Nothing Special   »   [go: up one dir, main page]

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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (62)

Search Parameters:
Keywords = contractile ring

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 11843 KiB  
Article
Effects of a Rosmarinus officinalis L. Extract and Rosmarinic Acid in Improving Streptozotocin-Induced Aortic Tissue Damages in Rats
by Irina Ielciu, Gabriela Adriana Filip, Alexandra C. Sevastre-Berghian, Ioana Bâldea, Neli-Kinga Olah, Ramona Flavia Burtescu, Vlad Alexandru Toma, Remus Moldovan, Ilioara Oniga and Daniela Hanganu
Nutrients 2025, 17(1), 158; https://doi.org/10.3390/nu17010158 - 31 Dec 2024
Viewed by 709
Abstract
Background/Aim: Rosmarinus officinalis L. (R. officinalis) is an aromatic medicinal species with important nutraceutical potential, having rosmarinic acid (RA) as one of its main metabolites. The present study aims to evaluate the effects of an extract obtained from the leaves of [...] Read more.
Background/Aim: Rosmarinus officinalis L. (R. officinalis) is an aromatic medicinal species with important nutraceutical potential, having rosmarinic acid (RA) as one of its main metabolites. The present study aims to evaluate the effects of an extract obtained from the leaves of this species and of its main metabolite in improving the streptozotocin-induced damage of hearts and aorta of diabetic rats. Methods: The leaves of the species were used to obtain a hydroethanolic extract, which was analyzed using the LC/MS method. Diabetes mellitus was induced by intraperitoneal streptozotocin administration in rats. After two weeks, oxidative stress parameters were evaluated from the heart and aorta homogenates. NOS3, AMPK, and adiponectin levels were quantified using ELISA tests, and thoracic aorta rings were isolated for contractility evaluation in the organ bath. Phospho-NF-κB, NRF2, HIF1 alfa, iNOS, and glyceraldehyde 3-phosphate dehydrogenase (GAPDH) quantification were performed using the Western blot technique. Results: Carnosic acid, together with rosmarinic acid, were proven to be the main metabolites identified in the composition of the tested extract. Administration of the extract and of RA improved the relaxation response to acetylcholine and the redox status, with the reduction in malondialdehyde (MDA), nitric oxide synthase 3 (NOS 3), AMP-activated protein kinase (AMPK), adiponectin, reduced (GSH) and oxidized glutathione (GSSG) levels, and superoxide dismutase (SOD) activity. RA significantly enhanced the expression of HIF 1α, NRF2, and pNFkB in the heart. Conclusions: Administration of the R. officinalis extract and of RA-alleviated oxidative stress, proving vascular and cardiac antioxidant properties in the hearts and aorta of diabetic rats. Full article
(This article belongs to the Section Clinical Nutrition)
Show Figures

Figure 1

Figure 1
<p>Contractile responses of aortic rings to cumulative concentrations of phenylephrine (Phe, <b>left</b>) and relaxation responses of aortic rings to cumulative concentrations of acetylcholine (Ach, <b>right</b>). The contractile and relaxing responses were tested for the control group and groups with DM treated with the vehicle, <span class="html-italic">R. officinalis</span> extract (Ro), and RA. No statistically significant differences between groups were noticed.</p>
Full article ">Figure 2
<p>Malondialdehyde (MDA) levels in the aorta and heart of diabetic rats. MDA levels increased after STZ administration (# <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.05) and diminished after <span class="html-italic">R. officinalis</span> extract and RA (* <span class="html-italic">p</span> &lt; 0.05) administration. Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 3
<p>Superoxide dismutase (SOD) activity in the aorta and heart of diabetic rats. SOD activity decreased after STZ administration (### <span class="html-italic">p</span> &lt; 0.001 in the aorta and # <span class="html-italic">p</span> &lt; 0.05 in the heart) and increased after <span class="html-italic">R. officinalis</span> extract (** <span class="html-italic">p</span> &lt; 0.05) and RA (DM + RA vs DM + Ro, α, <span class="html-italic">p</span> &lt; 0.05) administration. Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 4
<p>The level of oxidized glutathione (GSSG) in the heart and aorta homogenates of animals with experimentally induced DM. GSSG levels increased after STZ administration but insignificantly reduced after <span class="html-italic">R. officinalis</span> extract (* <span class="html-italic">p</span> &lt; 0.05) administration. Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 5
<p>Nitric oxide synthase 3 (NOS 3) levels in the aorta and heart of animals with experimentally induced DM. NOS3 levels decreased after STZ administration (## <span class="html-italic">p</span> &lt; 0.01) and further decreased after <span class="html-italic">R. officinalis</span> extract administration (* <span class="html-italic">p</span> &lt; 0.05) and RA (<span class="html-italic">p</span> &lt; 0.05) administration. Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 6
<p>AMPK levels in aorta and heart homogenates of animals with experimentally induced DM. Levels of activated AMP protein kinase (AMPK) in the aorta and heart of diabetic rats, increased after STZ administration (##, ### <span class="html-italic">p</span> &lt; 0.05) and diminished after <span class="html-italic">R. officinalis</span> extract (*, *** <span class="html-italic">p</span> &lt; 0.001) and RA (*, *** <span class="html-italic">p</span> &lt; 0.05) administration. Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 7
<p>Adiponectin levels in heart and aorta homogenates in DM and rats with DM treated with <span class="html-italic">R. officinalis</span> and RA. Adiponectin levels decreased in the aorta and heart homogenates of DM rats (#, ## <span class="html-italic">p</span> &lt; 0.01) and after the administration of the <span class="html-italic">R. officinalis</span> extract and RA (*, ** <span class="html-italic">p</span> &lt; 0.05). Each group consisted of 9 rats. Results are expressed as mean ± SD.</p>
Full article ">Figure 8
<p>The effects of the <span class="html-italic">R. officinalis</span> extract and RA administration on the expression of HIF 1α, iNOS, NRF2, and pNF-κB in the heart. (<b>A</b>) Expression of HIF 1α, iNOS, NRF2, and pNF-κB was analyzed using Western blotting (WB). (<b>B</b>) HIF 1α, (<b>C</b>) iNOS, (<b>D</b>) NRF2, and (<b>E</b>) pNF-κB. Image analysis of Western blot bands was carried out using densitometry, and the results were normalized to GAPDH. * DM vs. DM + Ro/DM + RA; <sup>α</sup> DM + Ro vs. DM + RA. Results are expressed as mean (n = 3) ± SD; *<sup>, α</sup> <span class="html-italic">p</span> &lt; 0.05; <sup>##, αα</sup> <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 9
<p>Representative photomicrographs of the hearts of the four experimental groups. Scale bar = 25 µm, 400× magnification, staining with HE. (<b>a</b>) Control, (<b>b</b>) DM, (<b>c</b>) DM + <span class="html-italic">R. officinalis</span> extract, (<b>d</b>) DM + RA.</p>
Full article ">
18 pages, 6375 KiB  
Article
The Myosin-V Myo51 and Alpha-Actinin Ain1p Cooperate during Contractile Ring Assembly and Disassembly in Fission Yeast Cytokinesis
by Zoe L. Tyree, Kimberly Bellingham-Johnstun, Jessica Martinez-Baird and Caroline Laplante
J. Fungi 2024, 10(9), 647; https://doi.org/10.3390/jof10090647 - 12 Sep 2024
Viewed by 1050
Abstract
Cytokinesis is driven in part by the constriction of a ring of actin filaments, myosin motors and other proteins. In fission yeast, three myosins contribute to cytokinesis including a Myosin-V Myo51. As Myosin-Vs typically carry cargo along actin filaments, the role of Myo51 [...] Read more.
Cytokinesis is driven in part by the constriction of a ring of actin filaments, myosin motors and other proteins. In fission yeast, three myosins contribute to cytokinesis including a Myosin-V Myo51. As Myosin-Vs typically carry cargo along actin filaments, the role of Myo51 in cytokinesis remains unclear. The previous work suggests that Myo51 may crosslink actin filaments. We hypothesized that if Myo51 crosslinks actin filaments, cells carrying double deletions of ain1, which encodes the crosslinker alpha-actinin, and myo51 (∆ain1 ∆myo51 cells) will exhibit more severe cytokinesis phenotypes than cells with the single ∆ain1 mutation. Contrary to our expectations, we found that the loss of Myo51 in ∆ain1 cells partially rescued the severity of the node clumping phenotype measured in ∆ain1 cells. Furthermore, we describe a normal process of contractile ring “shedding”, the appearance of fragments of ring material extending away from the contractile ring along the ingressing septum that occurs in the second half of constriction. We measured that ∆ain1 ∆myo51 cells exhibit premature and exaggerated shedding. Our work suggests that Myo51 is not a simple actin filament crosslinker. Instead, a role in effective node motion better recapitulates its function during ring assembly and disassembly. Full article
(This article belongs to the Special Issue Yeast Cytokinesis)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">Δmyo51 Δain1</span> cells partially rescue node clumping phenotype seen in <span class="html-italic">∆ain1</span> cells. (<b>A</b>) Confocal micrograph (Fire LUT) of contractile rings in cells expressing mEGFP-Myo2p. Arrowheads, clumping starts. Asterisks, clumping ends. Dashed lines, cell outlines. (<b>B</b>) Representative kymographs aligned at constriction onset (inverted grayscale LUT) of constricting contractile rings from cells expressing mEGFP-Myo2p. Green arrowheads, clumping starts. Magenta arrowheads, clumping ends. (<b>C</b>) Outcomes plot of the timing of assembly completion (n = 38–46), onset of constriction (n = 34–42), clumping start (n = 37–43) and clumping end (n = 37–43) in cells expressing mEGFP-Myo2p. Significance determined by log rank Test and reported in <a href="#app1-jof-10-00647" class="html-app">Table S3</a>. (<b>D</b>) Stacked bar chart of the percentage of rings that display mEGFP-Myo2p clumping phenotype. Significance determined by Pearson’s chi-square test. (<b>E</b>) Swarm plot of the duration of clumping for cells expressing mEGFP-Myo2p. Bars, means. Asterisks, <span class="html-italic">p</span> &lt; 0.05 by 2-tailed Student’s <span class="html-italic">t</span> test. (<b>F</b>) Stacked bar chart of the percentage of cells expressing mEGFP-Myo2p that display mild (23% of <span class="html-italic">Δain1</span> cells, 38% of <span class="html-italic">Δain1 Δmyo51</span> cells among whole population) or severe (29% of <span class="html-italic">Δain1</span> cells, 5% of <span class="html-italic">Δain1 Δmyo51</span> cells among whole population) clumping phenotype. Asterisk, <span class="html-italic">p</span> &lt; 0.05 by Pearson’s chi-square test. (<b>G</b>) Swarm plot of the mean fluorescence intensity of the clumps for cells expressing mEGFP-Myo2p. Bars, means. Asterisk, <span class="html-italic">p</span> &lt; 0.05 by 2-tailed Student’s <span class="html-italic">t</span> test.</p>
Full article ">Figure 2
<p>Outer ring proteins “shed” from contractile ring in wild-type cells. (<b>A</b>) Model of a shedding ring (left). Inset confocal micrograph (inverted grayscale LUT) of a representative wild-type cell expressing mEGFP-Myo2p undergoing shedding (right). Magenta arrows, shedding fragments. Dashed lines, cell outlines. (<b>B</b>) Confocal micrograph (inverted grayscale LUT, top and Fire LUT, bottom) of cells expressing different fluorescently labeled proteins. Brackets, contractile rings. Dashed lines, cell outlines. (<b>C</b>) Timelapse confocal micrograph (inverted grayscale LUT, top and Fire LUT, bottom) of a cell expressing Cdc12p-3GFP imaged in a yeast motel. Arrows, shedding fragments. Dashed lines, cell outlines. (<b>D</b>) Timelapse confocal micrograph (inverted grayscale LUT, top and Fire LUT, bottom) of a cell expressing mCherry-Myp2p. Arrows, shedding fragments. Dashed lines, cell outlines. (<b>E</b>) Timelapse confocal micrograph of a cell (inverted grayscale LUT and colored merged) expressing Myo51p-3GFP and Cdc15p-mCherry. Dashed lines, cell outlines. (<b>F</b>) Representative SMLM images of shedding in wild-type cells color-coded for time (top, Jet LUT) or density (bottom, Hot LUT). Brackets, contractile rings. Arrows, shedding material along septum. Dashed lines, cell outlines. (<b>G</b>) Enlarged SMLM images of shedding in wild-type cells from (<b>F</b>), color-coded for time. Arrowheads, shedding material. Dashed line, cell outline.</p>
Full article ">Figure 3
<p>Contractile ring shedding is exaggerated and premature in <span class="html-italic">Δain1 Δmyo51</span> cells. (<b>A</b>) Representative kymographs of contractile rings aligned at constriction onset from cells expressing mEGFP-Myo2p (inverted grayscale LUT, left and Fire LUT, right). Arrows, shedding starts. (<b>B</b>) Timelapse of confocal micrographs (inverted grayscale LUT) for cells expressing mEGFP-Myo2p. Magenta asterisks, first depicted timepoint with visible shedding. Dashed lines, cell outlines. (<b>C</b>) Swarm plot for the percentage of contractile ring constriction when shedding begins for cells expressing mEGFP-Myo2p. Bars, means. Single asterisks, <span class="html-italic">p</span> &lt; 0.05 by 2-tailed Student’s <span class="html-italic">t</span> test. Double asterisks, <span class="html-italic">p</span> &lt; 0.0001 by 2-tailed Student’s <span class="html-italic">t</span> test. (<b>D</b>) Swarm plot of the duration of shedding for cells expressing mEGFP-Myo2p. Bars, means. Double asterisks, <span class="html-italic">p</span> &lt; 0.0001 by 2-tailed Student’s <span class="html-italic">t</span> test. (<b>E</b>) Outcomes plot showing the timing of the onset of shedding (n = 39–54) and the completion of ring disassembly (n = 30) in cells expressing mEGFP-Myo2p. Significance determined by log rank test and reported in <a href="#app1-jof-10-00647" class="html-app">Table S3</a>. (<b>F</b>) Stacked bar chart of the percentage of cells expressing mEGFP-Myo2p that display an exaggerated shedding phenotype. Single asterisk, <span class="html-italic">p</span> &lt; 0.05 by Pearson’s chi-square test. Double asterisks, <span class="html-italic">p</span> &lt; 0.0001 by Pearson’s chi-square test.</p>
Full article ">Figure 4
<p>Myo51p localization to the inner layer of the contractile ring is actin dependent. (<b>A</b>) Timelapse confocal micrographs (inverted grayscale LUT and colored merged images) of cells imaged in a yeast motel expressing different pairs of fluorescently labeled cytokinetic markers. Model of orientation and method of line scan (top right). Graphs (right) depicting fluorescence intensity for both fluorescent signals along the line for each individual micrograph. Dashed lines, cell outlines. Magenta boxes, time frames used for line scans. Dashed arrows, lines used for fluorescence intensity scan. Arrowhead points to Myo51p-3GFP peel. (<b>B</b>) Timelapse confocal micrograph (inverted grayscale LUT) of 2.5 µM LatA treated (left) and DMSO treated (right) cells. Dashed lines, cell outlines.</p>
Full article ">Figure 5
<p>Model of clumping and shedding in the contractile ring. (<b>A</b>) Model of assembling contractile ring within a fission yeast cell (left). Models of assembling contractile rings to demonstrate clustering of nodes in wild-type, <span class="html-italic">Δmyo51</span>, <span class="html-italic">Δain1</span> and <span class="html-italic">Δmyo51 Δain1</span> cells (right). Magenta circles, nodes. Arrows, node movements. (<b>B</b>) Model of constricting contractile ring within a fission yeast cell (left). Models of constricting contractile rings in upright orientation to demonstrate shedding of material in wild-type, <span class="html-italic">Δmyo51</span>, <span class="html-italic">Δain1</span> and <span class="html-italic">Δmyo51 Δain1</span> cells (right). Magenta circles, nodes. Dim magenta ovals, node material in shedding fragments. Arrows, node movements.</p>
Full article ">
13 pages, 7661 KiB  
Article
Low Calcium–High Magnesium Krebs–Henseleit Solution Combined with Adenosine and Lidocaine Improved Rat Aortic Function and Structure Following Cold Preservation
by Aryadi Arsyad, Geni K. R. Lembang, Sesilia L. Linda, Yulia Y. Djabir and Geoffrey P. Dobson
Medicina 2024, 60(8), 1284; https://doi.org/10.3390/medicina60081284 - 9 Aug 2024
Viewed by 1193
Abstract
Background and objectives: The main problem of vascular preservation is the maintenance of vessel graft quality and function following extended storage. Conventional preservation solutions such as histidine–tryptophan–ketoglutarate (HTK) solution, Phosphate-Buffer Solution (PBS), or sodium chloride 0.9% has been shown to be inadequate in [...] Read more.
Background and objectives: The main problem of vascular preservation is the maintenance of vessel graft quality and function following extended storage. Conventional preservation solutions such as histidine–tryptophan–ketoglutarate (HTK) solution, Phosphate-Buffer Solution (PBS), or sodium chloride 0.9% has been shown to be inadequate in preserving vascular physiological function after 3 days of cold storage. This study aimed to evaluate whether adenosine and lidocaine (AL) in a modified Krebs–Henseleit (KH) solution can preserve the function and histological structure of rat aortic rings after 6 days. Materials and Methods: Thirty-five aortic rings from male Wistar rats (200–300 g) were harvested and immediately immersed in one of the assigned cold preservation solutions: standard KH, modified KH (mod KH) with lower calcium (Ca2+) and higher magnesium content (Mg2+) with or without adenosine and lidocaine (mod KH-AL), and modified KH with AL, insulin, and melatonin (Mod KH-ALMI). The contraction and relaxation function of the aortic rings were examined using an isometric force transducer after 6 days of cold preservation. Hematoxylin and eosin staining were used to analyze the rings’ histological structure. Results: Vascular contraction and relaxation functions were severely affected after a 6-day cold storage period in standard KH. Modifying the KH solution by reducing the Ca2+ and increasing the Mg2+ levels greatly recovered the vessel functions. The addition of AL or ALMI to the modified KH did not further recover vascular contractility. However, only the addition of AL to the modified KH increased the ACh-induced relaxation at 6 days when compared to the conventional KH, suggesting that endothelium preservation is improved. From histological analysis, it was found that the addition of AL but not ALMI further improved the endothelial lining and the structure of the elastic membrane layers of the preserved vessels after 6 days of cold preservation. Conclusions: The addition of AL to low calcium-high magnesium KH solution significantly enhanced endothelial preservation and improved endothelial-induced relaxation of preserved vessels after 6 days of cold storage. Full article
(This article belongs to the Section Hematology and Immunology)
Show Figures

Figure 1

Figure 1
<p>Representative vascular sections with degeneration scores of 0 (<b>A</b>), 1 (<b>B</b>), and 2 (<b>C</b>).</p>
Full article ">Figure 2
<p>Isolated rat aortic ring contraction after 6 days of cold preservation compared to the fresh preparation (control). Response to potassium chloride (KCl) (<b>A</b>) and norepinephrine (NE) (<b>B</b>). The values represent the mean ± S.E.M of the aortic ring contraction. * <span class="html-italic">p</span> &lt; 0.05 between the standard KH and control groups.</p>
Full article ">Figure 3
<p>Isolated rat aortic ring percent relaxation after 6 days of cold preservation. Response to acetylcholine (<b>A</b>) and nitric oxide donor (<b>B</b>) compared to the control. Relaxation is expressed as the percent of relaxation to 100 μM papaverine. The points represent the mean ± S.E.M of the aortic ring relaxation. * <span class="html-italic">p</span> &lt; 0.05 between the control and standard KH groups. # <span class="html-italic">p</span> &lt; 0.05 between the standard KH and modified KH−AL groups. ^ <span class="html-italic">p</span> &lt; 0.05 between the modified KH and modified KH−AL and KH−ALMI groups. ~ <span class="html-italic">p</span> &lt; 0.05 between the KH and the other groups.</p>
Full article ">Figure 4
<p>Concentration–response curve for SNP (NO-induced relaxation) after 6-day storage compared to control. (<b>A</b>) Acetylcholine-induced maximal relaxation, and (<b>B</b>) SNP-induced maximal relaxation. Relaxation is expressed as the percent of the control group maximal relaxation. The values are expressed as the mean ± S.E.M of the aortic ring relaxation. * <span class="html-italic">p</span> &lt; 0.05, statistical difference between the KH and the control group. # <span class="html-italic">p</span> &lt; 0.05, the KH group statistically differs from the modified KH + AL groups. ~ <span class="html-italic">p</span> &lt; 0.05 between the KH and the other groups.</p>
Full article ">Figure 5
<p>The representative of the hematoxylin and eosin section of freshly harvested aorta (<b>A</b>) and after 6-day preservation in the KH (<b>B</b>), modified KH (<b>C</b>), modified KH-AL (<b>D</b>), and modified KH-ALMI solution (<b>E</b>) in different microscopic magnifications. The black arrows show the endothelial cells; the red arrowheads show the denudation of the endothelium; EM: elastic membrane; SMCN: smooth muscle cell nucleus.</p>
Full article ">
16 pages, 2873 KiB  
Article
2-Amino-N-Phenethylbenzamides for Irritable Bowel Syndrome Treatment
by Miglena Milusheva, Mihaela Stoyanova, Vera Gledacheva, Iliyana Stefanova, Mina Todorova, Mina Pencheva, Kirila Stojnova, Slava Tsoneva, Paraskev Nedialkov and Stoyanka Nikolova
Molecules 2024, 29(14), 3375; https://doi.org/10.3390/molecules29143375 - 18 Jul 2024
Cited by 1 | Viewed by 1308
Abstract
Irritable bowel syndrome (IBS) is a common gastrointestinal (GI) disorder characterized by abdominal pain or discomfort. Mebeverine is an antispasmodic that has been widely used in clinical practice to relieve the symptoms of IBS. However, its systemic use usually leads to side effects. [...] Read more.
Irritable bowel syndrome (IBS) is a common gastrointestinal (GI) disorder characterized by abdominal pain or discomfort. Mebeverine is an antispasmodic that has been widely used in clinical practice to relieve the symptoms of IBS. However, its systemic use usually leads to side effects. Therefore, the current paper aimed to synthesize more effective medicines for IBS treatment. We used ring opening of isatoic anhydride for the synthesis in reaction with 2-phenylethylamine. In silico simulation predicted spasmolytic activity for 2-amino-N-phenethylbenzamides. The newly synthesized compounds demonstrated a relaxation effect similar to mebeverine but did not affect the serotonin or Ca2+-dependent signaling pathway of contractile activity (CA) in contrast. Having in mind the anti-inflammatory potential of antispasmodics, the synthesized molecules were tested in vitro and ex vivo for their anti-inflammatory effects. Four of the newly synthesized compounds demonstrated very good activity by preventing albumin denaturation compared to anti-inflammatory drugs/agents well-established in medicinal practice. The newly synthesized compounds also inhibited the expression of interleukin-1β and stimulated the expression of neuronal nitric oxide synthase (nNOS), and, consequently, nitric oxide (NO) synthesis by neurons of the myenteric plexus. This characterizes the newly synthesized compounds as biologically active relaxants, offering a cleaner and more precise application in pharmacological practice, thereby enhancing their potential therapeutic value. Full article
(This article belongs to the Special Issue Design, Synthesis and Evaluation of Small Molecule Drugs)
Show Figures

Figure 1

Figure 1
<p>Changes in SM tissue parameters: (<b>A</b>) tone in mN; and (<b>B</b>) amplitude in %, induced by compounds <b>3</b> and <b>4a</b>–<b>d</b> (5 × 10<sup>−5</sup> M) (<span class="html-italic">n</span> = 12). Statistically significant differences: * <span class="html-italic">p</span> &lt; 0.05 comparison is between mebeverine and <b>3</b>, <b>4a</b>–<b>d</b>.</p>
Full article ">Figure 1 Cont.
<p>Changes in SM tissue parameters: (<b>A</b>) tone in mN; and (<b>B</b>) amplitude in %, induced by compounds <b>3</b> and <b>4a</b>–<b>d</b> (5 × 10<sup>−5</sup> M) (<span class="html-italic">n</span> = 12). Statistically significant differences: * <span class="html-italic">p</span> &lt; 0.05 comparison is between mebeverine and <b>3</b>, <b>4a</b>–<b>d</b>.</p>
Full article ">Figure 2
<p>Representative tracings demonstrating the contractile responses of SMPs from the gastric corpus of rats following the application of mebeverine compared to the application of substances <b>4a</b> and <b>4c</b>, subsequently followed by an ACh reaction in their background.</p>
Full article ">Figure 3
<p>IC<sub>50</sub> inhibition of albumin denaturation (in mg/mL) of newly synthesized hybrids (yellow). Diclofenac (red) and acetylsalicylic acid (green) were used as standards. The lower the value, the higher the anti-inflammatory activity.</p>
Full article ">Figure 4
<p>Micrographs of SM stomach corpus preparations with newly synthesized compounds (the incubation period was 3 h): (<b>A</b>) control, IL-1β expression (black arrow), at ×400; (<b>B</b>,<b>C</b>) SMPs incubated with substance <b>3</b>, <b>4a</b> expression of IL-1β (black arrow), at ×400; (<b>D</b>) SMPs incubated with substance <b>4c</b>, lack of IL-1β expression in myenteric plexus, weak in mucosal glands, at ×400; (<b>E</b>) SMPs incubated with substance <b>4b</b>, lack of IL-1β expression, at ×400; (<b>F</b>) control, nNOS expression (orange arrow) at ×400; (<b>G</b>,<b>H</b>) SMPs incubated with substances <b>3</b> and <b>4a</b>, lack of nNOS expression, at ×400; and (<b>I</b>,<b>J</b>) SMPs incubated with substances <b>4b</b> and <b>4c</b>, increased expression of nNOS in the myenteric plexus (orange arrow), at ×400.</p>
Full article ">Figure 5
<p>Expression of IL-1β in the SMPs incubated with the newly synthesized compounds <b>3</b> and <b>4.</b> The asterisk indicates significant differences between groups—*** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>Expression of nNOS in the SMPs incubated with the newly synthesized compounds <b>3</b> and <b>4</b>. The asterisk indicates significant differences between groups—*** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Scheme 1
<p>Synthesis of the 2-amino-N-phenethylbenzamides <b>3</b> and <b>4a</b>–<b>d</b>.</p>
Full article ">
14 pages, 5662 KiB  
Article
The Mechanosensitive Pkd2 Channel Modulates the Recruitment of Myosin II and Actin to the Cytokinetic Contractile Ring
by Pritha Chowdhury, Debatrayee Sinha, Abhishek Poddar, Madhurya Chetluru and Qian Chen
J. Fungi 2024, 10(7), 455; https://doi.org/10.3390/jof10070455 - 28 Jun 2024
Cited by 1 | Viewed by 1241
Abstract
Cytokinesis, the last step in cell division, separates daughter cells through mechanical force. This is often through the force produced by an actomyosin contractile ring. In fission yeast cells, the ring helps recruit a mechanosensitive ion channel, Pkd2, to the cleavage furrow, whose [...] Read more.
Cytokinesis, the last step in cell division, separates daughter cells through mechanical force. This is often through the force produced by an actomyosin contractile ring. In fission yeast cells, the ring helps recruit a mechanosensitive ion channel, Pkd2, to the cleavage furrow, whose activation by membrane tension promotes calcium influx and daughter cell separation. However, it is unclear how the activities of Pkd2 may affect the actomyosin ring. Here, through both microscopic and genetic analyses of a hypomorphic pkd2 mutant, we examined the potential role of this essential gene in assembling the contractile ring. The pkd2-81KD mutation significantly increased the counts of the type II myosin heavy chain Myo2 (+18%), its regulatory light chain Rlc1 (+37%) and actin (+100%) molecules in the ring, compared to the wild type. Consistent with a regulatory role of Pkd2 in the ring assembly, we identified a strong negative genetic interaction between pkd2-81KD and the temperature-sensitive mutant myo2-E1. The pkd2-81KD myo2-E1 cells often failed to assemble a complete contractile ring. We conclude that Pkd2 modulates the recruitment of type II myosin and actin to the contractile ring, suggesting a novel calcium-dependent mechanism regulating the actin cytoskeletal structures during cytokinesis. Full article
(This article belongs to the Special Issue Yeast Cytokinesis)
Show Figures

Figure 1

Figure 1
<p>The <span class="html-italic">pkd2-81KD</span> mutant increased the recruitment of myosin II heavy chain Myo2 to the contractile ring. (<b>A</b>) Micrographs of wild type and <span class="html-italic">pkd2-81KD</span> cells expressing GFP-Myo2. White arrow: cytokinetic nodes; White arrowhead: the contractile ring. (<b>B</b>) Dot plot of the average intracellular fluorescence intensity of GFP-Myo2 in a cell. (<b>C</b>) Top: anti-GFP blot of the lysate from wild type and <span class="html-italic">pkd2-81KD</span> cells expressing GFP-Myo2. Number indicates the normalized intensity of GFP-Myo2 band ± standard deviations. Bottom: Coomassie blue stained SDS-PAGE gel of respective lysates. Representative data from three biological repeats are shown. (<b>D</b>) Top: a diagram of the contractile ring assembly, maturation and constriction in a fission yeast cell. Bottom: time series of the equatorial plane of dividing cells expressing GFP-Myo2. Dashed box: start of the contractile ring constriction. Number: time in minutes. (<b>E</b>) Time course of the fluorescence intensity of GFP-Myo2 at the equatorial plane after the start of contractile ring assembly (time zero). Cloud: standard deviations. Interval = 2 min. (<b>F</b>) Dot plot of the fluorescence intensity of GFP-Myo2 in a mature contractile ring just before it starts to constrict. **: <span class="html-italic">p</span> &lt; 0.01. NS: not significant. Statistics were calculated using two-tailed Student’s <span class="html-italic">t</span> tests. All the data are pooled from at least two independent biological repeats. Scale bar = 5 µm.</p>
Full article ">Figure 2
<p>The <span class="html-italic">pkd2-81KD</span> mutant increased the recruitment of myosin II regulatory light chain Rlc1 to the contractile ring. (<b>A</b>) Micrographs of <span class="html-italic">wild type</span> and <span class="html-italic">pkd2-81KD</span> cells expressing Rlc1-tdTomato. White arrowhead: the contractile ring. (<b>B</b>) Dot plot of the intracellular fluorescence intensity of Rlc1-tdTomato in a cell. (<b>C</b>) Time series of the equatorial plane of dividing cells expressing Rlc1-tdTomato. Dashed box: start of the contractile ring constriction. Number indicates time in minutes. (<b>D</b>) Time course of the fluorescence intensity of Rlc1-tdTomato in the equatorial plane. Cloud: standard deviations. (<b>E</b>) Dot plot of the fluorescence intensity of Rlc1-tdTomato in a mature contractile ring. **: <span class="html-italic">p</span> &lt; 0.01. ***: <span class="html-italic">p</span> &lt; 0.001. Statistics were calculated using two-tailed Student’s <span class="html-italic">t</span>-tests. All the data are pooled from at least two independent biological repeats. Scale bar = 5 µm.</p>
Full article ">Figure 3
<p>The <span class="html-italic">pkd2</span>-<span class="html-italic">81KD</span> mutation increased the assembly of actin filaments in the contractile ring. (<b>A</b>) Micrographs of the <span class="html-italic">wild type</span> (top) and <span class="html-italic">pkd2-81KD</span> (bottom) cells co-expressing GFP-Lifeact (green) and Rlc1-mCherry (magenta). White arrowhead: the contractile ring. (<b>B</b>) Dot plot of the average intracellular fluorescence intensity of GFP-Lifeact in a cell. (<b>C</b>) Time series of a <span class="html-italic">wild type</span> and a <span class="html-italic">pkd2-81KD</span> cell co-expressing GFP-Lifeact (green) and Rlc1-mCherry. Number: Time in minutes from the start of the contractile ring assembly based on the appearance of cytokinetic nodes marked by Rlc1-mCherry. (<b>D</b>) Dot plot of the total fluorescence of GFP-Lifeact in a mature contractile ring. ***: <span class="html-italic">p</span> &lt; 0.001. Statistics were calculated using two-tailed Student’s <span class="html-italic">t</span> tests. All the data are pooled from three independent biological repeats. Bar = 5 µm.</p>
Full article ">Figure 4
<p>Negative genetic interactions between <span class="html-italic">pkd2</span> and <span class="html-italic">myo2</span> mutants. (<b>A</b>) Ten-fold dilution series of yeast cells at the indicated temperatures. (<b>B</b>) Bright-field micrographs of live cells at 25 °C. Red arrowhead: lysed cells. (<b>C</b>) Micrographs of calcofluor-stained fixed cells at 25 °C. Arrow: abnormal septum. Arrowhead: thick septum (n &gt; 500). (<b>D</b>) A table summarizing the morphological defects of the <span class="html-italic">pkd2-81KD myo2-E1</span> mutant cells. All the data are pooled from at least two independent biological repeats. *: <span class="html-italic">p</span> &lt; 0.05; **: <span class="html-italic">p</span> &lt; 0.01. Statistics were calculated using two-tailed Student’s <span class="html-italic">t</span> tests. Scale bar = 5 µm.</p>
Full article ">Figure 5
<p>The <span class="html-italic">pkd2-81KD myo2-E1</span> mutant failed to assemble a complete contractile ring at the permissive temperature. (<b>A</b>) Micrographs of live cells expressing Rlc1-tdTomato. Arrowhead: the contractile ring. (<b>B</b>) Dot plot of average intracellular fluorescence intensity of Rlc1-tdTomato in a cell. (<b>C</b>) Time-lapse series of the equatorial plane of a <span class="html-italic">myo2-E1</span> and a <span class="html-italic">pkd2-81KD myo2-E1</span> cell expressing Rlc1-tdTomato. Dashed box: start of contractile ring constriction. Number: time in minutes. (<b>D</b>) Time course of the fluorescence intensity of Rlc1-tdTomato at the equatorial plane. Interval = 2 min. (<b>E</b>) Dot plot of average fluorescence intensity of Rlc1-tdTomato in a mature contractile ring before it starts to constrict. (<b>F</b>) Dot plot of the rate of the contractile ring constriction. All the experiments were carried out at the permissive temperature of 25 °C. ***: <span class="html-italic">p</span> &lt; 0.001. NS: not significant. Statistics were calculated using two-tailed Student’s <span class="html-italic">t</span> tests. All the data are pooled from at least two independent biological repeats. Scale bar = 5 µm.</p>
Full article ">
12 pages, 2032 KiB  
Article
Myogenic Anti-Nucleolin Aptamer iSN04 Inhibits Proliferation and Promotes Differentiation of Vascular Smooth Muscle Cells
by Mana Miyoshi, Takeshi Shimosato and Tomohide Takaya
Biomolecules 2024, 14(6), 709; https://doi.org/10.3390/biom14060709 - 15 Jun 2024
Cited by 1 | Viewed by 1829
Abstract
De-differentiation and subsequent increased proliferation and inflammation of vascular smooth muscle cells (VSMCs) is one of the mechanisms of atherogenesis. Maintaining VSMCs in a contractile differentiated state is therefore a promising therapeutic strategy for atherosclerosis. We have reported the 18-base myogenetic oligodeoxynucleotide, iSN04, [...] Read more.
De-differentiation and subsequent increased proliferation and inflammation of vascular smooth muscle cells (VSMCs) is one of the mechanisms of atherogenesis. Maintaining VSMCs in a contractile differentiated state is therefore a promising therapeutic strategy for atherosclerosis. We have reported the 18-base myogenetic oligodeoxynucleotide, iSN04, which serves as an anti-nucleolin aptamer and promotes skeletal and myocardial differentiation. The present study investigated the effect of iSN04 on VSMCs because nucleolin has been reported to contribute to VSMC de-differentiation under pathophysiological conditions. Nucleolin is localized in the nucleoplasm and nucleoli of both rat and human VSMCs. iSN04 without a carrier was spontaneously incorporated into VSMCs, indicating that iSN04 would serve as an anti-nucleolin aptamer. iSN04 treatment decreased the ratio of 5-ethynyl-2′-deoxyuridine (EdU)-positive proliferating VSMCs and increased the expression of α-smooth muscle actin, a contractile marker of VSMCs. iSN04 also suppressed angiogenesis of mouse aortic rings ex vivo, which is a model of pathological angiogenesis involved in plaque formation, growth, and rupture. These results demonstrate that antagonizing nucleolin with iSN04 preserves VSMC differentiation, providing a nucleic acid drug candidate for the treatment of vascular disease. Full article
Show Figures

Figure 1

Figure 1
<p>Nucleolin localization and iSN04 incorporation in A10 cells. (<b>A</b>) Representative fluorescence images of nucleolin staining of A10 cells in GM (day 0) and DM with or without 10 μM iSN04 (day 4). Scale bar, 50 μm. (<b>B</b>) Representative fluorescence images of A10 cells treated with 5 μg/mL 6-FAM-iSN04 in GM. Scale bar, 50 μm. 6-FAM, 6-carboxyfluorescein; DAPI, 4′,6-diamidino-2-phenylindole; DM, differentiation medium; GM, growth medium.</p>
Full article ">Figure 2
<p>The effect of iSN04 on proliferation and differentiation of A10 cells. (<b>A</b>) Representative fluorescence images of EdU staining of A10 cells pre-treated with 30 μM iSN04 in GM for 24 h and then with 10 μM EdU in GM for 3 h. Scale bar, 200 μm. The ratio of EdU<sup>+</sup> cells was quantified. * <span class="html-italic">p</span> &lt; 0.05 vs. control (Student’s <span class="html-italic">t</span>-test). <span class="html-italic">n</span> = 4. (<b>B</b>) Representative fluorescence images of α-SMA staining of A10 cells treated with 3 or 10 μM iSN04 in DM for 4 days. Scale bar, 200 μm. α-SMA signal intensities per number of DAPI<sup>+</sup> nuclei were quantified. ** <span class="html-italic">p</span> &lt; 0.01 vs. control; <sup>††</sup> <span class="html-italic">p</span> &lt; 0.01 vs. 3 μM iSN04 (Scheffe’s <span class="html-italic">F</span> test). <span class="html-italic">n</span> = 7. DAPI, 4′,6-diamidino-2-phenylindole; DM, differentiation medium; EdU, 5-ethynyl-2′-deoxyuridine; GM, growth medium; α-SMA, α-smooth muscle actin.</p>
Full article ">Figure 3
<p>The effect of iSN04 on proliferation and differentiation of hAoSMCs. (<b>A</b>) Representative fluorescence images of nucleolin staining of hAoSMCs in hGM (day 0) and hDM (day 4). Scale bar, 50 μm. (<b>B</b>) Representative fluorescence images of EdU staining of hAoSMCs pre-treated with 30 μM iSN04 in hGM for 48 h and then with 10 μM EdU in hGM for 12 h. Scale bar, 200 μm. The ratio of EdU<sup>+</sup> cells were quantified. ** <span class="html-italic">p</span> &lt; 0.01 vs. control (Student’s <span class="html-italic">t</span>-test). <span class="html-italic">n</span> = 4. (<b>C</b>) qPCR results of a cell-cycle marker gene, Ki-67 (<span class="html-italic">MKI67</span>), and contractile SMC marker genes, α-SMA (<span class="html-italic">ACTA2</span>), SM22α (<span class="html-italic">TAGLN</span>), and caldesmon (<span class="html-italic">CALD1</span>), in hAoSMCs treated with 30 μM iSN04 in hGM for 48 h. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01 vs. control (Student’s <span class="html-italic">t</span>-test). <span class="html-italic">n</span> = 3. DAPI, 4′,6-diamidino-2-phenylindole; DM, differentiation medium; EdU, 5-ethynyl-2′-deoxyuridine; GM, growth medium; hAoSMC, human aortic smooth muscle cell; hDM, differentiation medium for human aortic smooth muscle cell; hGM, growth medium for human aortic smooth muscle cell; qPCR, quantitative real-time RT-PCR; SMC, smooth muscle cell.</p>
Full article ">Figure 4
<p>iSN04 suppresses angiogenesis of aortic rings. Representative microscopic images of iSN04-treated mouse aortic rings. Aortas from 9-week-old C57BL/6J mice were embedded in type I-A collagen gel, serum starved for 24 h, and treated with 10 or 30 μM iSN04 in GM for 6 days. Neovascular sprouting area was quantified as the difference between the total area (contiguous outline of neovascular ends) and the aortic area (circumference of the aortic ring). Scale bar, 200 μm. ** <span class="html-italic">p</span> &lt; 0.01 vs. control (Scheffe’s <span class="html-italic">F</span> test). <span class="html-italic">n</span> = 5–6. GM, growth medium.</p>
Full article ">
16 pages, 2317 KiB  
Article
Chamaecyparis lawsoniana and Its Active Compound Quercetin as Ca2+ Inhibitors in the Contraction of Airway Smooth Muscle
by Edgar Flores-Soto, Bianca S. Romero-Martínez, Héctor Solís-Chagoyán, Edgar A. Estrella-Parra, Jose G. Avila-Acevedo, Juan C. Gomez-Verjan, Jorge Reyes-García, María F. Casas-Hernández, Bettina Sommer and Luis M. Montaño
Molecules 2024, 29(10), 2284; https://doi.org/10.3390/molecules29102284 - 12 May 2024
Cited by 1 | Viewed by 1494
Abstract
The Cupressaceae family includes species considered to be medicinal. Their essential oil is used for headaches, colds, cough, and bronchitis. Cedar trees like Chamaecyparis lawsoniana (C. lawsoniana) are commonly found in urban areas. We investigated whether C. lawsoniana exerts some of [...] Read more.
The Cupressaceae family includes species considered to be medicinal. Their essential oil is used for headaches, colds, cough, and bronchitis. Cedar trees like Chamaecyparis lawsoniana (C. lawsoniana) are commonly found in urban areas. We investigated whether C. lawsoniana exerts some of its effects by modifying airway smooth muscle (ASM) contractility. The leaves of C. lawsoniana (363 g) were pulverized mechanically, and extracts were obtained by successive maceration 1:10 (w:w) with methanol/CHCl3. Guinea pig tracheal rings were contracted with KCl, tetraethylammonium (TEA), histamine (HIS), or carbachol (Cch) in organ baths. In the Cch experiments, tissues were pre-incubated with D-600, an antagonist of L-type voltage-dependent Ca2+ channels (L-VDCC) before the addition of C. lawsoniana. Interestingly, at different concentrations, C. lawsoniana diminished the tracheal contractions induced by KCl, TEA, HIS, and Cch. In ASM cells, C. lawsoniana significantly diminished L-type Ca2+ currents. ASM cells stimulated with Cch produced a transient Ca2+ peak followed by a sustained plateau maintained by L-VDCC and store-operated Ca2+ channels (SOCC). C. lawsoniana almost abolished this last response. These results show that C. lawsoniana, and its active metabolite quercetin, relax the ASM by inhibiting the L-VDCC and SOCC; further studies must be performed to obtain the complete set of metabolites of the extract and study at length their pharmacological properties. Full article
(This article belongs to the Special Issue Advances in Natural Products and Their Biological Activities)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">C. lawsoniana</span> methanolic extract concentration-dependently diminishes KCl and tetraethylammonium (TEA)-induced contraction in guinea pig tracheal smooth muscle. (<b>A</b>) The upper panel shows original traces of the effects of different <span class="html-italic">C. lawsoniana</span> (CL) concentrations (37.5, 75, and 150 µg/mL) on the 60 mM KCl-induced contraction. Below, bar graph illustrating the significant diminution in KCl-induced tension produced by each CL concentration tested (<span class="html-italic">n</span> = 7). (<b>B</b>) Original recordings of the CL effects on the TEA (10 mM, <span class="html-italic">n</span> = 5–6) induced contraction. Bar graph depicts the significances reached for CL effect at 75 µg/mL and 150 µg/mL. Bars represent mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span>&lt; 0.01. Results are expressed as a percentage of the third KCl response.</p>
Full article ">Figure 2
<p><span class="html-italic">C. lawsoniana</span> methanolic extract diminishes histamine-induced contractions in guinea pig airway smooth muscle. The left panel illustrates original traces of the effects of different <span class="html-italic">C. lawsoniana</span> (CL) concentrations (9.37, 18.75, 37.5, 75, and 150 µg/mL) on the histamine (HIS, 10 µM, <span class="html-italic">n</span> = 6–7) induced contraction. On the right, bar graph depicting the significance reached at all CL concentrations used. Bars represent mean ± SEM, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Store-operated Ca<sup>2+</sup> channels (SOCC) might be blocked by the methanolic extract of <span class="html-italic">C. lawsoniana</span> (CL). (<b>A</b>) The upper panel shows original traces of smooth muscle contraction induced by Cch 1 µM and the effect of different CL concentrations (18.75, 37.5, 75, 150, and 250 µg/mL) (<span class="html-italic">n</span> = 6). The bar graph summarizes the data analysis and significant differences observed for CL 75, 150, and 250 µg/mL. (<b>B</b>) Original traces of smooth muscle contraction induced by Cch 1 µM and the relaxation developed by D-600 (30 µM) used to block L-type voltage-dependent Ca<sup>2+</sup> channels (L-VDCC) and the addition of CL (18.75, 37.5, 75, 150, and 250 µg/mL) (<span class="html-italic">n</span> = 6). Note that D-600 caused a new plateau that was almost abolished by the different CL concentrations tested, indicating a possible inhibition of store-operated Ca<sup>2+</sup> channels (SOCC). The bar graph in the right panel illustrates the relaxation induced by D-600 alone and after the addition of each CL concentration after D-600. Bars represent mean values ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>Voltage-dependent L-type Ca<sup>2+</sup> currents, but not K<sup>+</sup> currents, were diminished by the <span class="html-italic">C. lawsoniana</span> (CL) methanolic extract in tracheal myocytes. (<b>A</b>) Cultured guinea pig tracheal myocytes were held at a membrane potential of − 60 mV, and then depolarizing pulses were applied in 10 mV steps to +50 mV. This caused voltage-dependent Ba<sup>2+</sup> inward currents (IBa<sup>2+</sup>) corresponding to L-type Ca<sup>2+</sup> currents, as 1 µM nifedipine abolished them. The peak inward current reached its maximum amplitude at 0 mV. When multiple concentrations of CL extract (<span class="html-italic">n</span> = 5–7) were perfused, these currents decreased, reaching significance compared to the control current. The original recordings for each concentration tested are shown as insets. The symbols indicate the mean ± SEM, where * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the control group. (<b>B</b>) The cultured cells received a step depolarization protocol from −60 to +50 mV in 10 mV increments from a holding potential of −60 mV during 500 ms. These stimulations generated a voltage-dependent outward K<sup>+</sup> current (IK<sup>+</sup>). Myocytes perfused with the methanolic extract of <span class="html-italic">C. lawsoniana</span> (CL, 150 μg/mL) showed no changes in IK<sup>+</sup> compared to the control group (<span class="html-italic">n</span> = 3). The insets in the figure show the original traces of IK<sup>+</sup>.</p>
Full article ">Figure 5
<p>Both L-type voltage-dependent Ca<sup>2+</sup> channels (L-VDCC) and store-operated Ca<sup>2+</sup> channels (SOCC) were blocked by the methanolic extract of <span class="html-italic">C. lawsoniana</span> (CL) in guinea pig tracheal myocytes. (<b>A</b>) Once stimulated with carbachol (Cch), myocytes show an intracellular Ca<sup>2+</sup> peak followed by a plateau. The inset shows that this Ca<sup>2+</sup> plateau is sustained by L-VDCC, blocked by D-600 (30 µM), and SOCC, blocked by 2-APB (100 µM). (<b>B</b>) Cumulative addition of CL concentrations lowered the Cch-induced Ca<sup>2+</sup> plateau (<span class="html-italic">n</span> = 5), indicating that this extract blocks both L-VDCC and SOCC in tracheal myocytes. (<b>C</b>) Bar graph illustrating CL effects on the Cch-induced Ca<sup>2+</sup> plateau. Bars represent mean ± SEM, * <span class="html-italic">p</span>&lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>Phytochemical identification of quercetin from the MeOH extract of <span class="html-italic">C. lawsoniana.</span> Chromatograms for the standard (<b>A</b>) and MeOH extract of <span class="html-italic">C. lawsoniana</span> (<b>C</b>). Insets (<b>B</b>,<b>D</b>) illustrate the UV absorbance of the quercetin standard (<b>B</b>) and <span class="html-italic">C. lawsoniana</span> MeOH extract, respectively. Analysis revealed that the methanolic extract contained 0.019 mg (with a standard deviation of ±1.5209 × 10<sup>−6</sup>) of quercetin per 1 mg of the crude methanol extract. Additionally, the UV absorbance of quercetin in the <span class="html-italic">C. lawsoniana</span> methanolic extract, with a maximum wavelength (λmax) of 254.0 and 368.0 [MeOH], closely resembled that of the standard quercetin, which had a λmax of 255.0 and 369.0 [MeOH].</p>
Full article ">Figure 7
<p>The contractions induced by either KCl or Cch in airway smooth muscle were relaxed by quercetin. (<b>A</b>,<b>B</b>) Quercetin, in a concentration-dependent manner, inhibited the contraction induced by KCl or Cch, respectively, in tracheal rings (<span class="html-italic">n</span> = 5). Bars represent mean ± SEM, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Quercetin blocks the carbachol-induced Ca<sup>2+</sup> plateau. (<b>A</b>) In tracheal myocytes, carbachol (Cch) stimulation induced an intracellular Ca<sup>2+</sup> peak followed by a plateau. The cumulative curve of quercetin (QC) diminished the Cch-induced plateau. (<b>B</b>) Bar graph illustrating statistical significance at 50 and 100 µM QC (<span class="html-italic">n</span> = 5). Bars represent mean ± SEM, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
20 pages, 3165 KiB  
Article
Essential Role of COPII Proteins in Maintaining the Contractile Ring Anchoring to the Plasma Membrane during Cytokinesis in Drosophila Male Meiosis
by Yoshiki Matsuura, Kana Kaizuka and Yoshihiro H. Inoue
Int. J. Mol. Sci. 2024, 25(8), 4526; https://doi.org/10.3390/ijms25084526 - 20 Apr 2024
Viewed by 1732
Abstract
Coatomer Protein Complex-II (COPII) mediates anterograde vesicle transport from the endoplasmic reticulum (ER) to the Golgi apparatus. Here, we report that the COPII coatomer complex is constructed dependent on a small GTPase, Sar1, in spermatocytes before and during Drosophila male meiosis. COPII-containing foci [...] Read more.
Coatomer Protein Complex-II (COPII) mediates anterograde vesicle transport from the endoplasmic reticulum (ER) to the Golgi apparatus. Here, we report that the COPII coatomer complex is constructed dependent on a small GTPase, Sar1, in spermatocytes before and during Drosophila male meiosis. COPII-containing foci co-localized with transitional endoplasmic reticulum (tER)-Golgi units. They showed dynamic distribution along astral microtubules and accumulated around the spindle pole, but they were not localized on the cleavage furrow (CF) sites. The depletion of the four COPII coatomer subunits, Sec16, or Sar1 that regulate COPII assembly resulted in multinucleated cell production after meiosis, suggesting that cytokinesis failed in both or either of the meiotic divisions. Although contractile actomyosin and anilloseptin rings were formed once plasma membrane ingression was initiated, they were frequently removed from the plasma membrane during furrowing. We explored the factors conveyed toward the CF sites in the membrane via COPII-mediated vesicles. DE-cadherin-containing vesicles were formed depending on Sar1 and were accumulated in the cleavage sites. Furthermore, COPII depletion inhibited de novo plasma membrane insertion. These findings suggest that COPII vesicles supply the factors essential for the anchoring and/or constriction of the contractile rings at cleavage sites during male meiosis in Drosophila. Full article
(This article belongs to the Special Issue Cell Division: A Focus on Molecular Mechanisms)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Phase-contrast images of spermatids derived from spermatocytes harboring the silencing of mRNAs for components consisting of COPII coat and regulators for its assembly. (<b>A</b>–<b>G</b>) Phase-contrast images of living spermatids at onion stage. (<b>A</b>) Control (<span class="html-italic">bam&gt;+</span>) spermatids. Each cell contains a single nucleus (white) and a single Nebenkern, which is a mitochondrial aggregate (black) in a 1:1 ratio. The arrowheads indicate multinucleate cells harboring more than two nuclei and single Nebenkerns. The arrows indicate large cells in which two multinucleated cells have fused. (<b>B</b>) Spermatids that developed from <span class="html-italic">Sar1RNAi</span> spermatocytes (<span class="html-italic">bam&gt;Sar1RNAi</span>). They possessed four or more than four nuclei with single Nebenkerns. (<b>C</b>) Spermatids derived from <span class="html-italic">Sec23RNAi</span> spermatocytes (<span class="html-italic">bam&gt;Dcr-2</span>, <span class="html-italic">Sec23RNAi</span>). (<b>D</b>) Multinucleate spermatids derived from <span class="html-italic">Sec24CDRNAi</span> spermatocytes (<span class="html-italic">bam&gt;Dcr-2</span>, <span class="html-italic">Sec24CDRNAi</span>). An arrow indicates a large cell in which two multinucleated cells have fused. (<b>E</b>) Spermatids derived from <span class="html-italic">Sec13RNAi</span> spermatocytes (<span class="html-italic">bam&gt;Dcr-2</span>, <span class="html-italic">Sec13RNAi</span>). (<b>F</b>) Spermatids derived from <span class="html-italic">Sec31RNAi</span> spermatocytes (<span class="html-italic">bam&gt;Sec31RNAi</span>). (<b>G</b>) Spermatids derived from <span class="html-italic">Sec16RNAi</span> spermatocytes (<span class="html-italic">bam&gt;Sec16RNAi</span>). Bar, 10 µm. (<b>H</b>) Frequencies of control spermatids (single nuclei) and multinucleated spermatids (harboring two to more than four nuclei) at onion stage in the testes containing spermatocyte-specific silencing of COPII-related six genes. The bars represent the SEM, n &gt; 107.</p>
Full article ">Figure 2
<p>Co-localization and close association of two COPII components in pre-meiotic and meiotic spermatocytes. (<b>A</b>–<b>D</b>) Observation of spermatocytes expressing both RFP-Sec13 and Sec31-GFP before (<b>A</b>) and during meiosis I (<b>B</b>–<b>D</b>). The RFP fluorescence (red in (<b>A</b>–<b>D</b>), white in (<b>A</b>’–<b>D</b>’)). GFP fluorescence (green in (<b>A</b>–<b>D</b>) and (<b>A</b>’’–<b>D</b>’’)). DNA staining with DAPI (blue in (<b>A</b>–<b>D</b>) and (<b>A</b>’’’–<b>D</b>’’’). (<b>A</b>–<b>A</b>’’’) Spermatocyte at interphase. Yellow foci indicate co-localization of Sec13 and Sec31 (arrowheads). (<b>B</b>–<b>B</b>’’’) Spermatocyte at late anaphase. (<b>C</b>–<b>C</b>’’’) Spermatocyte at early telophase. (<b>D</b>–<b>D</b>’’’) Spermatocyte at mid-telophase. Scale bar: 10 µm. (<b>E</b>,<b>F</b>) In situ PLA signals (red in (<b>E</b>,<b>F</b>) and (<b>E</b>’,<b>F</b>’)) of normal spermatocytes expressing RFP-Sec13 (green in (<b>E</b>,<b>F</b>) and in (<b>E</b>”,<b>F</b>”)) to detect close association of Sec23 with Sec13. DNA staining with DAPI (blue in (<b>E</b>–<b>H</b>) and (<b>E</b>’’’–<b>H</b>’’’). (<b>E</b>) In situ PLA of the spermatocytes with both anti-Sec23 and anti-RFP antibodies, with magnified image of area shown in the inset. (<b>F</b>) Negative control for in situ PLA of cells with anti-RFP antibody. Few PLA signals appear in the spermatocytes. Scale bar: 10 μm. (<b>G</b>,<b>H</b>) Anti-Sec16 immunostaining (green in (<b>G</b>,<b>H</b>), white in (<b>G</b>’’,<b>H</b>’’)) of spermatocytes expressing RFP-Sec13 (red in (<b>G</b>,<b>H</b>) and (<b>G</b>’,<b>H</b>’)) in normal control (<b>G</b>) and <span class="html-italic">Sar1RNAi</span> spermatocyte (<b>H</b>) at anaphase I to telophase I. DNA staining with DAPI (blue in (<b>A</b>–<b>D</b>) and (<b>A</b>’’–<b>D</b>’’). Arrowheads indicate accumulation of COPII-containing foci in two cytoplasmic bands running from top to bottom of telophase cell. Bars: 10 μm. (<b>I</b>,<b>J</b>) Number (<b>I</b>) and size (<b>J</b>) of Sec13-positive foci in spermatocytes before meiosis (Interphase) and those undergoing meiosis I (meiosis). Control (<span class="html-italic">bam&gt;RFP-Sec13</span>) (n = 29 cells) and <span class="html-italic">Sar1RNAi</span> (<span class="html-italic">bam&gt;RFP-Sec13</span>, <span class="html-italic">Sar1RNAi</span>) (n = 40 cells). (<b>K</b>) Number of Sec13 foci in spermatocytes at telophase I. Control (<span class="html-italic">bam&gt;RFP-Sec13</span>) (n = 44 cells) and <span class="html-italic">Sar1RNAi</span> cells (<span class="html-italic">bam&gt;RFP-Sec13</span>, <span class="html-italic">Sar1RNAi</span>) (n = 51 cells). Bars represent the SEM. **** <span class="html-italic">p</span> &lt; 0.0001 (Welch’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>Time-lapse imaging of plasma membrane ingression during late anaphase I to the end of cytokinesis in male meiosis I. (<b>A</b>,<b>B</b>) Time-lapse observation of the plasma membrane labeled with GFP in meiosis I cells expressing <span class="html-italic">GFP-PLC</span>γ-<span class="html-italic">PH</span>. The observation started from the time when the ingression of the plasma membrane was initiated at the cell equator (t = 0′). (<b>A</b>) control cell (<span class="html-italic">bam&gt;PLC</span>γ-<span class="html-italic">PH-GFP</span>). The membrane ingression terminates at 24 min after the start at CF sites. (<b>B</b>) The <span class="html-italic">Sar1</span>-silenced cells (<span class="html-italic">bam&gt;PLC</span>γ-<span class="html-italic">PH-GFP</span>, <span class="html-italic">Sar1RNAi</span>). The ingression initiated at the cell equator of the silencing cell, terminates at the middle (t = 31′), and returns to its original state (t = 47′). Bar: 10 µm. (<b>C</b>,<b>D</b>) Time-lapse alteration in the distance between the edges of the CFs in control (<span class="html-italic">bam&gt;+</span>) (<b>C</b>) and <span class="html-italic">Sar1</span>-silenced (<span class="html-italic">bam&gt;Sar1RNAi</span>) (<b>D</b>) spermatocytes from late anaphase to the end of meiosis I. The recording was terminated at the point when the furrowing was completed or when no more changes in the cell diameter were observed in a control cell (<span class="html-italic">bam&gt;PLC</span>γ-<span class="html-italic">PH-GFP</span>) or the <span class="html-italic">Sar1</span>-silenced cells (<span class="html-italic">bam&gt;PLC</span>γ-<span class="html-italic">PH-GFP</span>, <span class="html-italic">Sar1RNAi</span>) (n = 5). The length of each cell is represented by a different color.</p>
Full article ">Figure 4
<p>Abnormal localization of contractile ring components in <span class="html-italic">Sar1</span>-silenced spermatocytes at later stages of meiosis I. (<b>A</b>–<b>H</b>) Observation of the CRs using fluorescence-tagged CR proteins (Ring) in spermatocytes at late anaphase I to telophase I. (<b>A</b>,<b>E</b>) Immunostaining of the cells expressing GFP-MLC (green in (<b>A</b>,<b>A</b>’’,<b>E</b>,<b>E</b>’’)) with anti-Myosin Heavy Chain (MHC) antibody (red in (<b>A</b>,<b>A</b>’,<b>E</b>,<b>E</b>’)). (<b>B</b>,<b>F</b>) Fluorescence of the cells stained with Rhodamine-conjugated phalloidin to visualize F-actin (red in (<b>B</b>,<b>B</b>’,<b>F</b>,<b>F</b>’)). (<b>C</b>,<b>G</b>) Fluorescence of the cells expressing Septin 1-GFP (green in (<b>C</b>,<b>C</b>’,<b>G</b>,<b>G</b>’)). (<b>D</b>,<b>H</b>) Fluorescence of the cells expressing RFP-Anillin (red in (<b>D</b>,<b>D</b>’,<b>H</b>,<b>H</b>’)). (<b>A</b>–<b>D</b>) Normal control cells (<span class="html-italic">bam&gt;+</span>). (<b>E</b>–<b>H</b>) <span class="html-italic">Sar1</span>-silenced cells (<span class="html-italic">bam&gt;Sar1RNAi</span>). DNA staining with DAPI (blue in (<b>A</b>–<b>H</b>) and (<b>A</b>’’’, <b>B</b>”–<b>D</b>”, <b>E</b>’’’, <b>F</b>”–<b>H</b>”). (<b>I</b>,<b>J</b>) Time-lapse observation of contractile ring (CR) formation in the spermatocytes expressing MLC-GFP at anaphase I to cytokinesis in male meiosis. Control (<b>I</b>) and <span class="html-italic">Sar1</span>-silenced spermatocytes (<b>J</b>) were selected for visualization. In control cells, the MLC was accumulated at presumptive CF sites on the plasma membrane immediately before ingression (t = 0′), and the membrane ingression completed 15 min after the initiation of ingression. (<b>J</b>) In <span class="html-italic">Sar1</span>-silenced cells, the CR is constructed as in control cells (t = 36′). Thereafter, it was removed from the CF site on the lower plasma membrane, and the ingression returned to the initial state (t = 32′). The time-lapse observation was initiated at late anaphase I in both cells when the fluorescence of MLC-GFP became clearer (t = 0′) in both genotypes. (<b>K</b>–<b>N</b>) Time-lapse observation of the Septin1 CR (<b>K</b>,<b>L</b>) and the Anillin ring (<b>M</b>,<b>N</b>) from anaphase I just before the plasma membrane ingression at the cleavage furrow sites (white in Septin1-GFP) (t = 0 min) to the end of cytokinesis. Normal control spermatocyte (<b>K</b>,<b>M</b>) and <span class="html-italic">Sar1RNAi</span> spermatocyte (<b>L</b>,<b>N</b>). (<b>O</b>,<b>P</b>) Frequencies of the spermatocytes harboring an abnormal Anillin ring (<b>O</b>) or Septin1 ring (<b>P</b>) at late telophase I spermatocytes from control (<span class="html-italic">bam&gt;+</span>) and <span class="html-italic">Sar1</span>-silenced (<span class="html-italic">bam&gt;Sar1RNAi</span>) spermatocytes. The bars represent the SEM. Black bars: the telophase I cells harboring normal CRs. Gray bars: the telophase I cells harboring abnormally shaped CRs. Bars, 10 µm.</p>
Full article ">Figure 5
<p><span class="html-italic">Sar1</span>-dependent formation and distribution of DE-cad-GFP foci during the later stages of meiosis. (<b>A</b>–<b>D</b>) Observation of DE-cad-containing vesicles in the control (<b>A</b>,<b>B</b>) and <span class="html-italic">Sar1</span>-silenced (<b>C</b>,<b>D</b>) spermatocytes expressing DE-cad-GFP before meiosis (<b>A</b>,<b>C</b>) and at telophase I (<b>B</b>,<b>D</b>). DE-cad (green in (<b>A</b>–<b>D</b>) and (<b>A</b>’–<b>D</b>’)). DNA staining (blue in (<b>A</b>–<b>D</b>) and (<b>A</b>’’–<b>D</b>’’)). (<b>C</b>,<b>D</b>) Overexposed images. (<b>E</b>) Time-lapse observation of GFP fluorescence in living spermatocytes expressing GFP-DE-cad (white in (<b>E</b>)) during the later stages of meiosis I. As meiosis progresses, DE-cad-containing vesicles, indicated by arrowheads (t = 0′), migrated toward another vesicle at the cell equator (t = 24′–27′). (<b>F</b>) Time-lapse observation of DE-cad foci in living <span class="html-italic">Sar1</span>-silenced spermatocyte. According to the phase contrast micrographs of the cells (<b>E</b>’,<b>F</b>’), the cell margins are encircled by dotted lines. Arrows indicate a migration of the same vesicle in the periphery of a right spindle pole toward another vesicle at the cell equator as meiosis progresses. Note that few of the DE-cad vesicles were contained in the <span class="html-italic">Sar1</span>-silenced cell except for a weak and constant signal on the astral and spindle envelopes. (<b>G</b>) Average numbers of DE-cad-GFP foci in control (<span class="html-italic">bam&gt;+</span>, <span class="html-italic">DE-cad-GFP</span>) (n = 58) and <span class="html-italic">Sar1</span>-silenced spermatocytes undergoing meiosis I (<span class="html-italic">bam&gt;Sar1RNAi</span>, <span class="html-italic">DE-cad-GFP</span>) (n = 62). The bars represent the SEM. Significance was tested between control cells and the <span class="html-italic">Sar1</span>-silenced cells in meiosis I. * <span class="html-italic">p</span> &lt; 0.05 (Welch’s <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>A loss of the plasma membrane newly added in the cleavage furrow by wheat germ agglutinin (WGA) in <span class="html-italic">Sar1</span>-silenced spermatocytes at telophase I. (<b>A</b>,<b>B</b>) A visualization of the plasma membrane stained with fluorescence-tagged WGA of the spermatocytes expressing RFP-tagged Anillin. The RFP-Anillin indicates the location of the CR in control (<span class="html-italic">bam&gt;RFP-Anillin</span>) (<b>A</b>) and in <span class="html-italic">Sar1</span>-silenced cells (<span class="html-italic">bam&gt;RFP-Sar1RNAi</span>, <span class="html-italic">RFP-Anillin</span>) (<b>B</b>) At telophase I. Note that the WGA signal is invisible in the cell midzone except for a weaker signal along the plasma membrane in the silenced cells, while the intense signal is mainly localized around the CR in control cells. WGA staining (green in (<b>A</b>,<b>B</b>), white in (<b>A</b>’,<b>B</b>’)), RFP-Anillin (red in (<b>A</b>,<b>A</b>’’,<b>B</b>,<b>B</b>’’)), and DNA (blue in (<b>A</b>,<b>A</b>’’’,<b>B</b>,<b>B</b>’’’)). (<b>C</b>) Frequencies of cells exhibiting no or reduced WGA signals (gray bars) and distinctive signals (black bars) in the midzone among the telophase I cells in control and the <span class="html-italic">Sar1</span>-silenced cells (n = 36 telophase I cells in total in both control and the silenced cells). The bars represent the SEM.</p>
Full article ">
16 pages, 2262 KiB  
Article
The Effect of an Elevated Dietary Copper Level on the Vascular Contractility and Oxidative Stress in Middle-Aged Rats
by Klaudia Kitala-Tańska, Katarzyna Socha, Jerzy Juśkiewicz, Magdalena Krajewska-Włodarczyk and Michał Majewski
Nutrients 2024, 16(8), 1172; https://doi.org/10.3390/nu16081172 - 15 Apr 2024
Cited by 3 | Viewed by 1483
Abstract
Copper (Cu), being an essential mineral, plays a crucial role in maintaining physiological homeostasis across multiple bodily systems, notably the cardiovascular system. However, an increased Cu level in the body may cause blood vessel dysfunction and oxidative stress, which is unfavorable for the [...] Read more.
Copper (Cu), being an essential mineral, plays a crucial role in maintaining physiological homeostasis across multiple bodily systems, notably the cardiovascular system. However, an increased Cu level in the body may cause blood vessel dysfunction and oxidative stress, which is unfavorable for the cardiovascular system. Middle-aged (7–8 months old) male Wistar rats (n/group = 12) received a diet supplemented with 6.45 mg Cu/kg (100% of the recommended daily dietary quantity of copper) for 8 weeks (Group A). The experimental group received 12.9 mg Cu/kg of diet (200%—Group B). An ex vivo study revealed that supplementation with 200% Cu decreased the contraction of isolated aortic rings to noradrenaline (0.7-fold) through FP receptor modulation. Vasodilation to sodium nitroprusside (1.10-fold) and acetylcholine (1.13-fold) was potentiated due to the increased net effect of prostacyclin derived from cyclooxygenase-1. Nitric oxide (NO, 2.08-fold), superoxide anion (O2•−, 1.5-fold), and hydrogen peroxide (H2O2, 2.33-fold) measured in the aortic rings increased. Blood serum antioxidant status (TAS, 1.6-fold), Cu (1.2-fold), Zn (1.1-fold), and the Cu/Zn ratio (1.4-fold) increased. An increase in Cu (1.12-fold) and the Cu/Zn ratio (1.09-fold) was also seen in the rats’ livers. Meanwhile, cyclooxygenase-1 (0.7-fold), cyclooxygenase-2 (0.4-fold) and glyceraldehyde 3-phosphate dehydrogenase (0.5-fold) decreased. Moreover, a negative correlation between Cu and Zn was found (r = −0.80) in rat serum. Supplementation with 200% Cu did not modify the isolated heart functioning. No significant difference was found in the body weight, fat/lean body ratio, and organ weight for either the heart or liver, spleen, kidney, and brain. Neither Fe nor Se, the Cu/Se ratio, the Se/Zn ratio (in serum and liver), heme oxygenase-1 (HO-1), endothelial nitric oxide synthase (eNOS), or intercellular adhesion molecule-1 (iCAM-1) (in serum) were modified. Supplementation with 200% of Cu potentiated pro-oxidant status and modified vascular contractility in middle-aged rats. Full article
(This article belongs to the Section Micronutrients and Human Health)
Show Figures

Figure 1

Figure 1
<p>Vascular contraction to noradrenaline (NA, 0.1 µM) in the isolated thoracic arteries dissected from rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) Cu/kg of diet for 8 weeks. Aortic rings were studied under the control conditions (<b>a</b>) or after preincubation with the selective cyclooxygenase-2 (COX-2) inhibitor (NS-398, 10 µM) (<b>b</b>), the EP and DP receptor antagonist (AH-6809, 30 µM) (<b>c</b>), the prostacyclin (PGI2) synthesis inhibitor (tranylcypromine, 10 µM) (<b>d</b>), the selective FP receptor antagonist (AL-8810, 10 µM) (<b>e</b>). Values are means ± SD, <span class="html-italic">p</span> ≤ 0.05 (<span class="html-italic">t</span>-test).</p>
Full article ">Figure 2
<p>Vascular contraction to noradrenaline (NA, 0.1 µM) in the isolated thoracic arteries dissected from rats supplemented with 6.45 mg (100%—Group A) (<b>a</b>) and 12.9 mg (200%—Group B) (<b>b</b>) Cu/kg of diet for 8 weeks. Values are means ± SD, <span class="html-italic">p</span> ≤ 0.05 (two-way ANOVA with Tukey’s multiple comparisons test).</p>
Full article ">Figure 3
<p>Vasodilation to cumulative concentrations of acetylcholine (ACh, 0.1 nM−10 µM) (<b>a</b>) and sodium nitroprusside (SNP, 0.1 nM−10 µM) (<b>b</b>) in the isolated thoracic arteries dissected from rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) Cu/kg of diet for 8 weeks. Values are means ± SEM, n = 12, * vs. control diet (100% copper), <span class="html-italic">p</span> ≤ 0.05 (two-way ANOVA with Šídák’s multiple comparisons test).</p>
Full article ">Figure 4
<p>The cumulative concentration–response curves to acetylcholine (ACh, 0.1 nM–10 µM) in the isolated thoracic arteries dissected from rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) Cu/kg of diet for 8 weeks. Aortic rings were preincubated (30 min) with the selective cyclooxygenase-2 (COX-2) inhibitor (NS-398, 10 µM) (<b>a</b>), the EP and DP receptor antagonist (AH-6809, 30 µM) (<b>b</b>), the prostacyclin (PGI2) synthesis inhibitor (tranylcypromine, 10 µM) (<b>c</b>), the selective FP receptor antagonist (AL-8810, 10 µM) (<b>d</b>). Values are means ± SEM, n varies, * vs. control conditions, <span class="html-italic">p</span> ≤ 0.05 (two-way ANOVA with Šídák’s multiple comparisons test). CC—control conditions.</p>
Full article ">Figure 5
<p>Total antioxidant status (TAS) (<b>a</b>), copper (Cu) (<b>b</b>), zinc (Zn) (<b>c</b>) and Cu/Zn molar ratio (<b>d</b>) measured in the blood serum of experimental rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) copper/kg of diet for 8 weeks. Values are means ± SD, n varies, <span class="html-italic">p</span> ≤ 0.05 (<span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>Correlation matrix. Pearson’s r correlation. Rats were supplemented with 6.45 mg (100%—Group A) (<b>a</b>) and 12.9 mg (200%—Group B) (<b>b</b>) Cu/kg of diet for 8 weeks. Positive correlation was found between TAS and Fe in both studied groups. Negative correlation was observed between Cu and Zn in the 200% copper group. Cu—copper, Zn—zinc.</p>
Full article ">Figure 7
<p>Copper (Cu) (<b>a</b>), and Cu/Zn molar ratio (<b>b</b>) measured in the liver of experimental rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) Cu/kg of diet for 8 weeks. Values are means ± SD, n varies, <span class="html-italic">p</span> ≤ 0.05 (<span class="html-italic">t</span>-test).</p>
Full article ">Figure 8
<p>ELISA study. COX-1 (<b>a</b>), COX-2 (<b>b</b>) and GAPDH (<b>c</b>) measured in the blood serum of experimental rats supplemented with 6.45 mg (100%—Group A) and 12.9 mg (200%—Group B) Cu/kg of diet for 8 weeks. Values are means ± SD, n varies, <span class="html-italic">p</span> ≤ 0.05 (<span class="html-italic">t</span>-test).</p>
Full article ">
13 pages, 1509 KiB  
Article
Spironolactone Induces Vasodilation by Endothelium-Dependent Mechanisms Involving NO and by Endothelium-Independent Mechanisms Blocking Ca2+ Channels
by Margarida Lorigo, João Amaro and Elisa Cairrao
J. Xenobiot. 2024, 14(1), 320-332; https://doi.org/10.3390/jox14010020 - 1 Mar 2024
Viewed by 1982
Abstract
Background: Spironolactone (SPI) is a diuretic widely used to treat cardiovascular diseases (CVD) and is non-specific for mineralocorticoid receptors (MR) and with an affinity for progesterone (PR) and androgen (AR) receptors. Since 2009, it has been suggested that pharmaceuticals are emerging contaminants (called [...] Read more.
Background: Spironolactone (SPI) is a diuretic widely used to treat cardiovascular diseases (CVD) and is non-specific for mineralocorticoid receptors (MR) and with an affinity for progesterone (PR) and androgen (AR) receptors. Since 2009, it has been suggested that pharmaceuticals are emerging contaminants (called EDC), and recently, it was reported that most EDC are AR and MR antagonists and estrogen receptors (ER) agonists. Concerning SPI, endocrine-disrupting effects were observed in female western mosquitofish, but there are still no data regarding the SPI effects as a possible human EDC. Methods: In this work, aortic rings were used to analyze the contractility effects of SPI and the mode of action concerning the involvement of Ca2+ channels and endothelial pathways. Moreover, cytotoxic effects were analyzed by MTT assays. Results: SPI induces vasodilation in the rat aorta by endothelium-dependent mechanisms involving NO and by endothelium-independent mechanisms blocking Ca2+ channels. Moreover, a non-monotonic effect characteristic of EDC was observed for SPI-induced decrease in cell viability. Conclusions: Our findings suggest that SPI may act as an EDC at a human level. However, ex vivo studies with human arteries should be carried out to better understand this drug’s implications for human health and future generations. Full article
Show Figures

Figure 1

Figure 1
<p>Cell viability of A7r5 cells under the effect of SPI (spironolactone, 0.0001–1000 μmol/L) (n = 12). Data are presented as a percentage (%) of sample absorbance on absorbances induced by control. Each bar represents mean values and vertical lines the S.D. of the mean. * (asterisk) represents differences between SPI and vehicle (ethanol 1%) (*** <span class="html-italic">p</span> &lt; 0.001) (<span class="html-italic">p</span> &lt; 0.05, one-way ANOVA followed by Dunnett’s post hoc tests).</p>
Full article ">Figure 2
<p>Vasorelaxant effects induced by Nif (nifedipine, 1 and 0.01 µmol/L) plus SPI (spironolactone, 0.001–100 μmol/L) on endothelium (E)-intact and -denuded aorta rings pre-contracted with (<b>a</b>) NA (noradrenaline, 1 μmol/L, n = 16), (<b>b</b>) PE (phenylephrine, 1 μmol/L, n = 10), and (<b>c</b>) KCl (potassium chloride, 60 mmol/L, n = 10). Data are presented as a percentage (%) of relaxation on maximal contraction induced by vasoactive agents. Each bar (left panel) or point (right panel) represents mean values and vertical lines the S.D. of the mean. * (asterisk) represents differences versus control (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001), and <sup>#</sup> (cardinal) shows significant differences between endothelium-denuded versus endothelium-intact (<sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001) (<span class="html-italic">p</span> &lt; 0.05, two-way ANOVA with interaction followed by Bonferroni’s post hoc <span class="html-italic">t</span>-tests).</p>
Full article ">Figure 3
<p>Maximum tensions produced by (<b>a</b>) NA (noradrenaline, 1 μmol/L, n = 28) and (<b>b</b>) PE (phenylephrine, 1 μmol/L, n = 24) on endothelium-denuded and -intact rings and in the presence of the inhibitors L-NAME (N(ω)-nitro-L-arginine methyl ester, an eNOS inhibitor, 100 µmol/L) and Indo (indomethacin, a non-selective COX inhibitor, 10 µmol/L). The values are expressed as tension (mean) ± S.D. (mN). Dispersion points represent each value. * (asterisk) represent statistical differences between conditions tested (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001); two-way ANOVA followed by Bonferroni’s <span class="html-italic">t</span>-tests).</p>
Full article ">Figure 4
<p>Vasorelaxant effects induced by SPI (spironolactone, 0.001–100 μmol/L) on endothelium-intact and -denuded aorta rings pre-contracted with (<b>a</b>) NA (noradrenaline, 1 μmol/L, n = 6) and (<b>b</b>) PE (phenylephrine, 1 μmol/L, n = 5) and in the presence of the inhibitors L-NAME (N(ω)-nitro-L-arginine methyl ester, an eNOS inhibitor, 100 µmol/L) and Indo (indomethacin, a non-selective COX inhibitor, 10 µmol/L). Data are presented as a percentage (%) of relaxation on maximal contraction induced by vasoactive agents. Each point represents mean values, and vertical lines are the S.D. of the mean. * (asterisk) represents differences between SPI vs. L-NAME + SPI in endothelium-intact rings (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001), and <sup>#</sup> (cardinal) shows significant differences between SPI vs. Indo + SPI in endothelium-intact rings (<sup>#</sup> <span class="html-italic">p</span> &lt; 0.05; <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01; <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001), (<span class="html-italic">p</span> &lt; 0.05, two-way ANOVA with interaction followed by Bonferroni’s post hoc <span class="html-italic">t</span>-tests).</p>
Full article ">
13 pages, 6059 KiB  
Article
Morphological Changes of 3T3 Cells under Simulated Microgravity
by Minh Thi Tran, Chi Nguyen Quynh Ho, Son Nghia Hoang, Chung Chinh Doan, Minh Thai Nguyen, Huy Duc Van, Cang Ngoc Ly, Cuong Phan Minh Le, Huy Nghia Quang Hoang, Han Thai Minh Nguyen, Han Thi Truong, Quan Minh To, Tram Thi Thuy Nguyen and Long Thanh Le
Cells 2024, 13(4), 344; https://doi.org/10.3390/cells13040344 - 15 Feb 2024
Cited by 3 | Viewed by 2462
Abstract
Background: Cells are sensitive to changes in gravity, especially the cytoskeletal structures that determine cell morphology. The aim of this study was to assess the effects of simulated microgravity (SMG) on 3T3 cell morphology, as demonstrated by a characterization of the morphology of [...] Read more.
Background: Cells are sensitive to changes in gravity, especially the cytoskeletal structures that determine cell morphology. The aim of this study was to assess the effects of simulated microgravity (SMG) on 3T3 cell morphology, as demonstrated by a characterization of the morphology of cells and nuclei, alterations of microfilaments and microtubules, and changes in cycle progression. Methods: 3T3 cells underwent induced SMG for 72 h with Gravite®, while the control group was under 1G. Fluorescent staining was applied to estimate the morphology of cells and nuclei and the cytoskeleton distribution of 3T3 cells. Cell cycle progression was assessed by using the cell cycle app of the Cytell microscope, and Western blot was conducted to determine the expression of the major structural proteins and main cell cycle regulators. Results: The results show that SMG led to decreased nuclear intensity, nuclear area, and nuclear shape and increased cell diameter in 3T3 cells. The 3T3 cells in the SMG group appeared to have a flat form and diminished microvillus formation, while cells in the control group displayed an apical shape and abundant microvilli. The 3T3 cells under SMG exhibited microtubule distribution surrounding the nucleus, compared to the perinuclear accumulation in control cells. Irregular forms of the contractile ring and polar spindle were observed in 3T3 cells under SMG. The changes in cytoskeleton structure were caused by alterations in the expression of major cytoskeletal proteins, including β-actin and α-tubulin 3. Moreover, SMG induced 3T3 cells into the arrest phase by reducing main cell cycle related genes, which also affected the formation of cytoskeleton structures such as microfilaments and microtubules. Conclusions: These results reveal that SMG generated morphological changes in 3T3 cells by remodeling the cytoskeleton structure and downregulating major structural proteins and cell cycle regulators. Full article
Show Figures

Figure 1

Figure 1
<p>Proliferation of 3T3 cells in control and SMG groups. (<b>A</b>,<b>B</b>) Cell morphology of 3T3 cells in control and SMG groups. (<b>C</b>,<b>D</b>) FCS values for control and SMG groups (<span class="html-italic">n</span> = 5). (<b>E</b>) Gravite<sup>®</sup> operation in CO<sub>2</sub> incubator. Scale bar = 223.64 µm.</p>
Full article ">Figure 2
<p>Analysis of 3T3 nuclear morphology. (<b>A</b>) Nuclear intensity value/cell (<span class="html-italic">n</span> = 24). (<b>B</b>) Nuclear shape value (<span class="html-italic">n</span> = 24). (<b>C</b>) Nuclear area (<span class="html-italic">n</span> = 24). (<b>D</b>,<b>E</b>) Distribution of 3T3 nuclear shape values relative to nuclear intensity. (<b>F</b>,<b>G</b>) Distribution of 3T3 nuclear area values relative to nuclear intensity. Gray indicates percentage of nuclei &lt; 2n, blue indicates percentage of nuclei in G0/G1 phase, red indicates percentage of nuclei in S phase, green indicates percentage of nuclei in G2/M phase, and yellow indicates percentage of nuclei &gt; 4n.</p>
Full article ">Figure 3
<p>Distribution of microfilament bundles of 3T3 cells. Microfilaments were stained with phalloidin (green color), and nuclei were counterstained with H33342. White arrows indicate microvilli. Scale bar = 223.64 µm.</p>
Full article ">Figure 4
<p>Distribution of microtubules of 3T3 cells. Microtubules were stained with SiR-tubulin (red color). White arrows indicate perinuclear accumulations of microtubules; dashed arrows indicate distribution of microtubules surrounding nucleus. Scale bar = 223.64 µm.</p>
Full article ">Figure 5
<p>Western blot analysis of major structural proteins in 3T3 cells. α-Tubulin 3 and β-actin were downregulated in 3T3 cells under SMG (<span class="html-italic">n</span> = 3). GAPDH was used as internal control.</p>
Full article ">Figure 6
<p>Cell cycle progression analysis. (<b>A</b>,<b>B</b>) Cell cycle of 3T3 cells in control and SMG groups was analyzed by cell cycle app of Cytell microscope (<span class="html-italic">n</span> = 24). Gray indicates percentage of nuclei &lt; 2n, blue indicates percentage of nuclei in G0/G1 phase, red indicates percentage of nuclei in S phase, green indicates percentage of nuclei in G2/M phase, and yellow indicates percentage of nuclei &gt; 4n. (<b>C</b>) Western blot analysis of major cell cycle-related proteins in 3T3 cells (<span class="html-italic">n</span> = 3). (<b>D</b>) Number of 3T3 cells was counted by cell cycle app (<span class="html-italic">n</span> = 24).</p>
Full article ">Figure 7
<p>Morphology of cell division related structures in 3T3 cells: (<b>A</b>,<b>A1</b>) contractile ring in 3T3 cells in control group; (<b>B</b>,<b>B1</b>) contractile ring in 3T3 cells in SMG group; (<b>C</b>,<b>C1</b>) polar spindle in 3T3 cells in control group; (<b>D</b>,<b>D1</b>) polar spindle in 3T3 cells in SMG group. Microfilaments were counterstained using phalloidin (green), and microtubules were stained with SiR-tubulin (red). White arrows indicate contractile rings, and dashed arrows indicate polar spindles. Scale bar = 100 µm.</p>
Full article ">
12 pages, 1784 KiB  
Review
Processes Controlling the Contractile Ring during Cytokinesis in Fission Yeast, Including the Role of ESCRT Proteins
by Imane M. Rezig, Wandiahyel G. Yaduma and Christopher J. McInerny
J. Fungi 2024, 10(2), 154; https://doi.org/10.3390/jof10020154 - 15 Feb 2024
Viewed by 2072
Abstract
Cytokinesis, as the last stage of the cell division cycle, is a tightly controlled process amongst all eukaryotes, with defective division leading to severe cellular consequences and implicated in serious human diseases and conditions such as cancer. Both mammalian cells and the fission [...] Read more.
Cytokinesis, as the last stage of the cell division cycle, is a tightly controlled process amongst all eukaryotes, with defective division leading to severe cellular consequences and implicated in serious human diseases and conditions such as cancer. Both mammalian cells and the fission yeast Schizosaccharomyces pombe use binary fission to divide into two equally sized daughter cells. Similar to mammalian cells, in S. pombe, cytokinetic division is driven by the assembly of an actomyosin contractile ring (ACR) at the cell equator between the two cell tips. The ACR is composed of a complex network of membrane scaffold proteins, actin filaments, myosin motors and other cytokinesis regulators. The contraction of the ACR leads to the formation of a cleavage furrow which is severed by the endosomal sorting complex required for transport (ESCRT) proteins, leading to the final cell separation during the last stage of cytokinesis, the abscission. This review describes recent findings defining the two phases of cytokinesis in S. pombe: ACR assembly and constriction, and their coordination with septation. In summary, we provide an overview of the current understanding of the mechanisms regulating ACR-mediated cytokinesis in S. pombe and emphasize a potential role of ESCRT proteins in this process. Full article
(This article belongs to the Special Issue Yeast Cytokinesis)
Show Figures

Figure 1

Figure 1
<p>Medial positioning of the ACR in fission yeast. (<b>A</b>) Pom1p gradient at the cell tips restricts the division site to the cell center. Mitotic entry is controlled by Pom1p phosphorylation of Cdr2p, preventing Cdr2p from binding the plasma membrane and the subsequent formation of cortical nodes. Upon mitotic entry, both active Mid1p and Cdr2p scaffold the formation of medial cortical nodes. (<b>B</b>) Upon microtubule–cortex contact, polymerization at the microtubule plus end generates a pushing force (large arrowhead) (1) that displaces the nucleus in the opposite direction (nuclear movement demonstrated by red arrows) (2). The antiparallel direction of the microtubule bundle ensures that, over time, the nucleus oscillates back and forth toward the center (3). References within the main text.</p>
Full article ">Figure 2
<p>Assembly of the ACR from node precursors. (<b>A</b>) During interphase, type 1 “stationary” (green: Mid1p, Cdr1p and Cdr2p) nodes and type 2 “anchoring” (orange: Blt1p, Klp8p, Gef2p and Nod1p) nodes bind the plasma membrane and scaffold other cytokinesis proteins. (<b>B</b>) Coalescence of type 1 and type 2 nodes leads to their maturation into cytokinesis nodes (green–orange gradient). Maturation of cytokinesis nodes leads to the recruitment of Myo2p, Cdc15p and Cdc12p and nucleation of actin filaments. Interactions between myosin-II and actin promote ACR formation. References within the main text.</p>
Full article ">Figure 3
<p>Septum biosynthesis is coupled to contractile ring constriction. (<b>A</b>) A primary septum (yellow) flanked by two secondary septa layers (purple) forms behind the ACR as it constricts. Deposition of the septum material by membrane trafficking through secretory vesicles carrying the septum glucan synthases: secondary septum proteins Ags1p, Bgs4p and primary septum protein Bgs1p. (<b>B</b>) Cross-section of the deposited septum and constricting ring with internal turgor pressure (black), opposed by the inward forces of the ACR constriction (blue) and septation (purple). (<b>C</b>) Following the ACR closure, the primary septum layer is degraded by glucanases (red: Eng1p and Agn1p), which are delivered to the septum via a septin-based pathway, the secondary septa represent the cell wall material at the new cell tips. Derived from Proctor et al. [<a href="#B53-jof-10-00154" class="html-bibr">53</a>]; references within the main text.</p>
Full article ">Figure 4
<p>The role of ESCRTs during mammalian cytokinesis. During mammalian cytokinesis, a cleavage furrow forms medially between the two dividing cells, while the ACR constricts to create the midbody. (<b>A</b>) Sequential recruitment (steps 1–3) of the ESCRT-I subunits/factors leads to the recruitment of ESCRT-III polymers forming a spiral at the constricted midbody. (<b>B</b>) The ATPase VPS4 is then recruited to the midbody (step 4) where it disassembles the ESCRT-III polymers, this ensures ESCRT-III subunits are recycled at the division site with such fission reaction leading to the separation of daughter cells. ACR: actin–myosin contractile ring. MT: microtubules. References within the main text.</p>
Full article ">
13 pages, 1459 KiB  
Article
Effects of the Heterodimeric Neurotoxic Phospholipase A2 from the Venom of Vipera nikolskii on the Contractility of Rat Papillary Muscles and Thoracic Aortas
by Alexey Averin, Vladislav Starkov, Victor Tsetlin and Yuri Utkin
Toxins 2024, 16(2), 100; https://doi.org/10.3390/toxins16020100 - 10 Feb 2024
Cited by 3 | Viewed by 2284
Abstract
Phospholipases A2 (PLA2s) are a large family of snake toxins manifesting diverse biological effects, which are not always related to phospholipolytic activity. Snake venom PLA2s (svPLA2s) are extracellular proteins with a molecular mass of 13–14 kDa. [...] Read more.
Phospholipases A2 (PLA2s) are a large family of snake toxins manifesting diverse biological effects, which are not always related to phospholipolytic activity. Snake venom PLA2s (svPLA2s) are extracellular proteins with a molecular mass of 13–14 kDa. They are present in venoms in the form of monomers, dimers, and larger oligomers. The cardiovascular system is one of the multiple svPLA2 targets in prey organisms. The results obtained previously on the cardiovascular effects of monomeric svPLA2s were inconsistent, while the data on the dimeric svPLA2 crotoxin from the rattlesnake Crotalus durissus terrificus showed that it significantly reduced the contractile force of guinea pig hearts. Here, we studied the effects of the heterodimeric svPLA2 HDP-1 from the viper Vipera nikolskii on papillary muscle (PM) contractility and the tension of the aortic rings (ARs). HDP-1 is structurally different from crotoxin, and over a wide range of concentrations, it produced a long-term, stable, positive inotropic effect in PMs, which did not turn into contractures at the concentrations studied. This also distinguishes HDP-1 from the monomeric svPLA2s, which at high concentrations inhibited cardiac function. HDP-1, when acting on ARs preconstricted with 10 μM phenylephrine, induced a vasorelaxant effect, similar to some other svPLA2s. These are the first indications of the cardiac and vascular effects of true vipers’ heterodimeric svPLA2s. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of different concentrations of HDP-1 on the contraction force of the papillary muscles (PMs). Representative traces show the control ((<b>a</b>) <span class="html-italic">n</span> = 5; here and in the next figures, this is the number of PMs) and the effects of HDP-1 at concentrations of 10 nM ((<b>b</b>) <span class="html-italic">n</span> = 5), 100 nM ((<b>c</b>) <span class="html-italic">n</span> = 5), 500 nM ((<b>d</b>) <span class="html-italic">n</span> = 6), and 1 μM ((<b>e</b>) <span class="html-italic">n</span> = 6). The arrow indicates the time point at which HDP-1 was added. (<b>f</b>) Quantitative data, where the ordinate shows the force of contraction at 0.3 Hz normalized to that obtained before the addition of HDP-1. Similar to the experimental groups, the contraction force in a separate control group (<span class="html-italic">n</span> = 5) was recorded 30 min after the start of the measurement. Data are presented as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Effect of HDP-1 (500 nM) on the force–frequency relation of the PMs. (<b>a</b>) Representative traces show the dependence of the contraction force on the stimulation frequency. The stimulation rate was increased stepwise to 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.8, 1.0, 1.5, 2.0, and 3.0 Hz. (<b>b</b>) Statistical data, where the ordinate shows the force of contraction normalized to the force of contraction obtained at 0.1 Hz in the control. Data are presented as the mean ± SEM. <span class="html-italic">n</span> = 6. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effect of HDP-1 (500 nM) on post-rest potentiation of PMs. (<b>a</b>) Representative traces show the dependence of the contraction force on the duration of the pause. (<b>b</b>) Statistical data, where the ordinate shows the force of contraction normalized to the force of contraction obtained at 0.1 Hz in the control. Data are presented as the mean ± SEM, <span class="html-italic">n</span> = 5. * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Schematic illustration of the experiments on the contraction of the ARs. KCl, isotonic solution with 80 mM KCl; ACh, 10 µM acetylcholine; PE, 10 μM phenylephrine. (<b>a</b>) Control. In (<b>b</b>), HDP-1 was added at a concentration of 100 nM. Horizontal lines indicate the presence of the corresponding reagent in the washing solution. The rectangle shows the registered relaxant effect of HDP-1 at the concentration applied (10 nM to 1 μM).</p>
Full article ">Figure 5
<p>Influence of HDP-1 on tension of ARs preconstricted with 10 μM PE. (<b>a</b>) Representative traces showing the control (<span class="html-italic">n</span> = 9) and the effects of HDP-1 at concentrations of 10 nM (<span class="html-italic">n</span> = 6), 100 nM (<span class="html-italic">n</span> = 7), 500 nM (<span class="html-italic">n</span> = 4), and 1 μM (<span class="html-italic">n</span> = 5), where n is the number of ARs studied. (<b>b</b>) Quantitative data, where the ordinate shows the level of relaxation. Data are presented as the mean ± SEM. * <span class="html-italic">p</span> &lt; 0.05 compared to the control. In the control experiment, no HDP-1 was added.</p>
Full article ">
22 pages, 7810 KiB  
Article
A Putative Role of Vasopressin/Oxytocin-Type Neuropeptide in Osmoregulation and Feeding Inhibition of Apostichopus japonicus
by Xiao Cong, Huachen Liu, Yingqiu Zheng and Muyan Chen
Int. J. Mol. Sci. 2023, 24(18), 14358; https://doi.org/10.3390/ijms241814358 - 20 Sep 2023
Cited by 2 | Viewed by 1560
Abstract
Vasopressin/oxytocin (VP/OT)-type neuropeptide is an ancient neurophysin-associated neuropeptide and has been intensively studied to be involved in multiple physiological processes in protostomian and deuterostome vertebrates. However, little is known about the functions of VP/OT-type neuropeptide in deuterostome invertebrates especially in echinoderms. Here, we [...] Read more.
Vasopressin/oxytocin (VP/OT)-type neuropeptide is an ancient neurophysin-associated neuropeptide and has been intensively studied to be involved in multiple physiological processes in protostomian and deuterostome vertebrates. However, little is known about the functions of VP/OT-type neuropeptide in deuterostome invertebrates especially in echinoderms. Here, we firstly report VP/OT-type neuropeptide signaling in an important economic species, Apostichopus japonicus, which is widely cultured in Asia, with high nutritional and medicinal values. Molecular characterization analysis of holotocin and its precursor revealed the highly conserved features of VP/OT family. The candidate receptor for holotocin (AjHOR) was confirmed to be able to activate the signaling via cAMP-PKA and possible Ca2+-PKC pathway, and further activated the downstream ERK1/2 cascade. Holotocin precursor expression profile showed that they were mainly concentrated in circumoral nerve ring. Furthermore, in vitro pharmacological experiments demonstrated that holotocin caused contractile responses in preparations from A. japonicus. And in vivo functional studies indicated that short-term injection of holotocin resulted in body bloat and long-term injection resulted in reduced body mass, suggesting potential roles of holotocin in osmoregulation and feeding co-inhibition with holotocin–CCK. Our findings provided a comprehensive description of AjHOR–holotocin signaling, revealed ancient roles of holotocin in osmoregulation and feeding inhibition by controlling muscle contractions. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Figure 1
<p>Characterization of holotocin, holotocin precursor and holotocin precursor genes in <span class="html-italic">A. japonicus.</span> (<b>A</b>) Amino acid sequences of <span class="html-italic">A. japonicus</span> holotocin precursor with the predicted signal peptide are shown in blue, a predicted cleavage site is shown in green, holotocin is shown in red, a C-terminal glycine that is a putative substrate for amidation is shown in orange and the neurophysin domain is shown in purple. The two red underlined cysteines form disulfide bridges, and the cysteines in neurophysin domain are shown with a purple underline. (<b>B</b>) Comparison of VP/OT-type neuropeptides from deuterostomes. The conserved pair of cysteine residues was shown by *. The GenBank numbers are shown in the <a href="#app1-ijms-24-14358" class="html-app">Supplementary Table S1</a>. (<b>C</b>) Conserved domains of protein sequences from VP/OT family. Gray line represent the length of proteins, and domains are represented by rectangles. (<b>D</b>) Comparison of VP/OT-type neuropeptide precursor proteins in Holothuroidea. The same amino acids are colored. (<b>E</b>) DNA structure of VP/OT-type neuropeptide precursor genes. Exons are represented by blue rectangles, introns are represented by lines and upstream and downstream are represented by red rectangles. The position of intron phase is represented by numbers. (<b>F</b>) Synteny of <span class="html-italic">A. rubens</span>, <span class="html-italic">L. variegatus</span> and <span class="html-italic">A. japonicus</span> VP/OT-type neuropeptide precursor genes. The chromosome location of VP/OT-type neuropeptide precursor genes is designated, and the relative orientation of adjacent genes is shown and various genes are marked in different colors. Transcriptional direction of genes is shown by arrows. <span class="html-italic">NCAP</span>, condensin complex subunit; <span class="html-italic">KCNK</span>, potassium channel subfamily K; <span class="html-italic">DOCK1</span>, dedicator of cytokinesis protein 1; <span class="html-italic">LPHN2</span>, latrophilin-2; <span class="html-italic">iPGAM</span>, 2,3-bisphosphoglycerate-independent phosphoglycerate mutase; and <span class="html-italic">NP</span>, nucleolar protein.</p>
Full article ">Figure 2
<p>Holotocin precursor and <span class="html-italic">AjHOR</span> relative transcript expression levels in different tissues of <span class="html-italic">A. japonicus</span>. (<b>A</b>) Holotocin precursor gene and (<b>B</b>) <span class="html-italic">AjHOR</span>. The significant difference (<span class="html-italic">p</span> &lt; 0.05) is shown by different lowercase letters. Values are mean ± S.E.M (n = 5 biological replicates).</p>
Full article ">Figure 3
<p>Localization of holotocin precursor in <span class="html-italic">A. japonicus</span> using mRNA in situ hybridization. (<b>A</b>) Intestine. (<b>B</b>) Mesentery. (<b>C</b>) Oral position: water vascular system, tentacle and circumoral nerve ring. (<b>D</b>) Longitudinal muscle. (<b>E</b>) Body wall. (<b>F</b>) Water canal. (<b>G</b>) Tube feet. Ce, cuticle; CE, coelomic epithelium; CNR, circumoral nerve ring; CT, connective tissue; Ds, dermis; Es, epidermis; IL, intestine lumen; My, mesentery; NP, nerve plexus; RNC, radial nerve cord; SCE, simple columnar epithelium; Te, tentacle; WC, water canal; WVS, water vascular system; WVE, water vascular epidermis; and WVLM, water vascular longitudinal muscle. Holotocin precursor signals in different tissues were shown by arrows. Scale bar: 200 µm in (<b>C</b>); 100 µm in (<b>A</b>,<b>D</b>,<b>E</b>,<b>G</b>); 50 µm in (<b>B</b>,<b>F</b>).</p>
Full article ">Figure 4
<p>Functional characteristics of holotocin signaling system. (<b>A</b>) CRE-driven luciferase activities in <span class="html-italic">AjHOR</span>-expressing HEK293T cells was detected. (<b>B</b>) Quantitative detection of cAMP accumulation in <span class="html-italic">AjHOR</span>-expressing cells under the stimulation of holotocin (10<sup>−9</sup>–10<sup>−5</sup> M). Intracellular Ca<sup>2+</sup> mobilization in <span class="html-italic">AjHOR</span>-expressing HEK293T cells was measured in response to holotocin (10<sup>−8</sup>–10<sup>−5</sup> M) (<b>C</b>), and holotocin caused concentration-dependent activation of <span class="html-italic">AjHOR</span> (<b>D</b>). (<b>E</b>) Internalization of <span class="html-italic">AjHOR</span>/pEGFP-N1 was initiated by 10<sup>−6</sup> M holotocin in <span class="html-italic">AjHOR</span>/pEGFP-N1-expressing HEK293T cells after incubation with indicated times. Cell nucleus were stained by cell nucleus probe (DAPI). Scale bar: 100 µm. (<b>F</b>) Immunoblot intensity of representative bands. Holotocin stimulated phosphorylation of ERK1/2 in <span class="html-italic">AjHOR</span>-expressing HEK293T cells, which were incubated with indicated times. (<b>G</b>) The normalized p-ERK1/2 according to t-ERK1/2. Error bars represent SEM for three independent experiments. All pictures and data are representative for at least three independent experiments. * Significant differences between 0 min and treated groups with <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Holotocin caused contractions in longitudinal muscle, tentacle and intestine from <span class="html-italic">A. japonicus</span>. (<b>A</b>) A representative recording showed that holotocin (10<sup>−9</sup>–10<sup>−5</sup> M) caused tonic contraction of longitudinal muscle preparation. (<b>B</b>) Graph showed the dose-dependent contracting effect of holotocin (10<sup>−9</sup>–10<sup>−5</sup> M) and NGIWYamide (10<sup>−7</sup>–10<sup>−5</sup> M) on longitudinal muscle preparations. (<b>C</b>) Comparison of contraction efficiency of holotocin and NGIWYamide at different concentrations, and the contraction efficiency of holotocin in 10<sup>−5</sup> M was defined as 100%. (<b>D</b>) A representative recording showed that holotocin (10<sup>−7</sup>–10<sup>−5</sup> M) caused tonic contraction of intestine preparation. (<b>E</b>) Graph showed the dose-dependent contracting effect of holotocin on intestine preparations. Responses were expressed as the mean percentage contraction (mean ± S.E.M.; n = 3). (<b>F</b>) The effect of holotocin (10<sup>−5</sup> M) on tentacle preparation in vitro. * The effect of muscle contractions induced by holotocin and NGIWYamide at the same concentration was significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Physiological response of holotocin in <span class="html-italic">A. japonicus</span>. (<b>A</b>) Morphological changes in <span class="html-italic">A. japonicus</span> after injection with seawater or 10<sup>−5</sup> M holotocin. (<b>B</b>) Comparison of wet weight before and after 10 min or 90 min of holotocin injection. The significant difference (<span class="html-italic">p</span> &lt; 0.05) is shown by *, and <span class="html-italic">p</span> &lt; 0.01 is shown by **.</p>
Full article ">Figure 7
<p><span class="html-italic">AjCCKP1</span> and <span class="html-italic">AjCCKP2</span> relative transcript expression levels in different stages after injection of holotocin in <span class="html-italic">A. japonicus</span>. (<b>A</b>) <span class="html-italic">AjCCKP1</span> was tested after injection of holotocin with indicated times. (<b>B</b>) <span class="html-italic">AjCCKP2</span> was tested after injection of holotocin with indicated times. The significant difference (<span class="html-italic">p</span> &lt; 0.05) is shown by *. Values are mean ± S.E.M. (n = 5 biological replicates). CO: control group (sterilized seawater); HOL: holotocin low concentration group (5 × 10<sup>−3</sup> mg/mL); and HOH: holotocin high concentration group (5 × 10<sup>−1</sup> mg/mL).</p>
Full article ">Figure 8
<p>Effect of holotocin injection on growth of <span class="html-italic">A. japonicus</span>. (<b>A</b>) Wet weight changes before and after injection of holotocin in different groups. The significant difference (<span class="html-italic">p</span> &lt; 0.05) is shown by *. (<b>B</b>) Calculation of the residual bait weight in different phases. (<b>C</b>) The feces weight in different phases after injection of holotocin in different phases. CO: control group (sterilized seawater); HOL: holotocin low concentration group (5 × 10<sup>−3</sup> mg/mL); and HOH: holotocin high concentration group (5 × 10<sup>−1</sup> mg/mL).</p>
Full article ">Figure 9
<p>Diagram showing VP/OT-type neuropeptides from representative species in the Bilateria and the corresponding pharmacological effects in vitro. Species are classified by different colors, a dash (−) indicates that VP/OT-type neuropeptides were not functionally validated in vitro. Because of the specificity of asterotocin function, the diagram summarizes the functions of echinoderms by presenting the function of <span class="html-italic">A. rubens</span> in addition to summarizing the functions of <span class="html-italic">A. japonicus</span> in this study.</p>
Full article ">Figure 10
<p>A schematic of holotocin’s potential physiological functions in <span class="html-italic">A. japonicus</span>. Holotocin are represented by the yellow circles, <span class="html-italic">AjHOR</span> are represented by green line. AjCCK1 and AjCCK2 are represented by pink and green circles respectively.</p>
Full article ">
18 pages, 5569 KiB  
Article
Lamin A/C Ablation Restricted to Vascular Smooth Muscle Cells, Cardiomyocytes, and Cardiac Fibroblasts Causes Cardiac and Vascular Dysfunction
by Alberto Del Monte-Monge, Íñigo Ruiz-Polo de Lara, Pilar Gonzalo, Carla Espinós-Estévez, María González-Amor, Miguel de la Fuente-Pérez, María J. Andrés-Manzano, Víctor Fanjul, Juan R. Gimeno, Roberto Barriales-Villa, Beatriz Dorado and Vicente Andrés
Int. J. Mol. Sci. 2023, 24(13), 11172; https://doi.org/10.3390/ijms241311172 - 6 Jul 2023
Viewed by 2507
Abstract
Mutations in the LMNA gene (encoding lamin A/C proteins) cause several human cardiac diseases, including dilated cardiomyopathies (LMNA-DCM). The main clinical risks in LMNA-DCM patients are sudden cardiac death and progressive left ventricular ejection fraction deterioration, and therefore most human [...] Read more.
Mutations in the LMNA gene (encoding lamin A/C proteins) cause several human cardiac diseases, including dilated cardiomyopathies (LMNA-DCM). The main clinical risks in LMNA-DCM patients are sudden cardiac death and progressive left ventricular ejection fraction deterioration, and therefore most human and animal studies have sought to define the mechanisms through which LMNA mutations provoke cardiac alterations, with a particular focus on cardiomyocytes. To investigate if LMNA mutations also cause vascular alterations that might contribute to the etiopathogenesis of LMNA-DCM, we generated and characterized Lmnaflox/floxSM22αCre mice, which constitutively lack lamin A/C in vascular smooth muscle cells (VSMCs), cardiac fibroblasts, and cardiomyocytes. Like mice with whole body or cardiomyocyte-specific lamin A/C ablation, Lmnaflox/floxSM22αCre mice recapitulated the main hallmarks of human LMNA-DCM, including ventricular systolic dysfunction, cardiac conduction defects, cardiac fibrosis, and premature death. These alterations were associated with elevated expression of total and phosphorylated (active) Smad3 and cleaved (active) caspase 3 in the heart. Lmnaflox/floxSM22αCre mice also exhibited perivascular fibrosis in the coronary arteries and a switch of aortic VSMCs from the ‘contractile’ to the ‘synthetic’ phenotype. Ex vivo wire myography in isolated aortic rings revealed impaired maximum contraction capacity and an altered response to vasoconstrictor and vasodilator agents in Lmnaflox/floxSM22αCre mice. To our knowledge, our results provide the first evidence of phenotypic alterations in VSMCs that might contribute significantly to the pathophysiology of some forms of LMNA-DCM. Future work addressing the mechanisms underlying vascular defects in LMNA-DCM may open new therapeutic avenues for these diseases. Full article
(This article belongs to the Special Issue Cardiovascular Diseases: Molecular Mechanisms and Potential Therapy)
Show Figures

Figure 1

Figure 1
<p>Lamin A/C ablation in VSMCs in <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. Representative immunofluorescence images of aorta, liver, kidney, and lung from 4-week-old <span class="html-italic">Lmna<sup>flox/flox</sup></span> (<b>A</b>) and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice (<b>B</b>). Lamin A/C is visualized in white, endothelial cells in green (anti-CD31 antibody), VSMCs in red (anti-smooth muscle α-actin (SMA) antibody), and nuclei in blue (DAPI staining). Magnified images show vessel-containing regions (*) and vessel-free regions (**).</p>
Full article ">Figure 2
<p>Lamin A/C ablation in heart VSMCs, cardiomyocytes, and cardiac fibroblasts in <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. (<b>A</b>,<b>B</b>) Representative immunofluorescence images of heart tissue from 4-week-old <span class="html-italic">Lmna<sup>flox/flox</sup></span> (<b>A</b>) and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice (<b>B</b>). Lamin A/C is visualized in white, endothelial cells in green (anti-CD31 antibody), VSMCs in red (anti-smooth muscle α-actin (SMA) antibody), and nuclei in blue (DAPI staining). Magnifications show vessel-containing (*) and vessel-free (**) regions. (<b>C</b>) Representative immunofluorescence images of heart tissue from 4-week-old <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. Lamin A/C is visualized in red, cell membranes in green (wheat germ agglutinin; WGA), cardiac fibroblasts in white (anti-FSP-1 antibody), and nuclei in blue (Hoechst 33342 staining). Graphs show the percentages of lamin A/C-positive nuclei in cardiomyocytes and cardiac fibroblasts. Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test. Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 3
<p><span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice exhibit reduced body weight and lifespan. (<b>A</b>) Circulating blood cell counts in 4-week-old <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. (<b>B</b>) Representative photograph of male and female mice of both genotypes. The graph shows body weight at 4 weeks of age. (<b>C</b>) Kaplan–Meier survival curves of <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> (median survival 33 days) and control <span class="html-italic">Lmna<sup>flox/flox</sup></span> mice (n = 19 mice per genotype). Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test (<b>A</b>,<b>B</b>) and the log-rank (Mantel–Cox) test (<b>C</b>). Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 4
<p><span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice show increased vascular collagen content. Representative images of Masson’s trichrome staining in the (<b>A</b>) liver, lung, kidney, (<b>B</b>) aortic arch, thoracic aorta, and (<b>C</b>) coronary arteries of 4-week-old <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. Graphs show collagen content calculated relative to the content in <span class="html-italic">Lmna<sup>flox/flox</sup></span> mice (=1). Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test. Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 5
<p><span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice develop cardiac fibrosis. <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice were examined at 4 weeks of age. (<b>A</b>) Representative images of Masson’s trichrome and Sirius red staining; graphs show collagen content in heart vessel-free regions calculated relative to the content in <span class="html-italic">Lmna<sup>flox/flox</sup></span> mice (=1). One outlier identified with the GraphPad outlier calculator in the <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> group was eliminated. (<b>B</b>) Representative immunofluorescence images of heart tissue stained with wheat germ agglutinin to visualize cell membranes (WGA, green), anti-FSP-1 antibody to identify cardiac fibroblasts (white), and anti-smooth muscle actin antibody to identify fibrogenic activated fibroblasts (SMA, red). Graphs show the positive area relative to the total area of tissue. Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test. Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 6
<p>Increased total Smad3, phosphorylated Smad3, and active caspase 3 protein levels in <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mouse hearts. <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice were examined at 4 weeks of age. (<b>A</b>) Representative immunofluorescence images of heart tissue showing phosphorylated Smad3 (S423 + S425; p-Smad3; white) and nuclei (stained with Hoechst 33342; blue). Graphs show p-Smad3-positive nuclei (top) and median intensity fluorescence (MIF) of p-Smad3-positive nuclei (bottom). (<b>B</b>) Representative Western blots of heart protein lysates probed with antibodies against p-Smad3, Smad3, and vinculin (the latter used as a housekeeping loading control). Each lane corresponds to the heart tissue from one mouse. Membranes incubated with anti-p-Smad3 antibody were stripped off and used for incubation with anti-Smad3 antibody. Graphs show relative p-Smad3 and total Smad3 expression normalized to vinculin expression (left and middle graphs, respectively) and the p-Smad3/Smad3 ratio after normalization to vinculin (right graph). One outlier identified in the <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> group using the Grubbs’ test was not included in the left graph. (<b>C</b>) Representative immunofluorescence images of the active (cleaved) form of caspase-3 (white) and nuclei (Hoechst 33342; blue). The graph shows the active caspase-3 positive area as a percentage of the total area of tissue. Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test. Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 7
<p><span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice show a severe loss of cardiac function and electrocardiographic defects. <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice were examined at 4 weeks of age. (<b>A</b>) Representative echocardiography images (sagittal plane) and quantification of left ventricle (LV) function (EF, ejection fraction), right ventricle (RV) function (TAPSE, tricuspid annular plane systolic excursion), and LV wall thickness. These results are also shown in table format in <a href="#app1-ijms-24-11172" class="html-app">Supplementary Table S1</a>. Sagittal and longitudinal planes are shown in <a href="#app1-ijms-24-11172" class="html-app">Supplementary Videos S1–S4</a>. (<b>B</b>) Representative images of hearts and tibia bones and quantification of tibia length and heart weight. (<b>C</b>) Quantification of PQ, QRS, and QT intervals and T-wave steepness obtained by electrocardiography. (<b>D</b>) Plasma levels of creatine kinase MB isoform (CK-MB) and troponin. One outlier identified in the <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> group using the Grubbs’ test was not included in the analysis of plasma troponin. Statistical analysis was conducted using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test for CK-MB and by the non-parametric Mann–Whitney test for troponin (troponin data did not follow a normal distribution). Data are mean ± SEM. Each symbol represents one animal.</p>
Full article ">Figure 8
<p><span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice exhibit contractile-to-synthetic phenotypic switching in vascular smooth muscle cells and vascular dysfunction in the aorta. <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice were examined at 4 weeks of age. (<b>A</b>) Protocol for processing mouse thoracic aorta (TA) samples for real-time quantitative PCR (RT-qPCR). Aortas were isolated from <span class="html-italic">Lmna<sup>flox/flox</sup></span> and <span class="html-italic">Lmna<sup>flox/flox</sup>SM22αCre</span> mice. After removing the aortic arch and perivascular tissue, TA samples were incubated with type I collagenase. Adventitia was then removed manually, and samples from mice of the same genotype and sex were paired for isolation of total RNA. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>. (<b>B</b>) RT-qPCR analysis of adventitia-free thoracic aorta, examining the expression of vascular smooth muscle cell ‘contractile’ and ‘synthetic’ genes (n = 5). Each replicate contained the thoracic aortas from two mice of the same genotype and sex. Data are presented as the ΔCt fold change relative to control samples. <span class="html-italic">Hprt</span> was used as the housekeeping gene. The heatmap shows the log<sub>2</sub> of the fold change relative to control <span class="html-italic">Lmna<sup>flox/flox</sup></span> mice. (<b>C</b>–<b>H</b>) Thoracic aorta rings were mounted in a wire myograph system to examine the following parameters (n = 10 each genotype): diameter–force relationship and its linear regression slope (an estimation of vessel stiffness) (<b>C</b>); estimated aortic ring diameter at 100 mmHg (<b>D</b>); maximum response induced by 120 mmol/L KCl (<b>E</b>); concentration–response curves to phenylephrine, and the concentration of phenylephrine giving the half-maximal response (EC<sub>50</sub>) (<b>F</b>); endothelium-dependent vasodilation induced by increasing concentrations of acetylcholine; (<b>G</b>); and endothelium-independent vasodilation induced by increasing concentrations of diethylamine NONOate (DEA-NO) (<b>H</b>). Statistical differences were analyzed using an unpaired two-tailed Student’s <span class="html-italic">t</span>-test in (<b>B</b>,<b>C</b>) (right graph) and (<b>D</b>–<b>F</b>) (right graph) or by two-way ANOVA and Fisher’s LSD multiple comparisons test in the dose–response curves in (<b>F</b>–<b>H</b>). Data are mean ± SEM.</p>
Full article ">
Back to TopTop