations of wt HAdV-5 and vector genome copy numbers were measured at 48 h post-infection using E3- and CMV promoter-specific qPCR probes, respectively.</p>
Full article ">Figure 7
<p>Improvement of rAdV amiRNA vector performance by inclusion of an AGO2 expression cassette. (<b>A</b>) In a prophylactic treatment scenario, 1.5 × 10<sup>4</sup> HeLa cells were transduced with vectors at an MOI of 100 24 h prior to infection with HAdV-5 at an MOI of 0.1. Concentrations of wt HAdV-5 infectious particles (TCID50/mL) were measured at timepoints 0 (D0), 48 h (D2), 96 h (D4), and 144 h (D6). Data were derived from a total of 3 experiments and display mean ± standard deviations. **** (<span class="html-italic">p</span> &lt; 0.0001). Significances in green: 6xNT+EGFP vs. 6xpTP-mi5+EGFP; significances in red: 6xNT+AGO2 vs. 6xpTP-mi5+AGO2. (<b>B</b>) In a therapeutic treatment scenario 1.5 × 10<sup>4</sup> HeLa cells underwent concomitant transduction and infection with vectors at an MOI of 100 as well as HAdV-5 at an MOI of 0.1, respectively. Concentrations of wt HAdV-5 infectious particles (TCID50/mL) were measured at timepoints 0 (D0), 48 h (D2), 96 h (D4), and 144 h (D6). Data were derived from a total of 3 experiments and display mean ± standard deviations. **** (<span class="html-italic">p</span> &lt; 0.0001); ** (<span class="html-italic">p</span> &lt; 0.01); * (<span class="html-italic">p</span> &lt; 0.05). Significances in green: 6xNT+EGFP vs. 6xpTP-mi5+EGFP; significances in red: 6xNT+AGO2 vs. 6xpTP-mi5+AGO2.</p>
Full article ">
27 pages, 1592 KiB  
Review
A Glimpse into Humoral Response and Related Therapeutic Approaches of Takayasu’s Arteritis
by Shuning Guo, Yixiao Tian, Jing Li and Xiaofeng Zeng
Int. J. Mol. Sci. 2024, 25(12), 6528; https://doi.org/10.3390/ijms25126528 - 13 Jun 2024
Viewed by 948
Abstract
Takayasu’s arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing [...] Read more.
Takayasu’s arteritis (TAK) manifests as an insidiously progressive and debilitating form of granulomatous inflammation including the aorta and its major branches. The precise etiology of TAK remains elusive, with current understanding suggesting an autoimmune origin primarily driven by T cells. Notably, a growing body of evidence bears testimony to the widespread effects of B cells on disease pathogenesis and progression. Distinct alterations in peripheral B cell subsets have been described in individuals with TAK. Advancements in technology have facilitated the identification of novel autoantibodies in TAK. Moreover, emerging data suggest that dysregulated signaling cascades downstream of B cell receptor families, including interactions with innate pattern recognition receptors such as toll-like receptors, as well as co-stimulatory molecules like CD40, CD80 and CD86, may result in the selection and proliferation of autoreactive B cell clones in TAK. Additionally, ectopic lymphoid neogenesis within the aortic wall of TAK patients exhibits functional characteristics. In recent decades, therapeutic interventions targeting B cells, notably utilizing the anti-CD20 monoclonal antibody rituximab, have demonstrated efficacy in TAK. Despite the importance of the humoral immune response, a systematic understanding of how autoreactive B cells contribute to the pathogenic process is still lacking. This review provides a comprehensive overview of the biological significance of B cell-mediated autoimmunity in TAK pathogenesis, as well as insights into therapeutic strategies targeting the humoral response. Furthermore, it examines the roles of T-helper and T follicular helper cells in humoral immunity and their potential contributions to disease mechanisms. We believe that further identification of the pathogenic role of autoimmune B cells and the underlying regulation system will lead to deeper personalized management of TAK patients. We believe that further elucidation of the pathogenic role of autoimmune B cells and the underlying regulatory mechanisms holds promise for the development of personalized approaches to managing TAK patients. Full article
Show Figures

Figure 1

Figure 1
<p>Two pathways of naïve B cells into antibody-secreting cells. In the follicular response (<b>left</b>), activated B cells engage in interactions with Th cells and follicle dendritic cells to form GC in secondary lymphoid organs. Following iterative rounds of somatic hypermutation and antigen affinity-driven selection, resting naïve B cells differentiate into antibody secreting cells or switched memory B cells derived from the germinal center. Extrafollicular responses (<b>right</b>) emerge preceding the formation of germinal centers, displaying distinctive phenotypic and transcriptional profiles compared to GC B cells. In healthy individuals, TLR7 and IFN-γ induce resting naïve B cells to differentiate into activated counterparts, DN2 cells and antibody-secreting cells in an IL-21-dependent manner. Neither pathway is T cell-dependent. In particular, the extrafollicular response includes a T cell-independent pathway. In addition, both pathways have mainly been reported in systemic lupus erythematosus. In TAK, the pathogenic role of extrafollicular responses is unknown. Therefore, we have marked a question mark on extrafollicular responses. Th: T helper; FDC: follicle dendritic cell; Mø: macrophage; DN2: double negative 2 cells; Ab: antibody; TLR7: toll-like receptor 7; IFNγ: interferon gamma; IL21: interleukin 21; TAK: Takayasu’s arteritis; GC: germinal center.</p>
Full article ">Figure 2
<p>A profile of artery involvement in TAK. In the left part, the color gradient illustrates the typical frequency of arterial segment involvement in TAK, with a predilection for the brachiocephalic arteries, as well as the thoracic and abdominal arterial territories. The right part shows the profile of the peripheral blood and vascular wall of TAK. The pathological process of TAK initiates in the vasa vasorum of the adventitia and is marked by the rupture of elastic laminae and smooth muscle cell migration. Several immune cells including memory B cells, antigen-experienced B cells as well as Tfh cells infiltrate the adventitia. The granulomas are located in the medial layer, and TLOs are distributed deeper within the adventitial layer which involves a dense network of HEVs. TLO: tertiary lymphoid organ; HEV: high endothelial venule; DC: dendritic cell; RBC: red blood cell; Tfh: T follicular helper; TAK: Takayasu’s arteritis.</p>
Full article ">Figure 3
<p>Abnormal activation of B cell checkpoints in TAK. The activation of BCRs, TLRs and several co-stimulatory molecules (including CD40, CD80 and CD86) was documented in TAK. Serum APRIL and BAFF levels and cytokines related to humoral immunity, including IL2, IL4, IL6, IL9, IL21, IL23 and IFN-γ, exhibited enhanced levels in TAK patients compared with healthy individuals. IL-5 induces B cell development and Ig secretion, the role of which is unclear in TAK. The bottom half of the figure is the cytokines and their receptors that are involved in B cell activation. The top half of the figure includes BCRs, TLRs and several co-stimulatory molecules. IL: interleukin; IFNγ: interferon-gamma; R: receptor; BAFF: B cell activating factor; BCMA: B cell maturation antigen; APRIL: A proliferation-inducing ligand; TACI: transmembrane activator and calcium modulator and cyclophilin ligand interactor; BCR: B cell receptor; TLR: toll-like receptor; Ig: immunoglobulin; gp130: glycoprotein 130; TAK: Takayasu’s arteritis; PAMP: pathogen-associated molecular pattern; DAMP: damage-associated molecular patterns.</p>
Full article ">
10 pages, 1354 KiB  
Article
Optic Disc Drusen in Pseudoxanthoma Elasticum Are Associated with the Extent of Bruch’s Membrane Calcification
by Kristin Raming, Sandrine H. Künzel, Maximilian Pfau, Doris Hendig, Frank G. Holz and Kristina Pfau
J. Clin. Med. 2024, 13(12), 3395; https://doi.org/10.3390/jcm13123395 - 10 Jun 2024
Viewed by 599
Abstract
Background/Objectives: To assess the frequency, extent, localization and potential progression of optic disc drusen (ODD) and the correlation with the angioid streak (AS) length and retinal atrophy in patients with pseudoxanthoma elasticum (PXE). Methods: This retrospective study included patient data from [...] Read more.
Background/Objectives: To assess the frequency, extent, localization and potential progression of optic disc drusen (ODD) and the correlation with the angioid streak (AS) length and retinal atrophy in patients with pseudoxanthoma elasticum (PXE). Methods: This retrospective study included patient data from a dedicated PXE clinic at the Department of Ophthalmology, University of Bonn, Germany (observation period from February 2008 to July 2023). Two readers evaluated the presence, localization, and the extent of the ODD on fundus autofluorescence (FAF) imaging at baseline and the follow-up assessments. Additionally, we measured the length of the longest AS visible at baseline and follow-up and the area of atrophy at baseline, both on FAF. Results: A total of 150 eyes of 75 PXE patients (median age at baseline 51.8 years, IRQ 46.3; 57.5 years, 49 female) underwent retrospective analysis. At baseline, 23 of 75 patients exhibited ODD in a minimum of one eye, resulting in an ODD prevalence of 30.7% in our cohort of PXE patients. Among these, 14 patients showed monocular and 9 binocular ODD that were localized predominantly nasally (46.9%). During the observational period (mean 97.5 ± 44.7 months), only one patient developed de novo ODD in one eye and one other patient showed a progression in the size of the existing ODD. The group of patients with ODD had significantly longer ASs (median 7020 µm, IQR 4604; 9183, vs. AS length without ODD: median 4404 µm, IQR 3512; 5965, p < 0.001). No association with the size of the atrophy was found at baseline (p = 0.27). Conclusions: This study demonstrates a prevalence of ODD of 30.7%. ODD presence is associated with longer ASs (an indicator of the severity and extent of ocular Bruch’s membrane calcification), suggesting that ODD formation is tightly related to ectopic calcification—possibly secondary to calcification of the lamina cribrosa. Prospective studies investigating the impact of ODD (in conjunction with intraocular pressure) on visual function in PXE warrant consideration. Full article
(This article belongs to the Special Issue Advances in Ophthalmic Imaging)
Show Figures

Figure 1

Figure 1
<p>Image analysis: fundus autofluorescence (FAF) images of a 25-year-old (<b>A</b>–<b>C</b>) and a 28-year-old (<b>D</b>–<b>F</b>) PXE patient (both female). (<b>A</b>,<b>D</b>): Dashed line boxes: Snippet of the images shown enlarged in (<b>B</b>) and (<b>E</b>), respectively. (<b>B</b>,<b>E</b>): The white overlay shows the grading of ODD regarding their location. (<b>C</b>): White arrow indicates angioid streak length, (<b>F</b>): Directions of reference lengths if the angioid streak extended the FAF image.</p>
Full article ">Figure 2
<p>Characteristics of optic disc drusen (ODD) in eyes of patients with pseudoxanthoma elasticum (PXE). (<b>A</b>) ODD are present monocularly in 60.9% and binocularly in 39.1% in patients with ODD. (<b>B</b>) The majority of ODD are localized nasally. (<b>C</b>) Illustration of the extent of the ODD by the number of quadrants involved, which are widely distributed.</p>
Full article ">Figure 3
<p>Clinical examples of the progression of optic disc drusen (ODD). (<b>A</b>,<b>B</b>) show fundus autofluorescence (FAF) images in 2017 and 2022 of a 25-year-old female PXE patient with unilateral ODD, which increased in size (arrow). The longest AS visible stayed stable, but a new AS developed (marked by the two arrowheads). (<b>C</b>) illustrates the FAF of a 56-year-old female PXE patient with suspected ODD nasal superior in the right eye. These become visible on FAF t the follow-up visit 7.8 years later (see (<b>D</b>)), with additional ODD temporal-inferior. Furthermore, the atrophy progressed markedly in size.</p>
Full article ">Figure 4
<p>Association of optic disc drusen (ODD) with the angioid streak (AS) length. The ASs are significantly longer in the eyes with ODD (median 7020 µm, IQR 4604; 9183) than the eyes without ODD (median 4404 µm, IQR 3512; 5965, <span class="html-italic">p</span> &lt; 0.001), see (<b>A</b>). No association with age was found (<span class="html-italic">p</span> = 0.37). (<b>B</b>) Correlation between the AS lengths of the left and right eyes (<span class="html-italic">p</span> = 0.03). Notably, ODD are only visible beyond a distinct AS length of approximately 5000 µm in one eye.</p>
Full article ">Figure 5
<p>Fundus autofluorescence (FAF) imaging (<b>A</b>), visual field examination (<b>B</b>) and optical coherence tomography (OCT) B-scan (<b>C</b>) to illustrate the morphological and possible functional aspects of optic disc drusen (ODD).</p>
Full article ">
Back to TopTop