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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (25)

Search Parameters:
Keywords = auraptene

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 1005 KiB  
Review
Activation of Nrf2 and FXR via Natural Compounds in Liver Inflammatory Disease
by Marta Belka, Aleksandra Gostyńska-Stawna, Maciej Stawny and Violetta Krajka-Kuźniak
Int. J. Mol. Sci. 2024, 25(20), 11213; https://doi.org/10.3390/ijms252011213 - 18 Oct 2024
Cited by 1 | Viewed by 1218
Abstract
Liver inflammation is frequently linked to oxidative stress and dysregulation of bile acid and fatty acid metabolism. This review focuses on the farnesoid X receptor (FXR), a critical regulator of bile acid homeostasis, and its interaction with the nuclear factor erythroid 2-related factor [...] Read more.
Liver inflammation is frequently linked to oxidative stress and dysregulation of bile acid and fatty acid metabolism. This review focuses on the farnesoid X receptor (FXR), a critical regulator of bile acid homeostasis, and its interaction with the nuclear factor erythroid 2-related factor 2 (Nrf2), a key modulator of cellular defense against oxidative stress. The review explores the interplay between FXR and Nrf2 in liver inflammatory diseases, highlighting the potential therapeutic effects of natural FXR agonists. Specifically, compounds such as auraptene, cafestol, curcumin, fargesone A, hesperidin, lycopene, oleanolic acid, resveratrol, rutin, ursolic acid, and withaferin A are reviewed for their ability to modulate both the FXR and Nrf2 pathways. This article discusses their potential to alleviate liver inflammation, oxidative stress, and damage in diseases such as metabolic-associated fatty liver disease (MAFLD), cholestatic liver injury, and viral hepatitis. In addition, we address the molecular mechanisms driving liver inflammation, including oxidative stress, immune responses, and bile acid accumulation, while also summarizing relevant experimental models. This review emphasizes the promising therapeutic potential of targeting both the Nrf2 and FXR pathways using natural compounds, paving the way for future treatments for liver diseases. Finally, the limitations of the clinical application were indicated, and further research directions were proposed. Full article
Show Figures

Figure 1

Figure 1
<p>Canonical and non-canonical mechanisms of Nrf2 activation. Abbreviations: ARE, Antioxidant Response Element; CAT, catalase; Cul3, Cullin 3; GCLC, glutamate-cysteine ligase catalytic subunit; GPx, glutathione peroxidase; GSTs, glutathione S-transferases; GSR1, glutathione reductase 1; HO-1, heme oxygenase-1; Keap1, Kelch-like ECH-associated protein 1; MAPKs, mitogen-activated protein kinases; NQO1, NAD(P)H oxidoreductase 1; Nrf2, Nuclear factor erythroid 2-related factor 2; p62, Sequestosome 1; PI3K, phosphatidylinositol 3-kinase; ROS, reactive oxygen species; sMAF, small musculoaponeurotic fibrosarcoma oncogene homolog; SOD, superoxide dismutase; Ub, ubiquitination. Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>.</p>
Full article ">Figure 2
<p>Interconnection between Nrf2 and FXR in inflammation. ↑/↓ increase/decrease. Abbreviations: BSEP, bile salt export pump; CAT, catalase; FXR, farnesoid X receptor; GCLC, glutamate-cysteine ligase catalytic subunit; GPx, glutathione peroxidase; GSTs, glutathione S-transferases; GSR1, glutathione reductase 1; HO-1, heme oxygenase-1; MRP2, multidrug resistance-associated proteins 2; MRP3, multidrug resistance-associated proteins 3; NQO1, NAD(P)H oxidoreductase 1; Nrf2, nuclear factor erythroid 2-related factor 2; OSTß, organic solute transporter beta; P300, P300 protein; β-catenin, beta-catenin; ROS, reactive oxygen species; RXRα, retinoid X receptor; SHP, small heterodimer partner, sMAF, small musculoaponeurotic fibrosarcoma oncogene homolog; SOD, superoxide dismutase; UGT1A1, UDP-glucuronyl transferase 1A1. Created with BioRender.com.</p>
Full article ">
20 pages, 7024 KiB  
Article
Auraptene Boosts the Efficacy of the Tamoxifen Metabolites Endoxifen and 4-OH-Tamoxifen in a Chemoresistant ER+ Breast Cancer Model
by Angel Pulido-Capiz, Brenda Chimal-Vega, Luis Pablo Avila-Barrientos, Alondra Campos-Valenzuela, Raúl Díaz-Molina, Raquel Muñiz-Salazar, Octavio Galindo-Hernández and Victor García-González
Pharmaceutics 2024, 16(9), 1179; https://doi.org/10.3390/pharmaceutics16091179 - 6 Sep 2024
Viewed by 950
Abstract
Approximately 80% of breast cancer (BC) cases are estrogen receptor positive (ER+) and sensitive to hormone treatment; Tamoxifen is a prodrug, and its main plasmatic active metabolites are 4-hydroxytamoxifen (4-OH Tam) and endoxifen. Despite the effectiveness of tamoxifen therapy, resistance can be developed. [...] Read more.
Approximately 80% of breast cancer (BC) cases are estrogen receptor positive (ER+) and sensitive to hormone treatment; Tamoxifen is a prodrug, and its main plasmatic active metabolites are 4-hydroxytamoxifen (4-OH Tam) and endoxifen. Despite the effectiveness of tamoxifen therapy, resistance can be developed. An increment in eukaryotic initiation factor-4A complex (eIF4A) activity can result in tamoxifen-resistant tumor cells. For this work, we developed a cell variant resistant to 4-OH Tam and endoxifen, denominated MCF-7Var E; then, the aim of this research was to reverse the acquired resistance of this variant to tamoxifen metabolites by incorporating the natural compound auraptene. Combination treatments of tamoxifen derivatives and auraptene successfully sensitized the chemoresistant MCF-7Var E. Our data suggest a dual regulation of eIF4A and ER by auraptene. Joint treatments of 4-OH Tam and endoxifen with auraptene identified a novel focus for chemoresistance disruption. Synergy was observed using the auraptene molecule and tamoxifen-derived metabolites, which induced a sensitization in MCF-7Var E cells and ERα parental cells that was not observed in triple-negative breast cancer cells (TNBC). Our results suggest a synergistic effect between auraptene and tamoxifen metabolites in a resistant ER+ breast cancer model, which could represent the first step to achieving a pharmacologic strategy. Full article
(This article belongs to the Special Issue Natural Products for Anticancer Application)
Show Figures

Figure 1

Figure 1
<p>Resistance development in MCF-7 cells and its characterization. (<b>A</b>) Illustration of the protocol used to carry out chemoresistance, showing the procedure for resistance acquisition in MCF-7<sup>Var E</sup> cells. (<b>B</b>) Comparison of the EC<sub>50</sub> values and SD among MCF-7 and MCF-7<sup>var E</sup> cells under the 4-OH Tam (<b>C</b>) and endoxifen (<b>D</b>) treatments (0–16 µM).</p>
Full article ">Figure 2
<p>Role of estrogen receptor (ER) in chemoresistance. (<b>A</b>) Expression of ER and cathepsin D. The comparative expression between MCF-7 and MCF-7<sup>Var E</sup> under the indicating conditions, with E2 (2 µM) and tamoxifen metabolites treatment (8 µM). (<b>B</b>) Quantitative analysis of ER expression. Comparative expression by densitometry analysis in three independent experiments of ER in MCF-7 and MCF-7<sup>Var E</sup> cells. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as fold-change in regard to loading control; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 in regard to control. GAPDH were used as loading controls.</p>
Full article ">Figure 3
<p>eIF4F complex characterization. (<b>A</b>) Expression of eIF4A and eIF4G targets. The comparative expression between MCF-7 and MCF-7<sup>Var E</sup> cells under different treatments, with E2 (2 µM) and tamoxifen metabolites (8 µM) for 12 h. Densitometry analysis of eIF4A (<b>B</b>) and eIF4G (<b>C</b>) in MCF-7 and MCF-7<sup>Var E</sup> cells under the same conditions. (<b>D</b>) Expression of eIF4E and p-4E-BP1,2,3. The comparative expression between MCF-7 and MCF-7<sup>Var E</sup> under different treatments with E2 and tamoxifen metabolites is shown. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as fold-change in regard to loading control; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.002 in regard to control. β-actin and GAPDH were used as loading controls.</p>
Full article ">Figure 4
<p>Effect of auraptene on the eIF4A regulation. (<b>A</b>) Coomassie-stained SDS-PAGE of eIF4A-purified fractions corresponding to TL: total lysate, T: tagged protein, and U: untagged protein. (<b>B</b>) Immunodetection for the fractions of eIF4A overexpression. Antibodies to detect eIF4A and polyhistidine tags were used. (<b>C</b>) Fluorescence assay of eIF4A and auraptene. Emission fluorescence spectra of eIF4A, eIF4A plus 50 µM auraptene, and eIF4A plus 100 µM auraptene are shown in blue, red, and green, respectively. (<b>D</b>) Docking of rocaglamide in the eIF4A RNA binding site (PDB ID: 5ZC9). eIF4A protein is shown in beige and rocaglamide molecule in green. (<b>E</b>) Docking of auraptene in the eIF4A RNA interaction site. eIF4A is shown in beige and auraptene in green. (<b>F</b>) Rocaglamide–eIF4A ligand interactions, showing the residues and type of interaction. (<b>G</b>) Auraptene–eIF4A binding residues and the nature of the interactions with molecules are shown. (<b>H</b>) E-score values obtained for each molecule in the molecular docking simulation.</p>
Full article ">Figure 5
<p>Cell viability of MCF-7 and MCF-7<sup>Var E</sup> cells with auraptene treatments. Comparison of auraptene and auraptene plus tamoxifen metabolite treatments. Viability percentages of MCF-7 cells under auraptene (<b>A</b>), auraptene and 4-OH tamoxifen (<b>B</b>), and auraptene and endoxifen treatments (<b>C</b>). Results of auraptene and auraptene plus tamoxifen metabolites shown in black and gray columns, respectively. Viability percentages of MCF-7<sup>Var E</sup> cells under auraptene (<b>D</b>), auraptene and 4-OH tamoxifen (<b>E</b>), and auraptene and endoxifen (<b>F</b>) treatments. Results of auraptene and auraptene plus tamoxifen metabolites shown in black and gray columns, respectively. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3); *** <span class="html-italic">p</span> &lt; 0.0001 regard to control.</p>
Full article ">Figure 6
<p>Auraptene and mixed auraptene plus tamoxifen metabolites treatments on ER+ and ER- cell variants. Comparative cell viability percentages of MCF-7, MCF-7<sup>Var E</sup>, and MDA-MB-231 cells are shown in black, light gray, and pattern gray, respectively, under increasing doses of auraptene plus 4-OH Tam 8 µM (<b>A</b>) and endoxifen 8 µM (<b>B</b>). Results are reported as mean ± SD (<span class="html-italic">n</span> = 3); # <span class="html-italic">p</span> &lt; 0.0001 regard to control.</p>
Full article ">Figure 7
<p>Characterization of eIF4F complex and ER. Molecular docking of molecules of auraptene and tamoxifen metabolites on estrogen receptor. Molecular docking of auraptene that is shown in green (<b>A</b>), endoxifen in purple (<b>B</b>), and 4-OH tamoxifen in light blue (<b>C</b>) on the ERα (PDB: 3ERT). ERα is shown in beige. Binding site residues and their types of interactions with auraptene (<b>D</b>), endoxifen (<b>E</b>), and 4-OH tamoxifen (<b>F</b>). (<b>G</b>) Comparison between MCF-7 and MCF-7<sup>Var E</sup> under treatments with metabolites of tamoxifen and aurapteno for ER, Cateptsin D, eIF4A, eIF4G, and eIF4E. Densitometry analysis of ER (<b>H</b>) and eIF4G (<b>I</b>) in MCF-7 and MCF-7<sup>Var E</sup> cells under the same conditions. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as fold-change in regard to loading control; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.002, *** <span class="html-italic">p</span> &lt; 0.001 in regard to control. β-actin and Actin were used as loading controls.</p>
Full article ">
13 pages, 5296 KiB  
Article
Natural Product Auraptene Targets SLC7A11 for Degradation and Induces Hepatocellular Carcinoma Ferroptosis
by Donglin Li, Yingping Li, Liangjie Chen, Chengchang Gao, Bolei Dai, Wenjia Yu, Haoying Yang, Junxiang Pi and Xueli Bian
Antioxidants 2024, 13(8), 1015; https://doi.org/10.3390/antiox13081015 - 20 Aug 2024
Cited by 2 | Viewed by 1075
Abstract
The natural product auraptene can influence tumor cell proliferation and invasion, but its effect on hepatocellular carcinoma (HCC) cells is unknown. Here, we report that auraptene can exert anti-tumor effects in HCC cells via inhibition of cell proliferation and ferroptosis induction. Auraptene treatment [...] Read more.
The natural product auraptene can influence tumor cell proliferation and invasion, but its effect on hepatocellular carcinoma (HCC) cells is unknown. Here, we report that auraptene can exert anti-tumor effects in HCC cells via inhibition of cell proliferation and ferroptosis induction. Auraptene treatment induces total ROS and lipid ROS production in HCC cells to initiate ferroptosis. The cell death or cell growth inhibition of HCC cells induced by auraptene can be eliminated by the ROS scavenger NAC or GSH and ferroptosis inhibitor ferrostatin-1 or Deferoxamine Mesylate (DFO). Mechanistically, the key ferroptosis defense protein SLC7A11 is targeted for ubiquitin–proteasomal degradation by auraptene, resulting in ferroptosis of HCC cells. Importantly, low doses of auraptene can sensitize HCC cells to ferroptosis induced by RSL3 and cystine deprivation. These findings demonstrate a critical mechanism by which auraptene exhibits anti-HCC effects via ferroptosis induction and provides a possible therapeutic strategy for HCC by using auraptene or in combination with other ferroptosis inducers. Full article
(This article belongs to the Special Issue Antioxidant Capacity of Natural Products)
Show Figures

Figure 1

Figure 1
<p>Auraptene exerts anti-tumor effects in HCC cells. (<b>A</b>) The molecular formula of auraptene with a molecular weight of 298.38. (<b>B</b>) HCCLM3 and HLE cells were plated into a 96-well plate at a density of 20,000 cells/well and treated with the indicated concentrations of auraptene for 24 h. Cell viability was detected with CCK-8 reagent and the IC50 was calculated. (<b>C</b>) HLE and HCCLM3 cells treated with the indicated concentrations of auraptene for 24 h were stained with crystal violet and photographed. (<b>D</b>) HLE and HCCLM3 cells treated with the indicated concentrations of auraptene for 16 h were photographed. Scale bar: 200 μm. (<b>E</b>) HLE and HCCLM3 cells treated with the indicated concentrations of auraptene for 16 h were stained with PI for flow cytometry analysis. Calculated cell death rate (Top) and representative pictures (Bottom) are shown. Aura: Auraptene. (<b>E</b>) Data are represented as the mean ± SD (n = 3), **** <span class="html-italic">p</span> &lt; 0.0001 (one-way ANOVA).</p>
Full article ">Figure 2
<p>ROS induction is responsible for auraptene-induced cell growth inhibition and cell death. (<b>A</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene and 5 mM NAC or 5 mM GSH were stained with DCFH-DA for 1 h, followed by flow cytometry analysis. The calculated relative cellular ROS levels (Top) and histogram of flow cytometric pictures are shown (Bottom). (<b>B</b>) The cell viability of HCCLM3 and HLE cells treated with or without 100 μM auraptene, 5 mM NAC, or 5 mM GSH for 24 h was analyzed with CCK-8. (<b>C</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 5 mM NAC or 5 mM GSH for 24 h were stained with crystal violet and the photographs are shown. (<b>D</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 5 mM NAC, or 5 mM GSH for 16 h were photographed and the representative images are shown. Scale bar: 200 μm. (<b>E</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 5 mM NAC, or 5 mM GSH for 16 h were harvested and stained with 10 μg/mL PI followed by flow cytometry analysis. Calculated cell death rate (left) and representative flow cytometric pictures (right) are shown. (<b>A</b>,<b>B</b>,<b>E</b>) Data are represented as the mean ± SD (n = 3); ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 (one-way ANOVA).</p>
Full article ">Figure 3
<p>Auraptene induces HCC cell ferroptosis. (<b>A</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 2 μM Fer-1, or 50 μM DFO for 4 h were incubated with the ROS probe DCFH-DA for 1 h followed by flow cytometry analysis. The calculated total ROS levels (Top) and histogram of flow cytometric pictures (Bottom) are shown. (<b>B</b>) HCCLM3 and HLE cells treated with or without auraptene (100 μM) for 10 h were incubated with lipid ROS probe C11-BODIPY 581/591 for 1 h followed by flow cytometry analysis. The calculated lipid ROS levels (Top) and histogram of flow cytometric pictures (Bottom) are shown. (<b>C</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 2 μM Fer-1, or 50 μM DFO for 24 h were stained with crystal violet and photographed; the images are shown. (<b>D</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 2 μM Fer-1 or 50 μM DFO for 24 h were incubated with CCK-8 followed by an analysis with a microplate reader. (<b>E</b>,<b>F</b>) HCCLM3 and HLE cells treated with or without 100 μM auraptene, 2 μM Fer-1 or 50 μM DFO for 16 h were photographed. Scale bar: 200 μm. (<b>E</b>) or stained with PI followed by analysis with flow cytometry. (<b>F</b>) The calculated cell death rate (<b>F</b>, <b>Top</b>) and the representative flow cytometric pictures (<b>F</b>, <b>Bottom</b>) are shown. (<b>A</b>,<b>B</b>,<b>D</b>,<b>F</b>) Data are represented as the mean ± SD (n = 3); *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (Unpaired Student’s <span class="html-italic">t</span> test for (<b>B</b>) and one-way ANOVA for (<b>A</b>,<b>D</b>,<b>F</b>)).</p>
Full article ">Figure 4
<p>A low dose of auraptene sensitizes HCC cells to ferroptosis. (<b>A</b>) HCCLM3 and HLE cells treated with or without the indicated concentrations of auraptene, RSL3 (2 μM) for 24 h, or cystine deprivation for 36 h were stained with crystal violet and photographed; the images are shown. (<b>B</b>) HCCLM3 and HLE cells treated with or without the indicated concentrations of auraptene, RSL3 (2 μM) for 24 h, or cystine deprivation for 36 h were photographed and the representative images are shown. Scale bar: 200 μm. (<b>C</b>) HCCLM3 and HLE cells treated with or without indicated concentration of auraptene or RSL3 (2 μM) for 24 h were stained with PI followed by flow cytometry analysis. The calculated cell death rate (left) and the representative flow cytometric pictures (right) are shown. (<b>D</b>) HCCLM3 and HLE cells treated with or without the indicated concentrations of auraptene or cystine deprivation for 36 h were stained with PI followed by flow cytometry analysis. The calculated cell death rate (left) and the representative flow cytometric pictures (right) are shown. (<b>C</b>,<b>D</b>) Data are represented as the mean ± SD (n = 3); ns: no significance, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 (one-way ANOVA).</p>
Full article ">Figure 5
<p>Auraptene degrades SLC7A11. (<b>A</b>) HCCLM3 and HLE cells treated with indicated concentrations of auraptene for 10 h were harvested for WB analysis with indicated antibodies, with Vinculin as the loading control. (<b>B</b>) HLE cells treated with 100 μM auraptene at the indicated time points were harvested for WB analysis with indicated antibodies, with Vinculin as the loading control. (<b>C</b>) HLE cells were pretreated with 100 μM auraptene for 2 h and then treated with or without CHX (100 μg/mL) or MG132 (10 μM) for another 8 h, then cells were harvested for WB analysis with indicated antibodies. (<b>A</b>–<b>C</b>) The data of SLC7A11/Vinculin are represented as the mean ± SD (n = 3); ns: no significance, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 (one-way ANOVA). (<b>D</b>) HLE cells were transfected with the specified plasmids for 15 h and then treated with or without 100 μM auraptene for 10 h. Cells were lysed for immunoprecipitated with anti-flag antibodies followed by Western blotting with the specified antibodies. (<b>E</b>) HCCLM3 and HLE cells were treated with or without 100 μM auraptene for 10 h and the cellular GSH levels were determined by a microplate reader at 412 nm. Data are represented as the mean ± SD (n = 3); **** <span class="html-italic">p</span> &lt; 0.0001 (Unpaired Student’s <span class="html-italic">t</span> test).</p>
Full article ">Figure 6
<p>The working model of auraptene in HCC ferroptosis induction. Auraptene, the major coumarin of citrus plants, targets SLC7A11 for ubiquitin–proteasomal degradation, leading to lipid ROS production and ferroptosis of HCC. Ub: ubiquitin.</p>
Full article ">
17 pages, 5061 KiB  
Article
Phytochemical Profiling Studies of Alkaloids and Coumarins from the Australian Plant Geijera parviflora Lindl. (Rutaceae) and Their Anthelmintic and Antimicrobial Assessment
by Deepika Dugan, Rachael J. Bell, Robert Brkljača, Colin Rix, Aya C. Taki, Robin B. Gasser and Sylvia Urban
Metabolites 2024, 14(5), 259; https://doi.org/10.3390/metabo14050259 - 30 Apr 2024
Viewed by 1378
Abstract
Phytochemical profiling followed by antimicrobial and anthelmintic activity evaluation of the Australian plant Geijera parviflora, known for its customary use in Indigenous Australian ceremonies and bush medicine, was performed. In the present study, seven previously reported compounds were isolated including auraptene, 6′-dehydromarmin, geiparvarin, [...] Read more.
Phytochemical profiling followed by antimicrobial and anthelmintic activity evaluation of the Australian plant Geijera parviflora, known for its customary use in Indigenous Australian ceremonies and bush medicine, was performed. In the present study, seven previously reported compounds were isolated including auraptene, 6′-dehydromarmin, geiparvarin, marmin acetonide, flindersine, and two flindersine derivatives from the bark and leaves, together with a new compound, chlorogeiparvarin, formed as an artefact during the isolation procedure and isolated as a mixture with geiparvarin. Chemical profiling allowed for a qualitative and quantitative comparison of the compounds in the leaves, bark, flowers, and fruit of this plant. Subsequently, a subset of these compounds as well as crude extracts from the plant were evaluated for their antimicrobial and anthelmintic activities. Anthelmintic activity assays showed that two of the isolated compounds, auraptene and flindersine, as well as the dichloromethane and methanol crude extracts of G. parviflora, displayed significant activity against a parasitic nematode (Haemonchus contortus). This is the first report of the anthelmintic activity associated with these compounds and indicates the importance of such fundamental explorations for the discovery of bioactive phytochemicals for therapeutic application(s). Full article
(This article belongs to the Section Plant Metabolism)
Show Figures

Figure 1

Figure 1
<p>Compounds from the bark and leaves of <span class="html-italic">Geijera parviflora</span>.</p>
Full article ">Figure 2
<p>HPLC-DAD comparison of <span class="html-italic">G. parviflora</span> DCM extracts at 220 nm (<b>top</b>) and 332 nm (<b>bottom</b>).</p>
Full article ">Figure 3
<p>HPLC-DAD comparison of <span class="html-italic">G. parviflora</span> MeOH extracts at 220 nm (<b>top</b>) and 332 nm (<b>bottom</b>).</p>
Full article ">Figure 4
<p>Dose-response curves of <span class="html-italic">G. parviflora</span> compounds on the motility of xL3s of <span class="html-italic">H. contortus</span> at 168 h.</p>
Full article ">
15 pages, 4104 KiB  
Article
Auraptene Enhances AMP-Activated Protein Kinase Phosphorylation and Thereby Inhibits the Proliferation, Migration and Expression of Androgen Receptors and Prostate-Specific Antigens in Prostate Cancer Cells
by Yasuyuki Akasaka, Shun Hasei, Yukino Ohata, Machi Kanna, Yusuke Nakatsu, Hideyuki Sakoda, Midori Fujishiro, Akifumi Kushiyama, Hiraku Ono, Akio Matsubara, Nobuyuki Hinata, Tomoichiro Asano and Takeshi Yamamotoya
Int. J. Mol. Sci. 2023, 24(21), 16011; https://doi.org/10.3390/ijms242116011 - 6 Nov 2023
Cited by 2 | Viewed by 1777
Abstract
Citrus hassaku extract reportedly activates AMPK. Because this extract contains an abundance of auraptene, we investigated whether pure auraptene activates AMPK and inhibits proliferation using prostate cancer cell lines. Indeed, auraptene inhibited the proliferation and migration of LNCaP cells and induced phosphorylation of [...] Read more.
Citrus hassaku extract reportedly activates AMPK. Because this extract contains an abundance of auraptene, we investigated whether pure auraptene activates AMPK and inhibits proliferation using prostate cancer cell lines. Indeed, auraptene inhibited the proliferation and migration of LNCaP cells and induced phosphorylation of AMPK or its downstream ACC in LNCaP, PC3, and HEK-293 cells, but not in DU145 cells not expressing LKB1. In addition, the mTOR-S6K pathway, located downstream from activated AMPK, was also markedly suppressed by auraptene treatment. Importantly, it was shown that auraptene reduced androgen receptor (AR) and prostate-specific antigen (PSA) expressions at both the protein and the mRNA level. This auraptene-induced downregulation of PSA was partially but significantly reversed by treatment with AMPK siRNA or the AMPK inhibitor compound C, suggesting AMPK activation to, at least partially, be causative. Finally, in DU145 cells lacking the LKB1 gene, exogenously induced LKB1 expression restored AMPK phosphorylation by auraptene, indicating the essential role of LKB1. In summary, auraptene is a potent AMPK activator that acts by elevating the AMP/ATP ratio, thereby potentially suppressing prostate cancer progression, via at least three molecular mechanisms, including suppression of the mTOR-S6K pathway, reduced lipid synthesis, and AR downregulation caused by AMPK activation. Full article
Show Figures

Figure 1

Figure 1
<p>Auraptene suppresses the proliferation and migration of prostate cancer LNCaP cells. (<b>a</b>) The effect of auraptene on the proliferation of LNCaP cells. Cells were treated with auraptene for 0, 24, or 48 h at the indicated concentrations (with or without DHT (10 nM)) (<span class="html-italic">n</span> = 4). (<b>b</b>–<b>d</b>) The effect of auraptene on the proliferation of (<b>b</b>) DU145, (<b>c</b>) PC3, and (<b>d</b>) HEK-293 cells (without DHT) (<span class="html-italic">n</span> = 4). (<b>e</b>,<b>f</b>) Determination of the IC<sub>50</sub> of auraptene against LNCaP and DU145 cells using CCK-8 assay. Cells were treated with auraptene for 24 h at the indicated concentrations, and then absorbances at 450 nm were measured. Absorbances relative to auraptene-untreated cells (red dots, <span class="html-italic">n</span> = 4) were used to calculate the IC<sub>50</sub> (blue lines: fitting curves). (<b>g</b>,<b>h</b>) Wound healing assay of LNCaP cells. Migration distances were measured under a microscope before and 96 h after administration of 30 μM auraptene (or vehicle as a control). Representative images (scale bar: 500 μm) (<b>g</b>) and quantification of wound closure (relative to control) (<span class="html-italic">n</span> = 4) (<b>h</b>). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 1 Cont.
<p>Auraptene suppresses the proliferation and migration of prostate cancer LNCaP cells. (<b>a</b>) The effect of auraptene on the proliferation of LNCaP cells. Cells were treated with auraptene for 0, 24, or 48 h at the indicated concentrations (with or without DHT (10 nM)) (<span class="html-italic">n</span> = 4). (<b>b</b>–<b>d</b>) The effect of auraptene on the proliferation of (<b>b</b>) DU145, (<b>c</b>) PC3, and (<b>d</b>) HEK-293 cells (without DHT) (<span class="html-italic">n</span> = 4). (<b>e</b>,<b>f</b>) Determination of the IC<sub>50</sub> of auraptene against LNCaP and DU145 cells using CCK-8 assay. Cells were treated with auraptene for 24 h at the indicated concentrations, and then absorbances at 450 nm were measured. Absorbances relative to auraptene-untreated cells (red dots, <span class="html-italic">n</span> = 4) were used to calculate the IC<sub>50</sub> (blue lines: fitting curves). (<b>g</b>,<b>h</b>) Wound healing assay of LNCaP cells. Migration distances were measured under a microscope before and 96 h after administration of 30 μM auraptene (or vehicle as a control). Representative images (scale bar: 500 μm) (<b>g</b>) and quantification of wound closure (relative to control) (<span class="html-italic">n</span> = 4) (<b>h</b>). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 2
<p>Auraptene induces AMPK activation in LNCaP cells. (<b>a</b>–<b>c</b>) LNCaP cells were treated with auraptene at the indicated concentrations for 8 h. AICAR (1 mM) was employed as a positive control. (<b>a</b>) AMPKα, phospho-AMPKα, ACC, phospho-ACC, and actin in LNCaP cells were detected using immunoblotting. (<b>b</b>,<b>c</b>) Quantification of relative band intensities (pAMPK/AMPK (<b>b</b>) and pACC/ACC (<b>c</b>)) (<span class="html-italic">n</span> = 4). (<b>d</b>) Western blot analysis. LNCaP cells were treated with 30 μM auraptene for 1, 2, 4, 8, or 24 h. (<b>e</b>) LNCaP cells were treated with 30 μM auraptene for 1 h, and the intracellular ADP/ATP ratio was determined using an ADP/ATP Ratio Assay Kit (<span class="html-italic">n</span> = 4). (<b>f</b>) Western blot analysis. LNCaP cells were treated with 30 μM auraptene for 1 h. (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>Auraptene induces AMPK activation in HEK-293 and PC3 cells but not in DU145 cells. (<b>a</b>,<b>c</b>,<b>e</b>) AMPKα, phosphor-AMPKα, ACC, phosphor-ACC, and actin were detected using immunoblotting after treatment with auraptene at the indicated concentrations for 8 h. AICAR (1 mM) was employed as a positive control. (<b>a</b>) HEK-293, (<b>c</b>) PC3, and (<b>e</b>) DU145. (<b>b</b>,<b>d</b>,<b>f</b>) Cells were treated with 30 μM auraptene for 1, 2, 4, 8, or 24 h, and immunoblotting was then performed. (<b>b</b>) HEK-293, (<b>d</b>) PC3, and (<b>f</b>) DU145.</p>
Full article ">Figure 4
<p>Auraptene attenuates the expressions of AR and PSA in LNCaP cells. (<b>a</b>) Protein levels of AR and PSA in LNCaP cells were examined using immunoblotting after treatment with 0, 3, 10, or 30 μM auraptene for 24 h. (<b>b</b>,<b>c</b>) Relative band intensities of (<b>b</b>) AR and (<b>c</b>) PSA protein levels are shown as bar graphs (<span class="html-italic">n</span> = 3). (<b>d</b>) Immunoblotting analysis of AR and PSA in LNCaP cells after treatment with 1 mM AICAR for 8 or 24 h. (<b>e</b>) LNCaP cells were treated with auraptene at 30 μM for 6 h, and the mRNAs of AR, PSA, and FKBP5 were examined using quantitative real-time PCR analysis. (<span class="html-italic">n</span> = 4–6). (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 5
<p>Auraptene-induced downregulation of PSA was partially reversed by suppressing AMPK. (<b>a</b>) Effects of the treatment with AMPK siRNA on the protein levels of AR and PSA. LNCaP cells were transfected with control siRNA or AMPK siRNA for 72 h and then treated with 10 μM of auraptene (or vehicle as a control) for 24 h. PSA, AR, AMPK, phosphor-AMPKα, ACC, phosphor-ACC, and actin were detected using immunoblotting. (<b>b</b>) Quantification of relative band intensities (PSA/actin) (<span class="html-italic">n</span> = 4). (<b>c</b>) Effects of compound C on the protein levels of AR and PSA. LNCaP cells were pre-treated with 5 μM of compound C for 1 h and then treated with 0, 3, or 10 μM of auraptene for 24 h. (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 6
<p>LKB1 is essential for auraptene to activate AMPK. (<b>a</b>) The protein expression profiles of AMPKα, phospho-AMPKα, ACC, phospho-ACC, LKB1, PSA, AR, CAMKK2, and actin were compared among LNCaP, DU145, and PC3 cells using immunoblotting. Notably, DU145 cells lack LKB1. (<b>b</b>) Effect of introduction of the LKB1 expression plasmid into DU145 cells lacking LKB1 on AMPKα phosphorylation. DU145 cells were transfected with pcDNA3.1(-) or MTF (myc-TEV-Flag-tagged)-LKB1 plasmids for 48 h, and then treated with vehicle or auraptene at 30 μM for 2 h. The cell lysates were subjected to immunoblotting. (<b>c</b>) Quantification of relative band intensities of pAMPK/AMPK (<span class="html-italic">n</span> = 3). (* <span class="html-italic">p</span> &lt; 0.05, n.s.: not significant).</p>
Full article ">
12 pages, 2144 KiB  
Article
Auraptene Has Antiviral Activity against Human Coronavirus OC43 in MRC-5 Cells
by Jung Sun Min, Young-Hee Jin and Sunoh Kwon
Nutrients 2023, 15(13), 2960; https://doi.org/10.3390/nu15132960 - 29 Jun 2023
Cited by 4 | Viewed by 2292
Abstract
Auraptene (7-geranyloxycoumarin) is the abundant prenyloxycoumarin found in the fruits of Citrus spp. Auraptene has a variety of pharmacological and therapeutic functions, such as anticancer, antioxidant, immunomodulatory, and anti-inflammation activities, with excellent safety profiles. In this study, we evaluated the anticoronaviral activity of [...] Read more.
Auraptene (7-geranyloxycoumarin) is the abundant prenyloxycoumarin found in the fruits of Citrus spp. Auraptene has a variety of pharmacological and therapeutic functions, such as anticancer, antioxidant, immunomodulatory, and anti-inflammation activities, with excellent safety profiles. In this study, we evaluated the anticoronaviral activity of auraptene in HCoV-OC43-infected human lung fibroblast MRC-5 cells. We found that auraptene effectively inhibited HCoV-OC43-induced cytopathic effects with 4.3 μM IC50 and 6.1 μM IC90, resulting in a selectivity index (CC50/IC50) of >3.5. Auraptene treatment also decreased viral RNA levels in HCoV-OC43-infected cells, as detected through quantitative real-time PCR, and decreased the expression level of spike proteins and nucleocapsid proteins in virus-infected cells, as detected through the Western blot analysis and immunofluorescence staining. Time-of-addition analysis showed auraptene’s inhibitory effects at the post-entry stage of the virus life cycle; however, auraptene did not induce the antiviral interferon families, IFN-α1, IFN-β1, and IFN-λ1. Additionally, auraptene-treated MRC-5 cells during HCoV-OC43 infection decreased the MMP-9 mRNA levels which are usually increased due to the infection, as auraptene is a previously reported MMP-9 inhibitor. Therefore, auraptene showed antiviral activity against HCoV-OC43 infection, and we suggest that auraptene has the potential to serve as a therapeutic agent against human coronavirus. Full article
(This article belongs to the Special Issue Nutraceuticals and Human Health and Disease)
Show Figures

Figure 1

Figure 1
<p>HCoV-OC43-induced cytopathic effects were inhibited by auraptene treatment in human lung fibroblast cells, MRC-5. (<b>A</b>) Chemical structure of auraptene. (<b>B</b>) MRC-5 cells were infected with 10<sup>4.5</sup> TCID<sub>50</sub>/mL HCoV-OC43 and treated with serially diluted concentrations of auraptene from 1.3 to 15 μM. At 4 days postinfection, an MTS assay was conducted to detect cell viability (left graph). The IC<sub>50</sub> and IC<sub>90</sub> of auraptene were 4.3 μM and 6.1 μM, respectively, as calculated via nonlinear regression analysis (right graph). Data were analyzed using a one-way analysis of variance followed by Bonferroni’s multiple comparisons test (F<sub>7,16</sub> = 120.5, <span class="html-italic">p</span> &lt; 0.0001; **** <span class="html-italic">p</span> &lt; 0.0001 vs. 0 μM of auraptene). (<b>C</b>) Optical microscope images of mock, 10 μM auraptene-treated cells, HCoV-OC43-infected cells, or virus-infected and 10 μM auraptene-treated cells at 4 days postinfection. Data are presented as mean ± SEM and represent three independent experiments.</p>
Full article ">Figure 2
<p>HCoV-OC43 replication and the expression of viral protein were inhibited by 10 μM auraptene treatment. (<b>A</b>) Extracellular (left graph) and intracellular viral RNA levels (right graph) in HCoV-OC43-infected MRC-5 cells treated with (circle) or without (rectangle) 10 μM auraptene assessed using qRT-PCR at 1, 2, 3, and 4 days postinfection. Data were analyzed using two-way ANOVA followed by Bonferroni’s multiple comparisons test (extracellular: time effect, F<sub>4,8</sub> = 3050, <span class="html-italic">p</span> &lt; 0.0001; auraptene effect, F<sub>1,8</sub> = 5590, <span class="html-italic">p</span> &lt; 0.0001; time × auraptene interaction, F<sub>4,8</sub> = 1690, <span class="html-italic">p</span> &lt; 0.0001; intracellular: time effect, F<sub>4,8</sub> = 273.4, <span class="html-italic">p</span> &lt; 0.0001; auraptene effect, F<sub>1,8</sub> = 1126, <span class="html-italic">p</span> &lt; 0.0001; time × auraptene interaction, F<sub>4,8</sub> = 281.4, <span class="html-italic">p</span> &lt; 0.0001; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 vs. HCoV-OC43 at the same day postinfection). Data are presented as mean ± SEM. (<b>B</b>) Expression of viral spike (S) proteins and nucleocapsid (N) protein in MRC-5 cells with HCoV-OC43 infection and treatment of vehicle or 10 μM auraptene at 1, 2, 3, and 4 days postinfection according to the Western blot analysis. The β-actin band was detected as a loading control. Data are representative of three independent experiments. (<b>C</b>) Immunofluorescence staining images at 1, 2, and 3 days postinfection. HCoV-OC43 spike protein (red) and nucleus visualized with DAPI (blue) (upper panel), and N protein (green) and nucleus visualized with DAPI (blue) (lower panel) in MRC-5 cells virus-infected with vehicle or 10 μM auraptene treatment. Scale bar = 100 μm. The experiments were performed independently ≥3 times.</p>
Full article ">Figure 3
<p>Time-of-addition assay of auraptene during HCoV-OC43 infection in MRC-5 cells. (<b>A</b>) Scheme for the time-of-addition assay. (<b>B</b>–<b>D</b>) Cell viability of pretreatment (<b>B</b>), cotreatment (<b>C</b>), and posttreatment (<b>D</b>) assay assessed using MTS at 4 days postinfection. Data were analyzed using a one-way analysis of variance (ANOVA) followed by Bonferroni’s multiple comparisons test (PRE-treatment: F<sub>10,22</sub> = 1.978, <span class="html-italic">p</span> = 0.0877; CO-treatment: F<sub>10,22</sub> = 1.232, <span class="html-italic">p</span> = 0.3251; POST-treatment: F<sub>10,22</sub> = 10.61, <span class="html-italic">p</span> &lt; 0.0001; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 and **** <span class="html-italic">p</span> &lt; 0.0001 vs. 0 μM of auraptene). Data are presented as mean ± SEM and representative of three independent experiments.</p>
Full article ">Figure 4
<p>IFN-related antiviral response was not induced by auraptene treatment during HCoV-OC43 infection. (<b>A</b>–<b>C</b>) Level of IFN-α1 (<b>A</b>), IFN-β1 (<b>B</b>), and IFN-λ1 (<b>C</b>) mRNA induction quantified in HCoV-OC43-infected MRC-5 cells with vehicle (pink bar) or 4, 7, or 10 μM auraptene (blue bar) treatment using qRT-PCR at 1, 2, 3, and 4 days postinfection. Data were normalized to β-actin gene expression level. Data were analyzed using one-way ANOVA followed by Bonferroni’s multiple comparisons test (IFN-α1: 2 days postinfection, F<sub>3,4</sub> = 18177, <span class="html-italic">p</span> &lt; 0.0001; 3 days postinfection, F<sub>3,4</sub> = 713.7, <span class="html-italic">p</span> &lt; 0.0001; 4 days postinfection, F<sub>3,4</sub> = 962.1, <span class="html-italic">p</span> &lt; 0.0001; IFN-β1: 2 days postinfection, F<sub>3,4</sub> = 46.39, <span class="html-italic">p</span> &lt; 0.0014; 3 days postinfection, F<sub>3,4</sub> = 804.6, <span class="html-italic">p</span> &lt; 0.0001; 4 days postinfection, F<sub>3,4</sub> = 31.04, <span class="html-italic">p</span> &lt; 0.0031; IFN-λ1: 2 days postinfection, F<sub>3,4</sub> = 670.4, <span class="html-italic">p</span> &lt; 0.0001; 3 days postinfection, F<sub>3,4</sub> = 472.7, <span class="html-italic">p</span> &lt; 0.0001; 4 days postinfection, F<sub>3,4</sub> = 810.5, <span class="html-italic">p</span> &lt; 0.0001; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and **** <span class="html-italic">p</span> &lt; 0.0001 vs. HCoV-OC43 at the same day postinfection). Data are presented as means ± SEMs and represent three independent experiments.</p>
Full article ">Figure 5
<p>Auraptene treatment decreased the induced MMP-9 expression by HCoV-OC43 infection. (<b>A</b>,<b>B</b>) Level of MMP-9 (<b>A</b>) and MMP-2 (<b>B</b>) mRNA quantified in HCoV-OC43-infected MRC-5 cells treated with vehicle (pink bar) or 4, 7, or 10 μM auraptene (blue bar) using qRT-PCR at 1, 2, 3, and 4 days postinfection. Data were normalized to β-actin gene expression level. Data were analyzed using one-way ANOVA followed by Bonferroni’s multiple comparisons test (MMP-9: 2 days postinfection, F<sub>3,4</sub> = 1093, <span class="html-italic">p</span> &lt; 0.0001; 3 days postinfection, F<sub>3,4</sub> = 811.7, <span class="html-italic">p</span> &lt; 0.0001; 4 days postinfection, F<sub>3,4</sub> = 527.3, <span class="html-italic">p</span> &lt; 0.0001; **** <span class="html-italic">p</span> &lt; 0.0001 vs. HCoV-OC43 at the same day postinfection). Data are presented as means ± SEMs and represent three independent experiments.</p>
Full article ">
14 pages, 1998 KiB  
Article
Protection of Mitochondrial Potential and Activity by Oxyprenylated Phenylpropanoids
by Francesco Epifano, Salvatore Genovese, Lucia Palumbo, Chiara Collevecchio and Serena Fiorito
Antioxidants 2023, 12(2), 259; https://doi.org/10.3390/antiox12020259 - 23 Jan 2023
Cited by 3 | Viewed by 1594
Abstract
A series of five naturally occurring oxyprenylated phenylpropanoids, namely, the coumarins auraptene (7-geranyloxycoumarin) 1 and 7-isopentenyloxycoumarin 2, and the coumaric acid and ferulic acid derivatives, 4’-isopentenyloxycoumaric acid 3, boropinic acid 4, and 4’-geranyloxyferulic acid 5 were tested for their effects [...] Read more.
A series of five naturally occurring oxyprenylated phenylpropanoids, namely, the coumarins auraptene (7-geranyloxycoumarin) 1 and 7-isopentenyloxycoumarin 2, and the coumaric acid and ferulic acid derivatives, 4’-isopentenyloxycoumaric acid 3, boropinic acid 4, and 4’-geranyloxyferulic acid 5 were tested for their effects on mitochondrial functionality using the organophosphate pesticides glyphosate and chlorpyrifos, and resveratrol, as the reference. While not showing an appreciable in vitro antioxidant activity, and virtually no or a little effect on the viability of non-cancer cell lines BEAS-2B and SHSY-5Y, all phytochemicals exhibited a marked protective effect on mitochondrial potential and activity, with values that were comparable to resveratrol. Auraptene 1 and 7-isopentenyloxycoumarin 2 were seen to be the most effective secondary metabolite to this concern, in particular in being able to completely abolish the decrease of mitochondrial potential induced by increasing concentration of both glyphosate and chlorpyrifos. All the compounds tested also exhibited a protective effect on mitochondrial activity. The potency displayed will shed more light on the molecular basis of the beneficial effects of auraptene, 7-isopentenyloxycoumarin, and structurally related oxyprenylated phenylpropanoids reported to date in the literature. Full article
Show Figures

Figure 1

Figure 1
<p>Structures of the phytochemicals under investigation. Compounds belong to two phenylpropanoids subclasses, such as <span class="html-italic">O</span>-prenylcoumarins (<b>1–2</b>), and cinnamic acid derivatives (<b>3–5</b>).</p>
Full article ">Figure 2
<p>Effects of compounds <b>1–5</b> on cell viability in non-cancer cells (REF = resveratrol) BEAS-2B (human bronchial epithelium) and SHSY-5Y (neuronal cells). Values expressed as mean (<span class="html-italic">n</span> = 6) ± SD. * <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Antioxidant properties of compounds <b>1–5</b> (ORAC assay).</p>
Full article ">Figure 4
<p>Effects of compounds <b>1–5</b> on mitochondrial destabilization (RSV = resveratrol), Values expressed as mean (<span class="html-italic">n</span> = 6) ± SD (* Significance non-treated vs. chlorpyrifos or glyphosate; ° Significance Ctrl vs. compounds).</p>
Full article ">Figure 5
<p>Effects of compounds <b>1–5</b> on mitochondrial activity (RSV = resveratrol) (* Significance non-treated vs. chlorpyrifos or glyphosate + compounds under investigation; ° Significance Ctrl vs. compounds).</p>
Full article ">Figure 6
<p>Effects of glyphosate and chlorpyrifos as individual compounds and in combination with compounds <b>1</b>, <b>2</b>, and resveratrol (REF) (all at the concentration level of 50 µM) on BEAS-2B cell viability (* Significance non-treated vs. chlorpyrifos or glyphosate + compounds under investigation).</p>
Full article ">
17 pages, 3371 KiB  
Article
The Antioxidant Auraptene Improves Aged Oocyte Quality and Embryo Development in Mice
by Yun-Hee Kim, Su-Yeon Lee, Eun-Young Kim, Kyeoung-Hwa Kim, Mi-Kyoung Koong and Kyung-Ah Lee
Antioxidants 2023, 12(1), 87; https://doi.org/10.3390/antiox12010087 - 30 Dec 2022
Cited by 9 | Viewed by 3277
Abstract
Decrease in quality of postovulatory aged oocytes occurs due to oxidative stress and leads to low fertilization and development competence. It is one of the main causes that exerting detrimental effect on the success rate in assisted reproductive technology (ART). Auraptene (AUR), a [...] Read more.
Decrease in quality of postovulatory aged oocytes occurs due to oxidative stress and leads to low fertilization and development competence. It is one of the main causes that exerting detrimental effect on the success rate in assisted reproductive technology (ART). Auraptene (AUR), a citrus coumarin, has been reported to possess an antioxidant effects in other tissues. In this study, we aimed to confirm the potential of AUR to delay the oocyte aging process by alleviating oxidative stress. Superovulated mouse oocytes in metaphase of second meiosis (MII) were exposed to 0, 1 or 10 μM AUR for 12 h of in vitro aging. AUR addition to the culture medium recovered abnormal spindle and chromosome morphology and mitigated mitochondrial distribution and mitochondrial membrane potential (ΔΨ) in aged oocytes. AUR-treated aged oocytes also showed suppressed oxidative stress, with lower reactive oxygen species (ROS) levels, higher glutathione (GSH) levels and increased expression of several genes involved in antioxidation. Furthermore, AUR significantly elevated the fertilization and embryo developmental rates. Oocytes aged with 1 μM AUR exhibited morphokinetics that were very similar to those of the control group. Altogether, these data allowed us to conclude that AUR improved the quality of aged oocytes and suggest AUR as an effective clinical supplement candidate to prevent postovulatory aging. Full article
Show Figures

Figure 1

Figure 1
<p>AUR protected the organizational dynamics of spindles and chromosomes during postovulatory aging. (<b>a</b>) A schematic diagram to illustrate the experimental design to investigate how AUR impact postovulatory aged oocytes. (<b>b</b>) Representative images of meiotic spindles and chromosomes in young and aged oocytes with or without AUR administration. Spindles were stained with α-tubulin (a spindle marker, green), and chromosomes were stained with DAPI (DNA, blue); scale bar = 20 μm. (<b>c</b>) Lengths of the spindles in each group of oocytes. Dots of boxplot show individual values of each observation. (<b>d</b>) Percentages of abnormal spindles in oocytes in each group. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.05, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>AUR affected mitochondrial distribution during postovulatory aging. (<b>a</b>) Representative images of mitochondrial distribution in young and aged oocytes with or without AUR administration. Oocytes were stained with MitoTracker (mitochondria marker, red), and chromosomes were stained with DAPI (DNA, blue); scale bar = 100 μm. (<b>b</b>) Percentages of abnormal mitochondrial distribution in each group of oocytes. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>AUR enhanced mitochondrial function by raising ΔΨm during postovulatory aging. (<b>a</b>) Representative images of mitochondrial ΔΨm in young and aged oocytes with or without AUR administration. Oocytes were stained with the cationic dye JC-1, which can selectively enter mitochondria and reversibly turn the emitted green fluorescence into red depending upon the mitochondrial membrane potential (aggregated JC-1: high potential marker, red; monomeric JC-1: low potential marker, green); scale bar = 20 μm. (<b>b</b>) The ratio of red to green fluorescence intensity in each group of oocytes. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>AUR decreased intracellular ROS and elevated GSH during postovulatory aging. (<b>a</b>) Representative images of reactive oxygen species (ROS) and glutathione (GSH) distribution in the oocytes in the Young, POA, POA+AUR 1 μM and POA+AUR 10 μM groups. Oocytes were stained with DCFH-DA (green) or CMF2HC (blue), scale bar = 20 μm. (<b>b</b>) Fluorescence intensities denoting intracellular ROS distribution in oocytes in all groups. (<b>c</b>) Fluorescence intensities of intracellular GSH staining distribution in oocytes in all groups. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>AUR upregulated the expression of NRF2-related genes during postovulatory aging. (<b>a</b>,<b>b</b>) Expression patterns of NRF2 mRNA and protein in postovulatory aged oocytes. (<b>c</b>–<b>h</b>) Transcription levels of NRF2-related genes (<span class="html-italic">Keap1</span>, <span class="html-italic">Gclc</span>, <span class="html-italic">Gclm</span>, <span class="html-italic">Gpx1</span>, <span class="html-italic">Sod1</span> and <span class="html-italic">Nqo1</span>), <span class="html-italic">p</span> &lt; 0.05. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>AUR promoted the fertilization and preimplantation development of postovulatory aged oocytes. (<b>a</b>) Representative images of blastocysts from young, aged and AUR-treated aged oocytes. Scale bar = 100 μm. (<b>b</b>,<b>c</b>) Quantitative analysis of embryo 2-cell and blastocyst formation rates. Data are presented as the means ± SEMs of at least three independent experiments. Significant difference indicated by lowercase letters of the alphabet, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>AUR advanced the morphokinetics of embryos from postovulatory aged oocytes. (<b>a</b>) Comparison of the morphokinetics of embryos derived from oocytes of the Young, POA, POA+AUR 1 μM and POA+AUR 10 μM groups. (<b>b</b>) Representative images of embryos from aged oocytes that had not reached the blastocyst stage. (<b>c</b>–<b>q</b>) t2, t3, t4, t5, t6, t7, and t8: time in hours post insemination (HPI) required for embryos to reach the 2-, 3-, 4-, 5-, 6-, 7-, and 8-cell stage, respectively; tErB: time to start formation of blastocoel cavity; tBL: half or more of blastocoel cavity had formed; CC: length of the cell cycle; S: synchronicity or round of cleavage division. Dots of boxplot show individual values of each observation. Data are presented as the means ± SEMs of at least three independent experiments, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 8
<p>Auraptene can effectively delay postovulatory oocyte aging in vitro by alleviating oxidative stress. Briefly, we found that AUR inhibited not only spindle defects but also mitochondrial dysfunctions. In addition, the AUR treatment showed exhibit high GSH level and increase expression of antioxidant enzymes (<span class="html-italic">Nrf2</span>, <span class="html-italic">Gclm</span>, <span class="html-italic">Gclc</span>, <span class="html-italic">Gpx1</span> and <span class="html-italic">Sod1</span>) while decreasing the ROS level. Moreover, AUR can improve fertilization and preimplantation embryo development. Thus, we conclude that AUR may be able to provide as a potent antioxidant in clinical.</p>
Full article ">
22 pages, 3802 KiB  
Article
LDL Promotes Disorders in β-Cell Cholesterol Metabolism, Implications on Insulin Cellular Communication Mediated by EVs
by Lizbeth Guevara-Olaya, Brenda Chimal-Vega, César Yahel Castañeda-Sánchez, Leslie Y. López-Cossio, Angel Pulido-Capiz, Octavio Galindo-Hernández, Raúl Díaz-Molina, Josefina Ruiz Esparza-Cisneros and Victor García-González
Metabolites 2022, 12(8), 754; https://doi.org/10.3390/metabo12080754 - 16 Aug 2022
Cited by 7 | Viewed by 2794
Abstract
Dyslipidemia is described as a hallmark of metabolic syndrome, promoting a stage of metabolic inflammation (metainflammation) that could lead to misbalances in energetic metabolism, contributing to insulin resistance, and modifying intracellular cholesterol pathways and the renin–angiotensin system (RAS) in pancreatic islets. Low-density lipoprotein [...] Read more.
Dyslipidemia is described as a hallmark of metabolic syndrome, promoting a stage of metabolic inflammation (metainflammation) that could lead to misbalances in energetic metabolism, contributing to insulin resistance, and modifying intracellular cholesterol pathways and the renin–angiotensin system (RAS) in pancreatic islets. Low-density lipoprotein (LDL) hypercholesterolemia could disrupt the tissue communication between Langerhans β-cells and hepatocytes, wherein extracellular vesicles (EVs) are secreted by β-cells, and exposition to LDL can impair these phenomena. β-cells activate compensatory mechanisms to maintain insulin and metabolic homeostasis; therefore, the work aimed to characterize the impact of LDL on β-cell cholesterol metabolism and the implication on insulin secretion, connected with the regulation of cellular communication mediated by EVs on hepatocytes. Our results suggest that β-cells can endocytose LDL, promoting an increase in de novo cholesterol synthesis targets. Notably, LDL treatment increased mRNA levels and insulin secretion; this hyperinsulinism condition was associated with the transcription factor PDX-1. However, a compensatory response that maintains basal levels of intracellular calcium was described, mediated by the overexpression of calcium targets PMCA1/4, SERCA2, and NCX1, together with the upregulation of the unfolded protein response (UPR) through the activation of IRE1 and PERK arms to maintain protein homeostasis. The LDL treatment induced metainflammation by IL-6, NF-κB, and COX-2 overexpression. Furthermore, LDL endocytosis triggered an imbalance of the RAS components. LDL treatment increased the intracellular levels of cholesterol on lipid droplets; the adaptive β-cell response was portrayed by the overexpression of cholesterol transporters ABCA1 and ABCG1. Therefore, lipotoxicity and hyperinsulinism induced by LDL were regulated by the natural compound auraptene, a geranyloxyn coumarin modulator of cholesterol-esterification by ACAT1 enzyme inhibition. EVs isolated from β-cells impaired insulin signaling via mTOR/p70S6Kα in hepatocytes, a phenomenon regulated by auraptene. Our results show that LDL overload plays a novel role in hyperinsulinism, mechanisms associated with a dysregulation of intracellular cholesterol, lipotoxicity, and the adaptive UPR, which may be regulated by coumarin-auraptene; these conditions explain the affectations that occur during the initial stages of insulin resistance. Full article
(This article belongs to the Special Issue The Role of β Cells in Diabetes)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>LDL endocytosis in β-cells is associated with the alteration of cholesterol targets. (<b>A</b>) RIN-m5F cells were treated with increasing concentrations of dil-LDL (0–40 µg/mL), representative images showing Hoescht (blue), dil-LDL (red), and merge for each treatment. (<b>B</b>) Flow cytometry analysis of dil-LDL endocytosis (0–20 µg/mL), under incubation for 20 h. PBS was used as a vehicle. (<b>C</b>) Effect of LDL treatment (0–40 µg/mL) on the expression of HMGCR and SREBP2 in β-cells, hepatocytes were evaluated as a control. (<b>D</b>) Densitometry analysis of SREBP2 in β-cells, results are reported as mean ± SD (<span class="html-italic">n</span> = 3), * <span class="html-italic">p</span> &lt; 0.05 concerning control. β-actin was used as a loading control.</p>
Full article ">Figure 2
<p>Insulin secretion promoted by LDL is associated with the modulation of targets controlling intracellular calcium levels. (<b>A</b>) Effect of LDL treatments (0–40 µg/mL) on insulin secretion (<span class="html-italic">n</span> = 3, mean ± SD), ** <span class="html-italic">p</span> &lt; 0.005. (<b>B</b>) Evaluation of insulin mRNA expression under the same LDL concentrations and 20 h of treatment, qPCR reactions were performed for triplicate, and GAPDH was used as a reference calibrator. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005. (<b>C</b>) Quantification of intracellular Ca<sup>2+</sup>. (<b>D</b>) Expression of PMCA1/4 and SERCA2 on cytoplasm lysates, representative Western blots are showed; quantitative characterization of the (<b>E</b>) SERCA2 and (<b>F</b>) PMCA1/4 expression (<span class="html-italic">n</span> = 3, mean ± SD), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005. (<b>G</b>) Western blot of NCX1 in cellular lysates; β-actin was used as a loading control.</p>
Full article ">Figure 3
<p>LDL modulates the extracellular insulin concentration through a transcriptional mechanism. (A) PDX-1 expression under LDL treatment (0–40 µg/mL). (B) Densitometry analysis of PDX-1 in β-cells, results are reported as mean ± SD (<span class="html-italic">n</span> = 3); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005 with respect to control. (C) Characterization of SERCA2 on cytosol (Cyt) and organelles (Org) isolates; for this experimentation, LDL (20 µg/mL) effect was evaluated. (D) Under this condition, insulin quantification was performed in organelles fraction; effect of KCl depolarization (30 mM) on intracellular Ca<sup>2+</sup> concentration (E) and insulin in extracellular media (F). Stimuli were performed for 40 min. (<b>D</b>–<b>F</b>) Results are reported as mean ± SD (<span class="html-italic">n</span> = 3), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.005. (G) Under the same condition, PDX-1 expression was characterized. In panels (<b>A</b>,<b>C</b>,<b>G</b>) β-actin was used as a loading control.</p>
Full article ">Figure 4
<p>In β-cells, activation of UPR induced by LDL is connected with inflammatory markers. (A) COX-2 expression under the LDL treatment (0–40 µg/mL) for 20 h in cytoplasm. Tunicamycin (Tum, 1 µg/mL) was used as an ER-stress inducer. (B) Densitometry analysis of COX-2, results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as % of control, * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.0005. (C) IL-6 and NF-κB expression under LDL increasing concentrations. (D) Effect of LDL treatment (0–40 µg/mL) on β-cell viability, results are reported as mean ± SD (<span class="html-italic">n</span> = 6), * <span class="html-italic">p</span> &lt; 0.05. (E) Under the same conditions, expression of apoptosis regulator, Bcl-2; (F) quantitative characterization of the Bcl-2 expression (<span class="html-italic">n</span> = 3, mean ± SD), * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005. (G) Western blot of the UPR targets XBP1s and CHOP under LDL stimuli (0–40 µg/mL) in nucleus isolates. Lamin B1 was used as a loading control. Tunicamycin (Tum) was used as a control (1 µg/mL). (H) PDI expression levels under the LDL treatment. (I) Densitometry analysis of PDI, results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as % of control. In panels A, C, E, and H, β-actin was used as a loading control.</p>
Full article ">Figure 5
<p>Effect of LDL treatment on renin–angiotensin system (RAS). (<b>A</b>) Western blot of HNF1α and ACE2 under increasing concentrations of LDL (0–40 µg/mL) at 20 h treatment. Quantitative characterization of HNF1α (<b>B</b>) and ACE2 (<b>C</b>), results are expressed as % of control, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005 (<span class="html-italic">n</span> = 3, mean ± SD). (<b>D</b>) Under the same experimental condition, expression of TMPRSS2 on cytoplasm extracts; (<b>E</b>) densitometry analysis of TMPRSS2 (<span class="html-italic">n</span> = 3, mean ± SD). GAPDH was used as a loading control. (<b>F</b>) AT<sub>1</sub>R expression levels under LDL incubation. (<b>G</b>) Densitometry analysis of AT<sub>1</sub>R, results are expressed as % of control, * <span class="html-italic">p</span> &lt; 0.05, **<span class="html-italic">p</span> &lt; 0.005 (<span class="html-italic">n</span> = 3, mean ± SD). In panels (<b>A</b>,<b>F</b>), β-actin was used as a loading control.</p>
Full article ">Figure 6
<p>LDL treatment promotes the intracellular accumulation of cholesterol and triggers ABCA1 and ABCG1 expression. Intracellular cholesterol concentrations (<b>A</b>) on fat droplets under treatment with LDL (0–20 µg/mL) at 20 h. Results are reported as mean ± SD (<span class="html-italic">n</span> = 3), * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.005. (B) Western blot of cholesterol transporters ABCA1 and ABCG1. β-actin was used as a loading control. Tunicamycin (Tum) was used as a control (1 µg/mL). (C) Densitometry analysis of ABCA1, results are reported as mean ± SD and expressed as % of control (<span class="html-italic">n</span> = 3), # <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005.</p>
Full article ">Figure 7
<p>Auraptene (Aur) treatment modulates insulin secretion through intracellular cholesterol metabolism in β-cells. (A) Characterization of ABCA1 under LDL (20 µg/mL) and concomitant treatment with Aur (4 and 8 µM) at 20 h, control treatments were performed with Aur. (B) Densitometry analysis of ABCA1, results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as % of control, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.005. (C) Effect of LDL (20 µg/mL) and Aur treatment (0–8 µM) on insulin secretion in extracellular media (<span class="html-italic">n</span> = 3, mean ± SD), * <span class="html-italic">p</span> &lt; 0.05 with respect to LDL treatment. (D) Under the same conditions, with ACAT1 expression in cellular lysates, β-actin was used as a loading control. DMSO was evaluated as a control. (E,F) Representation of the ACAT1 (blue, PDB: 6L47) binding to Aur (green) by molecular docking; the residues (pink) indicate the interactions with Aur. (G) Intracellular calcium quantification under LDL (20 µg/mL) and Aur treatment (4 µM) at 20 h of incubation; a statistical significance was not found. Under the same conditions, the cellular viability (H) and expression of PDX-1 by Western blot (I) were evaluated. (J) Densitometry analysis of PDX-1, results are reported as mean ± SD and expressed as % of control (<span class="html-italic">n</span> = 3), * <span class="html-italic">p</span> &lt; 0.05. In panels (<b>A</b>,<b>D</b>,<b>I</b>) β-actin was used as a loading control.</p>
Full article ">Figure 8
<p>The effect of extracellular vesicles (EVs) in β-cells on insulin signaling in hepatocytes. (A) Characterization of the purity of EVs isolated in extracellular media of β-cells was carried out through several steps; flotillin-2 was used as a marker. Pre-EVs: extracellular vesicles at 0 h; Post-EVs: extracellular vesicles at 20 h; FM-EVs: free medium of extracellular vesicles. β-cells and triple-negative breast cancer cells (TNBC) lysates were used as controls. In addition, Coomassie stain was performed on PVDF membranes. (B) Insulin quantification in EVs samples and extracellular media of β-cells employing the ultrasensitive insulin ELISA assay. (C) Characterization of insulin signaling in hepatocytes by p-p70S6kα, p70S6kα, eIF4E, and SREBP2 under the treatment of EVs derived from β-cells incubated with LDL (20 µg/mL) and concomitant Aur (4 µM) treatment, respectively. PBS was used as a control. Densitometry analysis of p-p70S6kα (D), p70S6kα (E), and SREBP2 (F). Results are reported as mean ± SD (<span class="html-italic">n</span> = 3) and expressed as % of control, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
12 pages, 765 KiB  
Article
Retrieval of High Added Value Natural Bioactive Coumarins from Mandarin Juice-Making Industrial Byproduct
by Eleni D. Myrtsi, Apostolis Angelis, Sofia D. Koulocheri, Sofia Mitakou and Serkos A. Haroutounian
Molecules 2021, 26(24), 7527; https://doi.org/10.3390/molecules26247527 - 12 Dec 2021
Cited by 3 | Viewed by 2727
Abstract
Cold pressed essential oil (CPEO) of mandarin (Citrus reticulata Blanco), a by-product of the juice-making industrial process known to contain large amounts of polymethoxyflavones, was exploited for its content in high added value natural coumarins. The study herein afforded a method referring [...] Read more.
Cold pressed essential oil (CPEO) of mandarin (Citrus reticulata Blanco), a by-product of the juice-making industrial process known to contain large amounts of polymethoxyflavones, was exploited for its content in high added value natural coumarins. The study herein afforded a method referring to the evaporation of CPEO volatile fraction under mild conditions (reduced pressure and temperature below 35 °C) as azeotrope with isopropanol. This allowed the isolation of high added value coumarins from the non-volatile fragment using preparative High Performance Liquid Chromatography (HPLC). Pilot-scale application of this procedure afforded for each kg of CPEO processed the following natural bioactive coumarins in chemically pure forms: heraclenol (38–55 mg), 8-gerayloxypsoralen (35–51 mg), auraptene (22–33 mg), and bergamottin (14–19 mg). The structures of coumarins were verified by Nuclear Magnetic Resonance (NMR) spectroscopy and HPLC co-injection with authentic standards. Thus, the low market value mandarin CPEO with current value of 17 to 22 EUR/kg can be valorized through the production of four highly bioactive natural compounds worth 3479 to 5057 EUR/kg, indicating the great potentials of this methodology in the terms of the circular economy. Full article
Show Figures

Figure 1

Figure 1
<p>HPLC preparative chromatogram of the isolated natural compounds. Peaks correspond to heraclenol (<b>1</b>, 40.7 min), 8-geranyloxypsoralene (<b>2</b>, 68.1 min), auraptene (<b>3</b>, 72.9 min), bergamottin (<b>4</b>, 76.5 min), nobiletin (<b>5</b>, 50.2 min), 5,6,7,4′-tetramethoxyflavone (<b>6</b>, 50.7 min), 3,5,6,7,8,3′,4′-heptamethoxyflavone (<b>7</b>, 51.9 min), and tangeretin (<b>8</b>, 56,3 min).</p>
Full article ">Figure 2
<p>Structures of retrieved natural coumarins and furanocoumarins. <b>1</b>: heraclenol; <b>2</b>: 8-geranyloxypsoralene; <b>3</b>: auraptene; <b>4</b>: bergamottin.</p>
Full article ">Figure 3
<p>Chemical structures of PMFs isolated from mandarin CPEO. <b>5</b>: nobiletin, <b>6</b>: 5,6,7,4′-tetramethoxyflavone, <b>7</b>: 3,5,6,7,8,3′4′-heptamethoxyflavone, and <b>8</b>: tangeretin.</p>
Full article ">
8 pages, 647 KiB  
Communication
Semisynthesis of Selenoauraptene
by Serena Fiorito, Francesco Epifano, Lorenzo Marchetti and Salvatore Genovese
Molecules 2021, 26(9), 2798; https://doi.org/10.3390/molecules26092798 - 10 May 2021
Cited by 2 | Viewed by 2335
Abstract
Selenium-containing compounds are gaining more and more interest due to their valuable and promising pharmacological properties, mainly as anticancer and antioxidant agents. Ebselen, the up to now only approved drugs, is well known to possess very good glutathione peroxidase mimicking effects. To date, [...] Read more.
Selenium-containing compounds are gaining more and more interest due to their valuable and promising pharmacological properties, mainly as anticancer and antioxidant agents. Ebselen, the up to now only approved drugs, is well known to possess very good glutathione peroxidase mimicking effects. To date, the most of efforts have been directed to build pure synthetic Se containing molecules, while less attention have been devoted to Se-based semisynthetic products resembling natural compounds like terpenes, polyphenols, and alkaloids. The aim of this short communication is to report the synthesis of the first example of a Se-phenylpropanoids, namely selenoauraptene, containing a selenogeranyl side chain in position 7 of the umbelliferone core. The key step was the Newman-Kwart rearrangement to obtain a selenocarbamate in which the Se atom was directly attached to umbelliferone (replacing its 7-OH function) followed by hydrolysis to get diumbelliferyl diselenide, which was finally easily converted to the desired Se-geranyl derivative in quite a good overall yield (28.5%). The synthesized adduct displayed a greater antioxidant and a radical scavenger in vitro activity than parent auraptene. The procedure we describe herein, to the best of our knowledge for the first time in the literature, represents an easy-to-handle method for the synthesis of a wide array of seleno analogues of naturally occurring biologically active oxyprenylated secondary metabolites. Full article
Show Figures

Figure 1

Figure 1
<p>Structure of auraptene (<b>1</b>) and selenoauraptene (<b>2</b>).</p>
Full article ">Figure 2
<p>Semisynthesis of selenoauraptene <b>2.</b> Reagents and conditions: (a) 2:1 molar ratio of NaBH<sub>4</sub> to powdered gray Se, iPrOH, 1 h, 5 °C; (b) CH<sub>2</sub>Cl<sub>2</sub>, 45 °C, 24 h; (c) iPrOH, 1.5 h, r.t., column chromatography; (d) neat, 200 °C; (e) KOH, MeOH, H<sub>2</sub>O, 24 h, r.t. (f) NaBH<sub>4</sub> (5 equiv.), iPrOH, geranyl bromide (1.1 equiv.), 1 h, r.t.; (g) crystallization (n-hexane).</p>
Full article ">Figure 3
<p>Monitoring the conversion of O-aryl selenocarbamate <b>6</b> into the Se-aryl carbamate <b>7</b> by <sup>1</sup>H NMR.</p>
Full article ">
13 pages, 2334 KiB  
Article
Auraptene Enhances Junction Assembly in Cerebrovascular Endothelial Cells by Promoting Resilience to Mitochondrial Stress through Activation of Antioxidant Enzymes and mtUPR
by Min Joung Lee, Yunseon Jang, Jiebo Zhu, Eunji Namgung, Dahyun Go, Changjun Seo, Xianshu Ju, Jianchen Cui, Yu Lim Lee, Hyoeun Kang, Hyeongseok Kim, Woosuk Chung and Jun Young Heo
Antioxidants 2021, 10(3), 475; https://doi.org/10.3390/antiox10030475 - 17 Mar 2021
Cited by 11 | Viewed by 3350
Abstract
Junctional proteins in cerebrovascular endothelial cells are essential for maintaining the barrier function of the blood-brain barrier (BBB), thus protecting the brain from the infiltration of pathogens. The present study showed that the potential therapeutic natural compound auraptene (AUR) enhances junction assembly in [...] Read more.
Junctional proteins in cerebrovascular endothelial cells are essential for maintaining the barrier function of the blood-brain barrier (BBB), thus protecting the brain from the infiltration of pathogens. The present study showed that the potential therapeutic natural compound auraptene (AUR) enhances junction assembly in cerebrovascular endothelial cells by inducing antioxidant enzymes and the mitochondrial unfolded protein response (mtUPR). Treatment of mouse cerebrovascular endothelial cells with AUR enhanced the expression of junctional proteins, such as occludin, zonula occludens-1 (ZO-1) and vascular endothelial cadherin (VE-cadherin), by increasing the levels of mRNA encoding antioxidant enzymes. AUR treatment also resulted in the depolarization of mitochondrial membrane potential and activation of mtUPR. The ability of AUR to protect against ischemic conditions was further assessed using cells deprived of oxygen and glucose. Pretreatment of these cells with AUR protected against damage to junctional proteins, including occludin, claudin-5, ZO-1 and VE-cadherin, accompanied by a stress resilience response regulated by levels of ATF5, LONP1 and HSP60 mRNAs. Collectively, these results indicate that AUR promotes resilience against oxidative stress and improves junction assembly, suggesting that AUR may help maintain intact barriers in cerebrovascular endothelial cells. Full article
(This article belongs to the Special Issue Natural Antioxidant in Cardiovascular and Cerebrovascular Diseases)
Show Figures

Figure 1

Figure 1
<p>Effects of Auraptene (AUR) on junctional proteins expression in bEnd.3 cells. (<b>A</b>) bEnd.3 cells (5 × 10<sup>3</sup> cells per well) seeded in 96-well plates were incubated in media containing 0, 1, 2, 4 µM of AUR for 24 h. Cell viability was measured by SRB assay. (<b>B</b>) bEnd.3 cells with vehicle (Veh) or 1 µM AUR were stained for ZO-1 (green) and DAPI (blue); Scale bar: 20 µm. Arrows indicate an increase of ZO-1 expression. (<b>C</b>) Relative ZO-1 intensity was quantified using ImageJ. (<b>D</b>) ZO-1, VE-cadherin and occludin expression were analyzed by Western blot after treatment of vehicle or 1 µM AUR for 24 h. (<b>E</b>–<b>G</b>) The protein levels of ZO-1, VE-cadherin and occludin were quantified using ImageJ. Data are presented as mean and ± SEM of three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 compared to Veh).</p>
Full article ">Figure 2
<p>Increase of genes encoding antioxidant enzymes in bEnd.3 cells by AUR treatment. (<b>A</b>,<b>B</b>) mRNA expressions for ROS scavenging antioxidant enzymes and GSH recycling-related genes were analyzed using qPCR with bEnd.3 cells treated with vehicle or 1 µM AUR. (<b>C</b>,<b>D</b>) bEnd.3 cells were incubated with vehicle or 1 µM AUR. The cells were stained with 5 µM CM-H<sub>2</sub>DCFDA or 5 µM MitoSOX<sup>TM</sup> and analyzed by flow cytometry. Total ROS was determined by DCFDA-stained cells (<b>C</b>) and mitochondrial ROS was determined by MitoSOX<sup>TM</sup>—Stained cells (<b>D</b>). Median fluorescence intensity (MFI) values are analyzed by FlowJo program. Data are presented as mean and ± SEM of three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared to Veh).</p>
Full article ">Figure 3
<p>Loss of mitochondrial membrane potential accompanying induction of mtUPR in bEnd.3 cells by AUR treatment. (<b>A</b>) Oxygen consumption rate (OCR) was measured in bEnd.3 cells treated with vehicle or 1 µM AUR for 24 h. (<b>B</b>) bEnd.3 cells were incubated with vehicle or 1 µM AUR. The cells were stained with 100 nM TMRE and analyzed by flow cytometry. (<b>C</b>) Mitochondrial membrane potential was determined by TMRE-stained cells. The Median fluorescence intensity (MFI) values are analyzed by FlowJo program. (<b>D</b>) Expression of mRNA for mitochondrial unfolded protein response (mtUPR) genes was examined after 1 µM AUR treatment using qPCR. (<b>E</b>) Expression of mRNA for mitochondrial unfolded protein response (mtUPR) genes was examined with bEnd.3 cells incubated in oxygen-glucose deprivation (OGD) condition for 3 h after pretreated vehicle or 1 µM AUR 24 h. Data are presented as mean and ± SEM of three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared to Veh, # <span class="html-italic">p</span> &lt; 0.05 compared to OGD).</p>
Full article ">Figure 4
<p>Protective effects of AUR pretreatment on the reduction of junctional protein by OGD in bEnd.3 cells.bEnd.3 cells were incubated in OGD condition for 3 h after treated with vehicle or 1 µM AUR 24 h. bEnd.3 cells were stained with claudin-5 (red) (<b>A</b>), ZO-1 (green) (<b>B</b>) and VE-cadherin (red) (<b>C</b>) with DAPI (blue); Scale bar: 20 µm. Arrows indicate junctional protein disruption. (<b>D</b>) Relative fluorescence intensity of markers was quantified using ImageJ. (<b>E</b>) ZO-1 and VE-cadherin expression were analyzed by Western blot with bEnd.3 cells incubated in OGD condition for 3 h after treatment of vehicle or 1 µM AUR for 24 h. Data are presented as mean and ± SEM of three independent experiments (* <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 compared to Veh, # <span class="html-italic">p</span> &lt; 0.05, ### <span class="html-italic">p</span> &lt; 0.001 compared to OGD).</p>
Full article ">Figure 5
<p>Schematic representation of the effect of AUR in normal condition and the protective mechanism in vitro ischemic injury model. AUR treatment can enhance junction assembly and induce resilience to oxidative stress in cerebrovascular endothelial cells by altering levels of mtUPR and antioxidant enzymes, resulting in low mitochondrial membrane potential. This reaction contributes mitochondrial stress resilience and it exhibits by alleviating degradation of junctional proteins when the cerebrovascular endothelial cells were put in OGD condition, in vitro ischemic injury model.</p>
Full article ">
12 pages, 952 KiB  
Article
Citrus hassaku Extract Powder Increases Mitochondrial Content and Oxidative Muscle Fibers by Upregulation of PGC-1α in Skeletal Muscle
by Shiori Akashi, Akihito Morita, Yusuke Mochizuki, Fuka Shibuya, Yasutomi Kamei and Shinji Miura
Nutrients 2021, 13(2), 497; https://doi.org/10.3390/nu13020497 - 3 Feb 2021
Cited by 6 | Viewed by 3265
Abstract
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is expressed in skeletal muscles and regulates systemic metabolism. Thus, nutraceuticals targeting skeletal muscle PGC-1α have attracted attention to modulate systemic metabolism. As auraptene contained in citrus fruits promotes lipid metabolism and improves mitochondrial respiration, it could increase [...] Read more.
Peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α) is expressed in skeletal muscles and regulates systemic metabolism. Thus, nutraceuticals targeting skeletal muscle PGC-1α have attracted attention to modulate systemic metabolism. As auraptene contained in citrus fruits promotes lipid metabolism and improves mitochondrial respiration, it could increase mitochondrial function through PGC-1α. Therefore, we hypothesized that PGC-1α is activated by auraptene and investigated its effect using Citrus hassaku extract powder (CHEP) containing >80% of auraptene. C2C12 myotubes were incubated with vehicle or CHEP for 24 h; C57BL/6J mice were fed a control diet or a 0.25% (w/w) CHEP-containing diet for 5 weeks. PGC-1α protein level and mitochondrial content increased following CHEP treatment in cultured myotubes and skeletal muscles. In addition, the number of oxidative fibers increased in CHEP-fed mice. These findings suggest that CHEP-mediated PGC-1α upregulation induced mitochondrial biogenesis and fiber transformation to oxidative fibers. Furthermore, as CHEP increased the expression of the protein sirtuin 3 and of phosphorylated AMP-activated protein kinase (AMPK) and the transcriptional activity of PGC-1α, these molecules might be involved in CHEP-induced effects in skeletal muscles. Collectively, our findings indicate that CHEP mediates PGC-1α expression in skeletal muscles and may serve as a dietary supplement to prevent metabolic disorders. Full article
(This article belongs to the Section Phytochemicals and Human Health)
Show Figures

Figure 1

Figure 1
<p>CHEP increases the amount of PGC-1α protein and mitochondria in C2C12 myotubes. Cells were incubated with vehicle (DMSO) or 37 μg/mL of CHEP for 24 h. (<b>a</b>) PGC-1α protein expression was evaluated using western blotting. Total PGC-1α protein levels were normalized to the total protein amount in the membrane as obtained after Ponceau S staining. Relative protein levels are expressed compared to those of cells treated with the vehicle. (<b>b</b>) CS activity was normalized to the protein content in the homogenate. (<b>c</b>) Mitochondrial DNA content, expressed as a percentage with respect to that in vehicle-treated cells. Relative mitochondrial DNA copy number was calculated as the ratio of <span class="html-italic">COX2</span> (mitochondrial) to <span class="html-italic">36B4</span> (nuclear) gene expression level using real-time PCR. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 3–7); * <span class="html-italic">p</span> &lt; 0.05 vs. vehicle. CHEP, <span class="html-italic">Citrus hassaku</span> extract powder; PGC-1α, peroxisome proliferator-activated receptor-γ coactivator-1α; CS, citrate synthase; <span class="html-italic">COX2</span>, cytochrome c oxidase subunit II; SEM, standard error of the mean.</p>
Full article ">Figure 2
<p>CHEP increases the amount of PGC-1α protein, mitochondria, and oxidative fibers in murine skeletal muscle. Seven-week-old male C57BL/6J mice were treated with an HC diet (control) or an HC diet containing 0.25% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) CHEP for 5 weeks. Protein expression levels of PGC-1α (<b>a</b>) and COX4 (<b>d</b>) in the gastrocnemius were evaluated using western blotting. Total PGC-1α and COX4 protein levels were normalized to the total protein amount on the membrane, obtained after Ponceau S staining. Relative protein levels were expressed compared to those of the control. (<b>b</b>) CS activity in the gastrocnemius was normalized to protein content in the homogenate. (<b>c</b>) Mitochondrial DNA content in the gastrocnemius is expressed as a percentage relative that in control. Relative mitochondrial DNA copy number was calculated as the ratio of <span class="html-italic">COX2</span> (mitochondrial) to <span class="html-italic">36B4</span> (nuclear) gene expression level, using real-time PCR. (<b>e</b>) Representative images of TA stained with anti-MHC type IIa (red), type IIb (green), and laminin (yellow) antibodies. Unlabeled fibers (black) are MHC IIx fibers. There are no MHC I (blue) fibers in this section. Scale bar: 100 μm. (<b>f</b>) Percentage of muscle fiber types in TA. Muscle fibers were counted in all cross sections based on immunofluorescence staining. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 5); * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 vs. control. HC, high carbohydrate; COX4, cytochrome c oxidase subunit IV; TA, tibialis anterior; MHC, myosin heavy chain.</p>
Full article ">Figure 3
<p>CHEP promotes SIRT3 expression, phosphorylation of AMPK, and PGC-1α transcription activity. Seven-week-old male C57BL/6J mice were fed an HC diet (control) or an HC diet containing 0.25% (<span class="html-italic">w</span>/<span class="html-italic">w</span>) CHEP, for 5 weeks. (<b>a</b>) Total SIRT3 protein levels in the gastrocnemius were measured using western blotting. The levels were normalized to the total protein amount on the membrane, obtained after Ponceau S staining. (<b>b</b>) The amount of phospho-AMPK (pAMPK) protein in the gastrocnemius was measured using western blotting. The levels were corrected by the amount of total AMPK protein. Relative protein levels were expressed compared to those of the control. Values are expressed as the mean ± SEM (<span class="html-italic">n</span> = 5); * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. control. (<b>c</b>) Transcriptional activation of PGC-1α by CHEP was measured using a reporter gene assay. HEK293T cells were co-transfected with pM-PGC-1α, pGL4.35, and pNL1.1.PGK. CHEP, at different concentrations (1, 3, 10, and 30 µg/mL), was added to the culture medium. The transcriptional activity of PGC-1α was analyzed using the Nano-Glo<sup>®</sup> luciferase assay. Mean values are indicated as the relative luciferase (Luc) activity, calculated as the ratio of firefly luciferase luminescence to NanoLuc luciferase luminescence. Values are expressed as mean ± SEM (<span class="html-italic">n</span> = 3); *** <span class="html-italic">p</span> &lt; 0.001 vs. 0 µg/mL. SIRT3, sirtuin 3; AMPK, AMP-activated protein kinase; HEK293T, human embryonic kidney 293T.</p>
Full article ">
14 pages, 5278 KiB  
Article
Regulation of Human Platelet Activation and Prevention of Arterial Thrombosis in Mice by Auraptene through Inhibition of NF-κB Pathway
by Chih-Wei Hsia, Ming-Ping Wu, Ming-Yi Shen, Chih-Hsuan Hsia, Chi-Li Chung and Joen-Rong Sheu
Int. J. Mol. Sci. 2020, 21(13), 4810; https://doi.org/10.3390/ijms21134810 - 7 Jul 2020
Cited by 10 | Viewed by 2955
Abstract
Platelets are major players in the occurrence of cardiovascular diseases. Auraptene is the most abundant coumarin derivative from plants, and it has been demonstrated to possess a potent capacity to inhibit platelet activation. Although platelets are anucleated cells, they also express the transcription [...] Read more.
Platelets are major players in the occurrence of cardiovascular diseases. Auraptene is the most abundant coumarin derivative from plants, and it has been demonstrated to possess a potent capacity to inhibit platelet activation. Although platelets are anucleated cells, they also express the transcription factor, nuclear factor-κB (NF-κB), that may exert non-genomic functions in platelet activation. In the current study, we further investigated the inhibitory roles of auraptene in NF-κB-mediated signal events in platelets. MG-132 (an inhibitor of proteasome) and BAY11-7082 (an inhibitor of IκB kinase; IKK), obviously inhibited platelet aggregation; however, BAY11-7082 exhibited more potent activity than MG-132 in this reaction. The existence of NF-κB (p65) in platelets was observed by confocal microscopy, and auraptene attenuated NF-κB activation such as IκBα and p65 phosphorylation and reversed IκBα degradation in collagen-activated platelets. To investigate cellular signaling events between PLCγ2-PKC and NF-κB, we found that BAY11-7082 abolished PLCγ2-PKC activation; nevertheless, neither U73122 nor Ro31-8220 had effect on NF-κB activation. Furthermore, both auraptene and BAY11-7082 significantly diminished HO• formation in activated platelets. For in vivo study, auraptene prolonged the occlusion time of platelet plug in mice. In conclusion, we propose a novel inhibitory pathway of NF-κB-mediated PLCγ2-PKC activation by auraptene in human platelets, and further supported that auraptene possesses potent activity for thromboembolic diseases. Full article
(This article belongs to the Special Issue Molecular Research on Platelet Activity in Health and Disease 2.0)
Show Figures

Figure 1

Figure 1
<p>Inhibitory profiles of NF-κB inhibitors and auraptene in platelet aggregation and ATP-release reaction stimulated by collagen in human platelets. Washed human platelets (3.6 × 10<sup>8</sup> cells/mL) were preincubated with MG-132 (50 and 100 μM), BAY11-7082 (5 and 10 μM) or auraptene (30 and 60 μM), followed by the addition of collagen (1 μg/mL) to trigger (<b>A</b>) platelet aggregation and (<b>B</b>) ATP-release reaction (AU; arbitrary unit). The corresponding statistical data are displayed on the below panel of each figure. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001, compared with the 0.1% DMSO-treated group.</p>
Full article ">Figure 2
<p>Effects of NF-κB activation by BAY11-7082, MG-132, and auraptene in platelets. (<b>A</b>) The confocal image (10 × 100 magnification) of NF-κB (p65) in resting (R) or collagen-activated (A) platelets. p65 or control IgG was labeled with goat anti-rabbit IgG-conjugated FITC (shown in green color) as described in Materials and Methods. For other experiments, washed platelets were preincubated with a solvent control (0.1% DMSO), BAY11-7082 (5 and 10 μM), MG-132 (50 and 100 μM), or auraptene (30 and 60 μM), followed by the addition of collagen (1 μg/mL) to trigger (<b>B</b>–<b>D</b>) IκBα and (<b>E</b>) p65 phosphorylation, or (<b>F</b>) IκBα protein degradation. Profiles (<b>A</b>) are representative examples of four similar experiments. The corresponding statistical data displayed in B–F. Data are presented as mean ± SEM (<span class="html-italic">n</span> = 4). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with the resting control (Tyrode’s solution); <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 and <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, compared with the 0.1% DMSO-treated group.</p>
Full article ">Figure 3
<p>Regulatory effects of NF-κB inhibitors and auraptene on PLCγ2 phosphorylation and PKC activation. Washed platelets were preincubated with 0.1% DMSO, auraptene (60 μM), BAY11-7082 (10 μM), U73122 (5 μM), or Ro31-8220 (2 μM) and then treated with collagen (1 μg/mL) or PDBu (150 nM) to trigger either (<b>A</b>) PLCγ2 phosphorylation or (<b>B</b>,<b>C</b>) PKC activation (p-p47). The corresponding statistical data are displayed on the right panel of each figure. Data are given as means ± SEM (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001, compared with the resting platelets (Tyrode’s solution); <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001, compared with the 0.1% DMSO-treated group.</p>
Full article ">Figure 4
<p>Regulatory profiles of auraptene and PLC/PKC inhibitors on IκBα protein degradation. Washed platelets were preincubated with 0.1% DMSO, auraptene (60 μM), BAY11-7082 (10 μM), U73122 (5 μM), or Ro31-8220 (2 μM) and then treated with collagen (1 μg/mL) to trigger (<b>A</b>,<b>B</b>) IκBα protein phosphorylation. The corresponding statistical data are displayed on the right panel of each figure. Data are given as means ± SEM (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt;0.01 and *** <span class="html-italic">p</span> &lt; 0.001, compared with the resting platelets (Tyrode’s solution); <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, compared with the 0.1% DMSO-treated group.</p>
Full article ">Figure 5
<p>Effects of auraptene on HO• formation in human platelets and vascular thrombosis in the mesenteric venules of mice. (<b>A</b>) For the electron spin resonance (ESR) study, washed platelets were incubated with Tyrode’s solution (<b>a</b>, resting group), solvent control (<b>b</b>, 0.1% DMSO), auraptene (<b>c</b>, 30 μM), or (<b>d</b>) BAY11-7082 (10 μM), followed by the addition of collagen (1 μg/mL) to trigger HO• (hydroxyl radical) formation. An asterisk (*) indicates the formation of HO•. Spectra are representative examples of four similar experiments. Data are given as means ± SEM (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt; 0.01, compared with the resting platelets (Tyrode’s solution); # p &lt; 0.05, compared with the 0.1% DMSO-treated group. (<b>B</b>) For animal study, mice were administered an intravenous bolus of the solvent control (0.1% DMSO) or auraptene (7.5 and 15 mg/kg), and the mesenteric venules were irradiated to induce microthrombus formation (occlusion time). Microscopic images (400× magnification) of 0.1% DMSO-treated groups and the auraptene (7.5 and 15 mg/kg)-treated groups were recorded at 5 and 150 s after irradiation, respectively. Data are given as means ± SEM (<span class="html-italic">n</span> = 8). ** <span class="html-italic">p</span> &lt; 0.01, compared with the 0.1% DMSO-treated group. The photographs are representative of eight similar experiments, and the arrows indicate platelet plug formation.</p>
Full article ">
8 pages, 1570 KiB  
Article
Citrus Auraptene Induces Expression of Brain-Derived Neurotrophic Factor in Neuro2a Cells
by Yoshiko Furukawa, Yu-suke Washimi, Ryu-ichi Hara, Mizuki Yamaoka, Satoshi Okuyama, Atsushi Sawamoto and Mitsunari Nakajima
Molecules 2020, 25(5), 1117; https://doi.org/10.3390/molecules25051117 - 3 Mar 2020
Cited by 11 | Viewed by 3323
Abstract
(1) Background: Our published data have indicated that (1) auraptene (AUR), a citrus ingredient, has neuroprotective effects on the mouse brain, owing to its ability to suppress inflammation, such as causing a reduction in hyperactivation of microglia and astrocytes; (2) AUR has the [...] Read more.
(1) Background: Our published data have indicated that (1) auraptene (AUR), a citrus ingredient, has neuroprotective effects on the mouse brain, owing to its ability to suppress inflammation, such as causing a reduction in hyperactivation of microglia and astrocytes; (2) AUR has the ability to trigger phosphorylation (activation) of extracellular signal-related kinase (ERK) and cAMP response element-binding protein (CREB) in neuronal cells; (3) AUR has the ability to induce glial cell line-derived neurotrophic factor (GDNF) synthesis/secretion in rat C6 glioma cells. The well-established fact that the ERK-CREB pathway plays an important role in the production of neurotrophic factors, including GDNF and brain-derived neurotrophic factor (BDNF), prompted us to investigate whether AUR would also have the ability to induce BDNF expression in neuronal cells. (2) Methods: Mouse neuroblastoma neuro2a cells were cultured and the effects of AUR on BDNF mRNA expression and protein content were evaluated by RT-PCR and ELISA, respectively. (3) Results: The levels of BDNF mRNA and secreted BDNF were significantly increased by AUR in a dose- and time-dependent manner in neuro2a cells. (4) Conclusion: The induction of BDNF in neuronal cells might be, in part, one of the mechanisms accounting for the neuroprotective effects of AUR. Full article
(This article belongs to the Section Medicinal Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of treatment with auraptene (AUR) on neuro2a cell viability. Cells were treated with various concentrations (10–80 μM) of AUR for 20 h (dotted bars) or with various concentrations (10–50 μM) of AUR for 40 h (hatched bar). The results represent the mean ± SEM (n = 4, different culture). Significance difference in values between the non-treated and AUR-treated cells: ** <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>Effects of treatment with AUR on brain-derived neurotrophic factor (BDNF) mRNA content in neuro2a cells. (<b>A</b>) Cells were incubated with (●) or without (○) 10 μM AUR for various times (0–50 h). (<b>B</b>) Cells were incubated with 10 or 20 μM AUR for 20 h. (<b>C</b>) Cells were incubated with 5 or 10 μM AUR for 40 h. Total RNA levels of untreated cells and of those treated with AUR were analyzed by the RT-PCR method. The results represent the mean ± SEM (n = 3, different culture). Significance difference in values between the non-treated and AUR-treated cells: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>Effects of treatment with AUR on BDNF content in medium conditioned by neuro2a cells. (<b>A</b>) Cells were incubated with 10, 30, or 50 μM AUR for 30 h. (<b>B</b>) Cells were incubated with 10 or 15 μM AUR for 50 h. The results represent the mean ± SEM (n = 3, different culture). Significance difference in values between the non-treated and AUR-treated cells: * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>Effect of U0126 on AUR-induced increase in BDNF mRNA content in neuro2a cells. Cells were preincubated with or without 10 μM U0126 for 30 min and then incubated with 20 μM AUR for 40 h. The results represent the mean ± SEM (n = 3, different culture). Significance difference in values between the non-treated and AUR-treated cells: * <span class="html-italic">p</span> &lt; 0.05; significant difference in values between the AUR-treated and AUR/inhibitor-treated cells: <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
Back to TopTop