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

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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (1,570)

Search Parameters:
Keywords = RAW 264.7 macrophages

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
25 pages, 13761 KiB  
Article
The Potential of a Novel Cold Atmospheric Plasma Jet as a Feasible Therapeutic Strategy for Gingivitis—A Cell-Based Study
by Andreea-Mariana Negrescu, Leonardo Zampieri, Emilio Martines and Anisoara Cimpean
Cells 2024, 13(23), 1970; https://doi.org/10.3390/cells13231970 - 28 Nov 2024
Viewed by 142
Abstract
Due to its antimicrobial, anti-inflammatory and pro-healing properties, the application of cold atmospheric plasma (CAP) has emerged as a new and promising therapeutic strategy in various fields of medicine, including general medicine and dentistry. In this light, the aim of the present study [...] Read more.
Due to its antimicrobial, anti-inflammatory and pro-healing properties, the application of cold atmospheric plasma (CAP) has emerged as a new and promising therapeutic strategy in various fields of medicine, including general medicine and dentistry. In this light, the aim of the present study was to investigate the effects of a homemade plasma jet on the cellular behaviour of two important cell types involved in gingivitis, namely gingival fibroblasts (HGF-1 cell line) and macrophages (RAW 264.7 cell line), by the direct application of CAP in different experimental conditions. The cellular behaviour of the HGF-1 cells was investigated in terms of viability/proliferation (LIVE/DEAD and CCK-8 assays), morphological features (immunofluorescent staining of the actin cytoskeleton) and fibronectin expression (immunocytochemical staining of the fibronectin network), while the macrophages’ response was evaluated through the assessment of the cellular survival/proliferation rate (LIVE/DEAD and CCK-8 assays), morphological behaviour (immunofluorescent staining of the actin cytoskeleton) and inflammatory activity (pro-inflammatory cytokine secretion profile (ELISA assay) and foreign body giant cells (FBGCs) formation (immunofluorescent staining of the actin cytoskeleton and multinuclearity index determination)). The in vitro biological assessment revealed an upward trend dependent on treatment time and number of CAP applications, in terms of fibroblasts proliferation (p < 0.0001) and fibronectin expression (p < 0.0001). On the other hand, the macrophages exposed to five consecutive CAP applications for longer treatment times (over 120 s) exhibited a strong pro-inflammatory activity, as evinced by their altered morphology, pro-inflammatory cytokine profile (p < 0.0001) and FBGCs formation. Overall, our results demonstrate that CAP exposure, when used with appropriate operating parameters, has a beneficial effect on the cellular response of HGF-1 and RAW 264.7 cells, thus paving the way for further in vitro and in vivo investigations that will allow the translation of CAP treatment from research to clinic as an alternative therapy for gingivitis. Full article
Show Figures

Figure 1

Figure 1
<p>CAP source during cell culture treatment.</p>
Full article ">Figure 2
<p>The interpolation of the nitrogen second positive system.</p>
Full article ">Figure 3
<p>The survival/proliferation potential of the HGF-1 cells exposed to single/multiple CAP applications, as assessed by (<b>a</b>) the LIVE/DEAD assay at 24 h after the final CAP treatment (live cells: green fluorescence; dead cells: red fluorescence). The size of the scale bar is 200 µm; (<b>b</b>) the CCK-8 technique results at the same time period of 24 h after the final CAP exposure. The results are expressed as means ± SD (n = 3, **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. TCPS; ●●●● <span class="html-italic">p</span> &lt; 0.0001, ●●● <span class="html-italic">p</span> &lt; 0.001 and ●● <span class="html-italic">p</span> &lt; 0.01 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 and ■ <span class="html-italic">p</span> &lt; 0.05 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001, ○○○ <span class="html-italic">p</span> &lt; 0.001, ○○ <span class="html-italic">p</span> &lt; 0.01 and ○ <span class="html-italic">p</span> &lt; 0.05 vs. 90 s; X <span class="html-italic">p</span> &lt; 0.05 vs. 120 s; # <span class="html-italic">p</span> &lt; 0.05 vs. 180 s). The significance level between the three groups: ♦♦♦♦ <span class="html-italic">p</span> &lt; 0.0001 vs. CAP single treatment; <b>^^^^</b> <span class="html-italic">p</span> &lt; 0.0001 vs. CAP multiple (3x) treatment.</p>
Full article ">Figure 4
<p>Effect of single (<b>a</b>)/multiple (<b>b</b>) CAP applications on the HGF-1 cells’ morphology, as assessed through the fluorescent labelling of the cytoskeletal proteins: actin (green fluorescence) and vinculin (red fluorescence). DAPI-labelled nuclei emit blue fluorescence. The size of the scale bar is 50 µm.</p>
Full article ">Figure 4 Cont.
<p>Effect of single (<b>a</b>)/multiple (<b>b</b>) CAP applications on the HGF-1 cells’ morphology, as assessed through the fluorescent labelling of the cytoskeletal proteins: actin (green fluorescence) and vinculin (red fluorescence). DAPI-labelled nuclei emit blue fluorescence. The size of the scale bar is 50 µm.</p>
Full article ">Figure 5
<p>(<b>a</b>) Immunofluorescent labelling of the fibronectin network synthesised and organised by the HGF-1 cells exposed to either single or multiple CAP applications (fibronectin network—green fluorescence; nuclei—blue fluorescence). Scale bar represents 50 µm. (<b>b</b>) Fluorescence intensity measurement (n = 10, mean ± SD, **** <span class="html-italic">p</span> &lt; 0.0001, *** <span class="html-italic">p</span> &lt; 0.001, ** <span class="html-italic">p</span> &lt; 0.01 and * <span class="html-italic">p</span> &lt; 0.05 vs. TCPS; ●●●● <span class="html-italic">p</span> &lt; 0.0001, ●●● <span class="html-italic">p</span> &lt; 0.001, ●● <span class="html-italic">p</span> &lt; 0.01 and ● <span class="html-italic">p</span> &lt; 0.05 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001, ■■ <span class="html-italic">p</span> &lt; 0.01 and ■ <span class="html-italic">p</span> &lt; 0.05 vs. 60 s). The significance level between the three groups: ♦♦♦♦ <span class="html-italic">p</span> &lt; 0.0001 vs. CAP single treatment; <b>^^^^</b> <span class="html-italic">p</span> &lt; 0.0001 vs. CAP multiple (3×) treatment.</p>
Full article ">Figure 6
<p>The survival potential of the RAW 264.7 cells exposed to either single or multiple CAP applications, as assessed by the LIVE/DEAD assay at 24 h after the final CAP treatment (live cells—green fluorescence; dead cells—red fluorescence) in both experimental culture conditions: (<b>a</b>) standard culture (−LPS); (<b>b</b>) macrophage activation with 100 ng mL<sup>−1</sup> LPS (+LPS). The size of the scale bar is 200 µm.</p>
Full article ">Figure 6 Cont.
<p>The survival potential of the RAW 264.7 cells exposed to either single or multiple CAP applications, as assessed by the LIVE/DEAD assay at 24 h after the final CAP treatment (live cells—green fluorescence; dead cells—red fluorescence) in both experimental culture conditions: (<b>a</b>) standard culture (−LPS); (<b>b</b>) macrophage activation with 100 ng mL<sup>−1</sup> LPS (+LPS). The size of the scale bar is 200 µm.</p>
Full article ">Figure 7
<p>The CCK-8 assay showing the proliferative status of the RAW 264.7 macrophages exposed to either single or multiple CAP applications under both standard (−LPS) and pro-inflammatory (+LPS) conditions (n = 3, mean ± SD, **** <span class="html-italic">p</span> &lt; 0.0001 vs. TCPS; ●●●● <span class="html-italic">p</span> &lt; 0.0001, ●● <span class="html-italic">p</span> &lt; 0.01 and ● <span class="html-italic">p</span> &lt; 0.05 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 and ■ <span class="html-italic">p</span> &lt; 0.05 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001 vs. 90 s; XXXX <span class="html-italic">p</span> &lt; 0.0001 vs. 120 s). The significance level between the three groups: ♦♦♦♦ <span class="html-italic">p</span> &lt; 0.0001 vs. CAP single treatment; <b>^^^^</b> <span class="html-italic">p</span> &lt; 0.0001 vs. CAP multiple (3x) treatment.</p>
Full article ">Figure 8
<p>The morphological features exhibited by the RAW 264.7 macrophages after CAP treatment under standard (−LPS) and pro-inflammatory (+LPS) culture conditions (green fluorescence—actin cytoskeleton). The size of the scale bar is 50 µm.</p>
Full article ">Figure 9
<p>Quantification of the pro-inflammatory cytokines’ extracellular secretion by the LPS-stimulated RAW 264.7 macrophages (100 ng/mL LPS) subjected to CAP-exposure. ELISA measurement of (<b>a</b>) IL-1β—CAP multiple (3x) treatment (**** <span class="html-italic">p</span> &lt; 0.0001 and *** <span class="html-italic">p</span> &lt; 0.001 vs. TCPS; •••• <span class="html-italic">p</span> &lt; 0.0001 and •• <span class="html-italic">p</span> &lt; 0.01 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 and ■■■ <span class="html-italic">p</span> &lt; 0.001 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001 and ○○○ <span class="html-italic">p</span> &lt; 0.001 vs.90 s; XXXX <span class="html-italic">p</span> &lt; 0.0001 and XX <span class="html-italic">p</span> &lt; 0.01 vs. 120 s; #### <span class="html-italic">p</span> &lt; 0.0001 vs. 180 s). (<b>b</b>) IL-1β—CAP multiple (5x) treatment (**** <span class="html-italic">p</span> &lt; 0.0001 and ** <span class="html-italic">p</span> &lt; 0.01 vs. TCPS; •••• <span class="html-italic">p</span> &lt; 0.0001 and • <span class="html-italic">p</span> &lt; 0.05 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001 vs. 90 s; XXXX <span class="html-italic">p</span> &lt; 0.0001 vs. 120 s; #### <span class="html-italic">p</span> &lt; 0.0001 vs. 180 s). (<b>c</b>) IL-6—CAP multiple (3x) treatment (**** <span class="html-italic">p</span> &lt; 0.0001 and * <span class="html-italic">p</span> &lt; 0.05 vs. TCPS; •••• <span class="html-italic">p</span> &lt; 0.0001 and •• <span class="html-italic">p</span> &lt; 0.01 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 and ■■ <span class="html-italic">p</span> &lt; 0.05 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001 and ○○ <span class="html-italic">p</span> &lt; 0.01 vs. 90 s; XXXX <span class="html-italic">p</span> &lt; 0.0001 and X <span class="html-italic">p</span> &lt; 0.05 vs. 120 s; #### <span class="html-italic">p</span> &lt; 0.0001 vs. 180 s). (<b>d</b>) IL-6—CAP multiple (5x) treatment (**** <span class="html-italic">p</span> &lt; 0.0001 and *** <span class="html-italic">p</span> &lt; 0.001 vs. TCPS; •••• <span class="html-italic">p</span> &lt; 0.0001 and •• <span class="html-italic">p</span> &lt; 0.0001 vs. 30 s; ■■■■ <span class="html-italic">p</span> &lt; 0.0001 and ■■ <span class="html-italic">p</span> &lt; 0.01 vs. 60 s; ○○○○ <span class="html-italic">p</span> &lt; 0.0001 and ○○ <span class="html-italic">p</span> &lt; 0.01 vs. 90 s; XXXX <span class="html-italic">p</span> &lt; 0.0001 vs. 120 s; #### <span class="html-italic">p</span> &lt; 0.0001 vs. 180 s). The results are expressed as means ± SD (n = 3).</p>
Full article ">Figure 10
<p>Fluorescent images of the multinucleated FBGCs generated through the RAW 264.7 macrophage fusion process after CAP treatment under stimulation with LPS (green fluorescence—actin cytoskeleton; blue fluorescence—nuclei). The size of the scale bar is 50 µm.</p>
Full article ">
14 pages, 1605 KiB  
Article
Hydroethanolic Extract of Polygonum aviculare L. Mediates the Anti-Inflammatory Activity in RAW 264.7 Murine Macrophages Through Induction of Heme Oxygenase-1 and Inhibition of Inducible Nitric Oxide Synthase
by Chan Ho Jang, You Chul Chung, Ami Lee and Youn-Hwan Hwang
Plants 2024, 13(23), 3314; https://doi.org/10.3390/plants13233314 - 26 Nov 2024
Viewed by 327
Abstract
Polygonum aviculare L. (PAL), commonly known as knotgrass, has been utilized as a traditional folk medicine across Asian, African, Latin American and Middle Eastern countries to treat various inflammatory diseases, including arthritis and airway inflammation. Numerous medicinal herbs exert anti-inflammatory and antioxidative effects [...] Read more.
Polygonum aviculare L. (PAL), commonly known as knotgrass, has been utilized as a traditional folk medicine across Asian, African, Latin American and Middle Eastern countries to treat various inflammatory diseases, including arthritis and airway inflammation. Numerous medicinal herbs exert anti-inflammatory and antioxidative effects that are mediated through the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2) and the inhibition of nuclear factor kappa B (NF-κB). However, the underlying molecular mechanisms linking the antioxidative and anti-inflammatory effects remain poorly understood. Heme oxygenase-1 (HO-1) is an antioxidant enzyme that catalyzes heme degradation, ultimately leading to the production of carbon monoxide (CO). Elevated levels of CO have been correlated with the decreased level of inducible nitric oxide synthase (iNOS). In this study, we examined whether HO-1 plays a key role in the relationship between the antioxidative and anti-inflammatory properties of PAL. The anti-inflammatory and antioxidative activities of PAL in an in vitro system were evaluated by determining NF-κB activity, antioxidant response element (ARE) activity, pro-inflammatory cytokine and protein levels, as well as antioxidant protein levels. To examine whether HO-1 inhibition interfered with the anti-inflammatory effect of PAL, we measured nitrite, reactive oxygen species, iNOS, and HO-1 levels in RAW 264.7 murine macrophages pre-treated with Tin protoporphyrin (SnPP, an HO-1 inhibitor). Our results demonstrated that PAL increased ARE activity and the Nrf2-regulated HO-1 level, exerting antioxidative activities in RAW 264.7 macrophages. Additionally, PAL reduced cyclooxygenase-2 (COX-2) and iNOS protein levels by inactivating NF-κB in lipopolysaccharide (LPS)-activated RAW 264.7 macrophages. Further investigation using the HO-1 inhibitor revealed that HO-1 inhibition promoted iNOS expression, subsequently elevating nitric oxide (NO) generation in LPS-activated RAW 264.7 macrophages treated with PAL compared to those in the macrophages without the HO-1 inhibitor. Overall, our findings suggest that HO-1 induction by PAL may exert anti-inflammatory effects through the reduction of the iNOS protein level. Hence, this study paves the way for further investigation to understand molecular mechanisms underlying the antioxidative and anti-inflammatory activities of medicinal herbs. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Inhibition of NF-κB activation by PAL hydroethanolic extract. Cell viability of PAL hydroethanolic extract in (<b>A</b>) NF-κB Luciferase Reporter-RAW 264.7 cells, and (<b>B</b>) RAW 264.7 macrophages was quantified using a CCK-8 assay. (<b>C</b>) Concentration-dependent inhibition of NF-κB luciferase activity by PAL hydroethanolic extract in LPS-activated NF-κB Luciferase Reporter-RAW 264.7 cells. (<b>D</b>) Expression level of nuclear NF-κB in LPS-activated RAW 264.7 macrophages were quantitatively analyzed. Data are presented as mean  ±  standard error of the mean (SEM) from three independent experiments (<span class="html-italic">N</span>  =  3). A statistical significance compared with LPS alone treatment at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was marked by an asterisk (*) and double asterisk (**), respectively. LPS, lipopolysaccharide; DEX, dexamethasone; PAL, <span class="html-italic">Polygonum aviculare</span> L.; NS, not significant.</p>
Full article ">Figure 2
<p>Anti-inflammatory effects of PAL hydroethanolic extract in LPS-activated RAW 264.7 macrophages. Expression levels of (<b>A</b>) COX-2 and (<b>B</b>) iNOS in LPS-activated RAW 264.7 macrophages were quantitatively analyzed. The levels of extracellular (<b>C</b>) PGE<sub>2</sub> and (<b>D</b>) NO were analyzed in LPS-activated RAW 264.7 macrophages. Data are presented as the mean  ±  SEM (<span class="html-italic">N</span>  =  3). A statistical significance compared with LPS alone treatment at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was marked by an asterisk (*) and double asterisk (**), respectively.</p>
Full article ">Figure 3
<p>Effects of PAL hydroethanolic extract on the production of pro-inflammatory cytokines in LPS-activated RAW 264.7 macrophages. Cellular inflammatory response was provoked by LPS in RAW 264.7 macrophages. Pro-inflammatory cytokines, including (<b>A</b>) IL-1β, (<b>B</b>) IL-6, and (<b>C</b>) TNF-α were analyzed in LPS-activated RAW 264.7 cells. Data are presented as the mean  ±  SEM (<span class="html-italic">N</span>  =  3). A statistical significance compared with LPS alone treatment at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was marked by an asterisk (*) and double asterisk (**), respectively.</p>
Full article ">Figure 4
<p>Antioxidant effects of PAL hydroethanolic extract by activation of Nrf2 signaling pathway in RAW 264.7 macrophages. Protein levels of (<b>A</b>) nuclear Nrf2 and (<b>B</b>) cytoplasmic HO-1 in RAW 264.7 macrophages were quantitatively analyzed. (<b>C</b>) ARE activity by PAL hydroethanolic extract in HepG2-ARE cells. (<b>D</b>) Intracellular ROS level by PAL hydroethanolic extract in RAW 264.7 macrophages. Data are presented as the mean  ±  SEM (<span class="html-italic">N</span>  =  3). A statistical significance compared with control group at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was marked by an asterisk (*) and double asterisk (**), respectively. SFN, sulforaphane; tBHP, tert-butyl hydroperoxide; tBHQ, tertiary-butylhydroquinone.</p>
Full article ">Figure 5
<p>HO-1 inhibition nullified anti-inflammatory effects by PAL hydroethanolic extract. (<b>A</b>) Cytoplasmic iNOS protein level was quantitatively analyzed in RAW 264.7 cells pre-treated with or without SnPP. (<b>B</b>) Extracellular NO level by PAL hydroethanolic extract in RAW 264.7 cells pre-treated with or without SnPP. (<b>C</b>) Cytoplasmic HO-1 protein level was quantitatively analyzed in RAW 264.7 cells pre-treated with or without SnPP. (<b>D</b>) Intracellular ROS level was analyzed in LPS-activated RAW 264.7 cells pre-treated with and without SnPP. Values are mean  ±  SEM (<span class="html-italic">N</span>  =  3). A significance difference compared with LPS alone or control group at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was indicated by an asterisk (*) and double asterisk (**), respectively. A hash (# <span class="html-italic">p</span> &lt; 0.05) and double hash (## <span class="html-italic">p</span> &lt; 0.01) indicate a significant difference between groups. SnPP, Tin Protoporphyrin IX dichloride.</p>
Full article ">Figure 6
<p>HO-1 inhibition abrogates anti-inflammatory effects by KAE and QUE abundant in PAL hydroethanolic extract. NF-κB luciferase activity by (<b>A</b>) KAE and (<b>B</b>) QUE in LPS-activated NF-κB Luciferase Reporter-RAW 264.7 cells pre-treated with or without SnPP. Extracellular NO level by (<b>C</b>) KAE and (<b>D</b>) QUE in LPS-activated RAW 264.7 macrophages pre-treated with and without SnPP. Data are presented as the mean  ±  SEM (<span class="html-italic">N</span>  =  3). A statistical significance compared with LPS alone group at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01 was marked by an asterisk (*) and double asterisk (**), respectively. A hash (# <span class="html-italic">p</span> &lt; 0.05) and double hash (## <span class="html-italic">p</span> &lt; 0.01) indicate a statistical significance between groups. NS, not significant.</p>
Full article ">
12 pages, 1728 KiB  
Article
Deodeokaloid, a New Indole Alkaloid N-Glycoside and Bioactive Phenolic Compounds from the Roots of Codonopsis lanceolata
by Yeo Rang Cho, Joo-Hyun Hong, Dong-Min Kang, Yoon-Joo Ko, Mi-Jeong Ahn and Ki Hyun Kim
Plants 2024, 13(22), 3243; https://doi.org/10.3390/plants13223243 - 19 Nov 2024
Viewed by 428
Abstract
Codonopsis lanceolata, commonly known as the bonnet bellflower or deodeok, is primarily found in Eastern Asia. Its roots have been used traditionally across Asia to treat various ailments such as bronchitis, coughs, asthma, and inflammation. In our ongoing efforts to discover bioactive [...] Read more.
Codonopsis lanceolata, commonly known as the bonnet bellflower or deodeok, is primarily found in Eastern Asia. Its roots have been used traditionally across Asia to treat various ailments such as bronchitis, coughs, asthma, and inflammation. In our ongoing efforts to discover bioactive natural products, a phytochemical investigation of the n-BuOH fraction of C. lanceolata root extracts led to the isolation and identification of a new indole alkaloid N-glycoside, deodeokaloid (D-indole-3-lactic acid N-β-D-glucopyranoside) (1), alongside known compounds tangshenoside I (2), tangshenoside IV (3), and chlorogenic acid (4) through HPLC purification. The structure of the new compound 1 was elucidated using 1D and 2D NMR spectroscopy and high-resolution electrospray ionization mass spectrometry (HR-ESIMS). Its absolute configuration was determined through a combination of DP4+ probability analysis and chemical reactions. The isolated compounds 14 were evaluated for their anti-Helicobacter pylori and antioxidant activities. In the anti-H. pylori assay, compound 3 showed antibacterial activity similar to that of quercetin as the positive control, inhibiting the bacterial growth by 36.8%. Compound 4 exhibited the most potent antioxidant activity, with an ABTS [2,2′-azino-bis(3-ethylbenzothiazoline-6-sulfonate)] radical scavenging activity of 1624.7 mmol TE/mol and a DPPH (2,2-diphenyl-1-picrylhydrazyl) radical scavenging capacity of 707.5 mmol Trolox equivalent (TE)/mol. Compounds 24 displayed significant intracellular reactive oxygen species (ROS) scavenging capacity in lipopolysaccharide-stimulated RAW 264.7 macrophage cells. This study highlights C. lanceolata roots as a promising natural source of bioactive compounds with potential therapeutic applications. Full article
(This article belongs to the Special Issue Alkaloids: Chemical Structures with Pharmaceutical Potential)
Show Figures

Figure 1

Figure 1
<p>Chemical structures of compounds <b>1</b>–<b>4</b>.</p>
Full article ">Figure 2
<p>Key <sup>1</sup>H-<sup>1</sup>H COSY (<span class="html-fig-inline" id="plants-13-03243-i001"><img alt="Plants 13 03243 i001" src="/plants/plants-13-03243/article_deploy/html/images/plants-13-03243-i001.png"/></span>) and HMBC (<span class="html-fig-inline" id="plants-13-03243-i002"><img alt="Plants 13 03243 i002" src="/plants/plants-13-03243/article_deploy/html/images/plants-13-03243-i002.png"/></span>) correlations for compound <b>1</b>.</p>
Full article ">Figure 3
<p>DP4+ analysis and probability scores for compound <b>1</b> with <b>1a</b>/<b>1b</b>.</p>
Full article ">Figure 4
<p>Photographs of aerial parts and roots of <span class="html-italic">C. lanceolata</span>.</p>
Full article ">Scheme 1
<p>Retrosynthetic analysis of deodeokaloid.</p>
Full article ">
23 pages, 7007 KiB  
Article
Amelioration of Toll-like Receptor-4 Signaling and Promotion of Mitochondrial Function by Mature Silkworm Extracts in Ex Vivo and in Vitro Macrophages
by Trinh Yen Binh Phan, Byungki Jang, Sang-Kuk Kang, Jongbok Seo, Seong-Ryul Kim, Kee-Young Kim and Young Ho Koh
Nutrients 2024, 16(22), 3932; https://doi.org/10.3390/nu16223932 - 18 Nov 2024
Viewed by 486
Abstract
Objectives: The unknown immune-enhancing effects of steamed mature silkworms (Bombyx mori L.), known as HongJam (HJ), were investigated. Methods: Supercritical fluid extracts from the White Jade variety of HJ (WJ-SCEs) were applied to in vitro RAW264.7 macrophages (RAWMs) and ex vivo bone [...] Read more.
Objectives: The unknown immune-enhancing effects of steamed mature silkworms (Bombyx mori L.), known as HongJam (HJ), were investigated. Methods: Supercritical fluid extracts from the White Jade variety of HJ (WJ-SCEs) were applied to in vitro RAW264.7 macrophages (RAWMs) and ex vivo bone marrow-derived macrophages (BMDMs). Results: WJ-SCE enhanced the proliferation and viability of both RAWMs and BMDMs. Supplementation with WJ-SCE significantly reduced the lipopolysaccharide (LPS)-induced expression of iNOS mRNA and protein, resulting in decreased nitric oxide (NO) production. Additionally, WJ-SCE lowered the mRNA and protein expression of COX-2 and reduced the levels of pro-inflammatory cytokines. The mitochondrial function, ATP levels, and reactive oxygen species levels in LPS-treated macrophages were restored following WJ-SCE treatment. WJ-SCE modulated LPS-Toll-like receptor-4 (TLR-4) signaling by reducing the levels of phosphorylated (p)-p38, p-ERK1/2, and p-p65. WJ-SCE also restored gene expression related to cytokines, chemokines, glucose transport, mitochondrial metabolism, and TLR-4 signaling, suggesting the inhibition of pro-inflammatory M1 macrophage polarization. Furthermore, WJ-SCE enhanced macrophage phagocytic and pinocytotic activity. Conclusions: WJ-SCE demonstrated anti-inflammatory effects by inhibiting LPS-induced M1 polarization in both macrophage types, potentially suppressing chronic inflammation while enhancing phagocytosis and pinocytosis. Full article
(This article belongs to the Section Nutritional Immunology)
Show Figures

Figure 1

Figure 1
<p>Altered cell viabilities and NO productions in RAWMs and BMDMs following treatment with various concentrations of WJ-SCE, either alone or in combination with LPS. (<b>A</b>) In RAWMs, treatment with WJ-SCE alone or in combination with LPS significantly increased cell viability (F<sub>(7, 40)</sub> = 27.909, <span class="html-italic">p</span> = 1.7 × 10<sup>−13</sup>); (<b>B</b>) WJ-SCE treatment alone significantly increased cell survival, and when combined with LPS, additive effect was observed in BMDMs (F<sub>(7, 40)</sub> = 27.909, <span class="html-italic">p</span> = 1.7 × 10<sup>−13</sup>); (<b>C</b>) In RAWMs, treatment with WJ-SCE alone slightly increased NO levels. LPS treatment led to 15.8-fold increase in NO levels, but when treated simultaneously with WJ-SCE, NO levels decreased in dose-dependent manner (F<sub>(7, 40)</sub> = 6182.752, <span class="html-italic">p</span> = 0.0007); (<b>D</b>) In BMDMs, NO levels surged with LPS treatment, but co-treatment with WJ-SCE significantly reduced them (F<sub>(7, 40)</sub> = 2134.3, <span class="html-italic">p</span> = 2.2 × 10<sup>−49</sup>). Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 2
<p>Enhanced phagocytic and pinocytotic activities in macrophages by WJ-SCE. (<b>A</b>) RAWMs treated with various concentrations of WJ-SCE showed significantly enhanced phagocytic activities with FITC-labeled <span class="html-italic">E. coli</span>; (<b>B</b>) phagocytic activities of RAWMs were significantly enhanced when supplemented with WJ-SCE at concentrations above 0.5 mg/mL (F<sub>(4, 245)</sub> = 8.433, 2.2 × 10<sup>−6</sup>); (<b>C</b>) pinocytotic activities of RAWMs were significantly and dose-dependently enhanced by WJ-SCE supplementation (F<sub>(5, 30)</sub> = 96.469, 1.5 × 10<sup>−17</sup>); (<b>D</b>) when RAWMs were co-treated with 100 ng/mL LPS and various concentrations of WJ-SCE, pinocytotic activities were significantly and dose-dependently enhanced by WJ-SCE (F<sub>(4, 25)</sub> = 15.838, 1.4 × 10<sup>−6</sup>); (<b>E</b>) BMDMs treated with various concentrations of WJ-SCE showed significantly enhanced phagocytic activities with FITC-labeled <span class="html-italic">E. coli</span>; (<b>F</b>) phagocytic activities of BMDMs were significantly enhanced when supplemented with WJ-SCE at concentrations above 0.5 mg/mL (F<sub>(5, 174)</sub> = 48.738, 2.3 × 10<sup>−31</sup>); (<b>G</b>) pinocytotic activities of BMDMs were significantly and LPS dose-dependently enhanced (F<sub>(6, 35)</sub> = 88.107, 1.2 × 10<sup>−19</sup>); (<b>H</b>) pinocytotic activities of BMDMs were significantly and dose-dependently enhanced by WJ-SCE supplementation (F<sub>(4, 25)</sub> = 110.01, 1.7 × 10<sup>−15</sup>); (<b>I</b>) when BMDMs were co-treated with 100 ng/mL LPS and various concentrations of WJ-SCE, pinocytotic activities were significantly and dose-dependently enhanced by WJ-SCE (F<sub>(5, 30)</sub> = 53.014, 7.5 × 10<sup>−12</sup>). Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 3
<p>Enhanced mitochondrial function, increased ATP amounts, and reduced ROS in RAWMs by WJ-SCE supplementation; (<b>A</b>) WJ-SCE dose-dependently increased MitoCom I activity in RAWMs. Reduced MitoCom I activity caused by LPS treatment in RAWMs was restored by WJ-SCE co-treatment, making it similar to Con (F<sub>(9, 30)</sub> = 91.612, <span class="html-italic">p</span> = 2.5 × 10<sup>−19</sup>); (<b>B</b>) Activity of MitoCom II in RAWMs was enhanced by WJ-SCE treatment. Reduction in MitoCom II activity caused by LPS treatment in RAWMs was restored by WJ-SCE treatment, similar to Con (F<sub>(9, 30)</sub> = 61.529, <span class="html-italic">p</span> = 7.1 × 10<sup>−17</sup>); (<b>C</b>) MitoCom III activity was significantly enhanced by WJ-SCE treatment. Reduction in MitoCom III activity caused by LPS treatment in RAWMs was partially restored by WJ-SCE treatment (F<sub>(9, 30)</sub> = 67.023, <span class="html-italic">p</span> = 2.1 × 10<sup>−17</sup>); (<b>D</b>) MitoCom IV activity was significantly enhanced by WJ-SCE treatment at doses greater than 1.0 mg/mL. Reduction in MitoCom IV activity caused by LPS treatment in RAWMs was partially restored by WJ-SCE treatment (F<sub>(9, 30)</sub> = 22.896, <span class="html-italic">p</span> = 4.3 × 10<sup>−11</sup>); (<b>E</b>) ATP levels in RAWMs significantly increased in dose-dependent manner with WJ-SCE treatment. While reduced ATP levels in LPS-treated RAWMs were significantly increased by WJ-SCE co-treatment, they remained significantly lower than those of Con (F<sub>(9, 30)</sub> = 251.64, <span class="html-italic">p</span> = 2.6 × 10<sup>−32</sup>); (<b>F</b>) ROS levels in BMDMs were not affected by WJ-SCE treatment, but significantly increased ROS levels in LPS-treated BMDMs were dose-dependently reduced by WJ-SCE, bringing them close to those of Con (F<sub>(9, 30)</sub> = 383.19, <span class="html-italic">p</span> = 1.9 × 10<sup>−28</sup>). Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 4
<p>Enhanced mitochondrial function, increased ATP amounts, and reduced ROS in BMDMs by WJ-SCE supplementation; (<b>A</b>) MitoCom I activity in BMDMs significantly and dose-dependently increased with WJ-SCE treatment alone. Activity reduced by LPS treatment to 40.7% was significantly and dose-dependently increased by WJ-SCE, and in group treated with 5 mg/mL WJ-SCE, there was no statistical difference from Con (F<sub>(9, 30)</sub> = 66.798, <span class="html-italic">p</span> = 2.2 × 10<sup>−17</sup>); (<b>B</b>) MitoCom II activity in BMDMs significantly and dose-dependently increased with WJ-SCE treatment alone. Activity reduced by LPS treatment to 42.2% increased with WJ-SCE treatment, but increase was not statistically significant (F<sub>(9, 30)</sub> = 112.767, <span class="html-italic">p</span> = 1.2 × 10<sup>−20</sup>); (<b>C</b>) MitoCom III activity in BMDMs significantly and dose-dependently increased with WJ-SCE treatment alone. Activity reduced by LPS treatment to 48.8% significantly increased in groups treated with 0.5 mg/mL or higher of WJ-SCE, but remained lower than in Con (F<sub>(9, 30)</sub> = 89.107, <span class="html-italic">p</span> = 3.7 × 10<sup>−19</sup>); (<b>D</b>) MitoCom IV activity in BMDMs significantly and dose-dependently increased with WJ-SCE treatment alone. Activity reduced by LPS treatment to 50.8% significantly increased to 73.5% in groups treated with 1.0 mg/mL or higher of WJ-SCE, but remained significantly lower than in Con (F<sub>(9, 30)</sub> = 75.049, <span class="html-italic">p</span> = 4.3 × 10<sup>−18</sup>); (<b>E</b>) ATP levels in BMDMs significantly and dose-dependently increased with WJ-SCE treatment alone. Levels reduced by LPS treatment to 34.9% significantly increased to 68.2% in groups treated with 0.5 mg/mL or higher of WJ-SCE, but remained significantly lower than in Con (F<sub>(9, 40)</sub> = 1213.09, <span class="html-italic">p</span> = 7.7 × 10<sup>−46</sup>); (<b>F</b>) ROS levels in BMDMs significantly and dose-dependently decreased with WJ-SCE treatment, whether administered alone or in combination with LPS (F<sub>(9, 50)</sub> = 22.487, <span class="html-italic">p</span> = 1.2 × 10<sup>−14</sup>). Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 5
<p>Modulation of pro-inflammatory cytokine production in macrophages by WJ-SCE supplementation. Significantly increased expressions of TNF-α ((<b>A</b>) RAWMs, F<sub>(4, 10)</sub> = 2037.5, <span class="html-italic">p</span> = 1.7 × 10<sup>−14</sup>; (<b>B</b>) BMDMs, F<sub>(5, 12)</sub> = 18,305.5, <span class="html-italic">p</span> = 7.4 × 10<sup>−23</sup>), IL-6 ((<b>C</b>) RAWMs, F<sub>(5, 12)</sub> = 31.563, <span class="html-italic">p</span> = 1.7 × 10<sup>−6</sup>; (<b>D</b>) BMDMs, F<sub>(5, 12)</sub> = 1585.6, <span class="html-italic">p</span> = 1.7 × 10<sup>−16</sup>), and IL-1β ((<b>E</b>) RAWMs, F<sub>(5, 12)</sub> = 30.424, <span class="html-italic">p</span> = 2.0 × 10<sup>−6</sup>; (<b>F</b>) BMDMs, F<sub>(5, 12)</sub> = 291.0, <span class="html-italic">p</span> = 4.4 × 10<sup>−12</sup>) by LPS treatment were significantly and dose-dependently reduced when WJ-SCE was co-treated. Effects of WJ-SCE were more prominent in RAWMs than BMDMs. Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 6
<p>WJ-SCE reduced LPS-induced increased NO production in macrophages by reducing expression of iNOS and COX2, as well as inhibition of activation of p38, ERK, and p65; (<b>A</b>) WJ-SCE reduced LPS-induced increased NO in RAWMs (F<sub>(5, 12)</sub> = 6374.9, <span class="html-italic">p</span> = 4.2 × 10<sup>−20</sup>). Expressions of iNOS mRNA in RAWMs ((<b>B</b>) F<sub>(5, 12)</sub> = 29,091, <span class="html-italic">p</span> = 2.6 × 10<sup>−6</sup>) and BMDMs ((<b>C</b>) F<sub>(5, 12)</sub> = 22.102, <span class="html-italic">p</span> = 1.1 × 10<sup>−5</sup>) were significant and WJ-SCE dose-dependent. Expressions of COX2 mRNA in RAWMs ((<b>D</b>) F<sub>(5, 12)</sub> = 20.872, <span class="html-italic">p</span> = 1.5 × 10<sup>−5</sup>) and BMDMs ((<b>E</b>) F<sub>(5, 12)</sub> = 4.065, <span class="html-italic">p</span> = 0.022) were significantly reduced. (<b>F</b>) Expression of iNOS and COX2 proteins was reduced by WJ-SCE supplementation in RAWMs. (<b>G</b>) Sharply increased iNOS protein expressions in RAWMs by LPS were significantly reduced in WJ-SCE-supplemented RAWMs. (<b>H</b>) Similarly, drastically increased COX2 protein expressions were significantly reduced in WJ-SCE-supplemented RAWMs. (<b>I</b>) After LPS treatments, activated p-p35, p-ERK, and p-p65 were detected. WJ-SCE co-treatment reduced expression of p-p35, p-ERK, and p-p65. (<b>J</b>,<b>K</b>) 30 min after LPS and WJ-SCE co-treatment, levels of p-p35, p-ERK, and p-65 were significantly reduced (<span class="html-italic">p</span> values: p-p38 = 0.00924, p-ERK = 0.00922, p-p65 = 0.00045). Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 7
<p>Expression changes in mRNAs of key regulators in TRL-4 signaling, pro-inflammatory responses, metabolism, and mitochondrial function induced by LPS treatments were recovered by WJ-SCE. Ex<span class="html-italic">p</span>ression levels of TRL-4 ((<b>A</b>) RAWMs, F<sub>(5, 12)</sub> = 23.6224, <span class="html-italic">p</span> = 8.0 × 10<sup>−6</sup>; (<b>B</b>) BMDMs, F<sub>(5, 12)</sub> = 23.6224, <span class="html-italic">p</span> = 8.0 × 10<sup>−6</sup>), NF-κB ((<b>C</b>) RAWMs, F<sub>(5, 12)</sub> = 4.534, <span class="html-italic">p</span> = 0.015; 6 (<b>D</b>) BMDMs, F<sub>(5, 12)</sub> = 15.833, <span class="html-italic">p</span> = 6.4 × 10<sup>−5</sup>), HIF-1α ((<b>E</b>) RAWMs, F<sub>(5, 12)</sub> = 55.146, <span class="html-italic">p</span> = 7.3 × 10<sup>−8</sup>; (<b>F</b>) BMDMs, F<sub>(5, 12)</sub> = 35.874, <span class="html-italic">p</span> = 8.2 × 10<sup>−7</sup>), IFN-γ ((<b>G</b>) RAWMs, F<sub>(5, 12)</sub> = 19.473, <span class="html-italic">p</span> = 2.2 × 10<sup>−5</sup>; (<b>H</b>), F<sub>(5, 12)</sub> = 19.473, <span class="html-italic">p</span> = 2.2 × 10<sup>−5</sup>), IL-18 ((<b>I</b>) RAWMs, F<sub>(5, 12)</sub> = 8.549, <span class="html-italic">p</span> = 0.001; (<b>J</b>) BMDMs, F<sub>(5, 12)</sub> = 5.755, <span class="html-italic">p</span> = 0.006), MC<span class="html-italic">p</span>1 ((<b>K</b>) RAWMs, F<sub>(5, 12)</sub> = 11.277, <span class="html-italic">p</span> = 0.0003; (<b>L</b>) BMDMs, F<sub>(5, 12)</sub> = 8.228, <span class="html-italic">p</span> = 0.0014), CCL3 ((<b>M</b>) RAWMs, F<sub>(5, 12)</sub> = 22.989, <span class="html-italic">p</span> = 9.2 × 10<sup>−6</sup>; (<b>N</b>) BMDMs, F<sub>(5, 12)</sub> = 45.319, <span class="html-italic">p</span> = 2.2 × 10<sup>−7</sup>), TRIF ((<b>O</b>) RAWMs, F<sub>(5, 12)</sub> = 23.412, <span class="html-italic">p</span> = 8.4 × 10<sup>−6</sup>; (<b>P</b>) BMDMs, F<sub>(5, 12)</sub> = 84.461, <span class="html-italic">p</span> = 6.3 × 10<sup>−9</sup>), MYD88 ((<b>Q</b>) RAWMs, F<sub>(5, 12)</sub> = 6.903, <span class="html-italic">p</span> = 0.003; (<b>R</b>) BMDMs, F<sub>(5, 12)</sub> = 12.011, <span class="html-italic">p</span> = 0.0002), ACOD1 ((<b>S</b>) RAWMs, F<sub>(5, 12)</sub> = 10.397, <span class="html-italic">p</span> = 0.0005; (<b>T</b>) BMDMs, F<sub>(5, 12)</sub> = 28.246, <span class="html-italic">p</span> = 3.1 × 10<sup>−6</sup>), Glut1 ((<b>U</b>) RAWMs, F<sub>(5, 12)</sub> = 38.663, <span class="html-italic">p</span> = 5.1 × 10<sup>−7</sup>; (<b>V</b>) BMDMs, F<sub>(5, 12)</sub> = 13.426, <span class="html-italic">p</span> =0.00015), and UC<span class="html-italic">p</span>-2 ((<b>W</b>) RAWMs, F<sub>(5, 12)</sub> = 20.558, <span class="html-italic">p</span> = 1.7 × 10<sup>−5</sup>; (<b>X</b>) BMDMs, F<sub>(5, 12)</sub> = 5.445, <span class="html-italic">p</span> = 0.008) were investigated by RT-q-<span class="html-italic">p</span>CR analyses. Different letters above error bars indicated statistically significant differences.</p>
Full article ">Figure 8
<p>WJ-SCE reprograms LPS-induced pro-inflammatory M1 macrophages into anti-inflammatory M2 macrophages. LPS induced TLR-4 signal transduction pathway activation by phosphorylating p38, ERK1/2, and p65, resulting in upregulated expressions of iNOS, COX2, HIF-1α, NF-κB, cytokines, chemokines, TRIF1, MYD88, and ACOD1 mRNAs. NO and ROS also significantly increased, and mitochondrial function and ATP amounts were reduced in two macrophages. Treatment of WJ-SCE together with LPS restored p-p38, p-ERK1/2, and p-p65 gene expression patterns, mitochondrial function, and amounts of ATP, NO, and ROS in two macrophages. In addition, phagocytic and pinocytotic activities of macrophages were significantly enhanced. Red and blue arrows indicate upregulation and downregulation of gene expression, as well as enzyme and mitochondrial activities, respectively, while yellow-black arrows represent signal transduction pathways.</p>
Full article ">
19 pages, 5471 KiB  
Article
Chamomile Tincture and Lidocaine Hydrochloride Gel Ameliorates Periodontitis: A Preclinical Study
by Jiahui Sun, Huiyi Wang, Junhong Xiao, Qiudong Yang, Heyu Liu, Zhengkun Yang, Yuqi Liu, Xin Huang, Liu Yang, Li Ma and Zhengguo Cao
Biomedicines 2024, 12(11), 2629; https://doi.org/10.3390/biomedicines12112629 - 17 Nov 2024
Viewed by 455
Abstract
Background/Objectives: Periodontitis is a common oral disease marked by gingival inflammation and alveolar bone loss. This study evaluated the efficacy of chamomile tincture and lidocaine hydrochloride (CLH) gel in mitigating periodontal inflammation and bone loss and uncovered the molecular mechanisms involved, both [...] Read more.
Background/Objectives: Periodontitis is a common oral disease marked by gingival inflammation and alveolar bone loss. This study evaluated the efficacy of chamomile tincture and lidocaine hydrochloride (CLH) gel in mitigating periodontal inflammation and bone loss and uncovered the molecular mechanisms involved, both in vitro and in vivo. Methods: A periodontitis model was induced in Sprague Dawley rats by ligating the mandibular first molars. Sixty rats were divided into four groups: control (C), periodontitis (PD), periodontitis + CLH gel once daily (G1), and periodontitis + CLH gel thrice daily (G3). Clinical, micro-computed tomography (micro-CT), biological, and histological evaluations were performed, focusing on osteoclastogenesis, osteogenesis, and inflammatory cytokine production. The effect of CLH gel on inflammatory responses in RAW264.7 cells was also assessed through co-culture assays under Porphyromonas gingivalis (P. gingivalis) infection, with RNA-sequencing, qPCR, and Western blot analyses to explore underlying mechanisms. Results: CLH gel significantly reduced gingival and systemic inflammation and mitigated bone loss by enhancing the bone volume to tissue volume ratio and trabecular thickness via the RANKL/OPG axis in rats. The G3 group showed marked reductions in osteoclasts and increases in osterix-positive cells compared to other groups. In vitro, CLH gel reduced the inflammatory phenotype of macrophages in the periodontitis microenvironment by modulating Type II interferon (IFN-γ) networks. Conclusions: CLH gel reduced inflammation and bone loss in rat periodontitis, promoting osteogenesis and inhibiting osteoclastogenesis. It also suppressed macrophage inflammation via Type II interferon networks under P. gingivalis stimulation. These findings suggest that CLH gel has potential as an adjunctive therapy for periodontitis. Full article
(This article belongs to the Section Drug Discovery, Development and Delivery)
Show Figures

Figure 1

Figure 1
<p>The experimental model and clinical parameters. Overview of the study schedule (<b>a</b>). C (no intervention), PD (ligature-induced periodontitis), G1 (ligature-induced periodontitis plus once-daily administration of CLH gel), G3 (ligature-induced periodontitis plus a thrice-daily administration of CLH gel). Average periodontal attachment loss (the distance between the cemento-enamel junction (CEJ) and the coronal position of the junctional epithelium) in the first molar (<b>b</b>). Results represent the means ± standard error of the mean (SEM) performed in four samples. H&amp;E staining of the interdental epithelial attachment (black arrows indicate the actual epithelial attachment) between the first and second molars at low and high magnifications (<b>c</b>). Results represent the means ± SEM performed in four samples in each group. Clinical measurements of the PPD (distance from the gingival margin to the pocket bottom) at three sites (mesial, medial, distal) on the buccal side of the first molar (<b>d</b>). Results represent the means ± SEM of 10 samples. Immunohistochemical analysis of collagen fibers at low and high magnifications (<b>e</b>). Results represent the means ± SEM of collagen fibers of four samples in each group (<b>f</b>). Statistical analysis was performed with one-way ANOVA. 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; 0.0001.</p>
Full article ">Figure 2
<p>Alterations in the alveolar bone and the microarchitecture of the trabecular bone after modeling. Three-dimensional and bi-dimensional views of the mandibular molars from micro-CT scanning (<b>a</b>). Reconstructive images in red depict areas of bone loss. The reconstructive images of furcation areas in first molars (<b>b</b>). The distance between the CEJ and alveolar bone crest (ABC) on the buccal and lingual sides (<b>c</b>). Results represent the means ± SEM of 10 samples. Microarchitectural parameters of the alveolar bone (<b>d</b>). Bone volume to total tissue volume (BV/TV%), trabecular number (Tb.N), trabecular thickness (Tb.Th), trabecular separation (Tb.Sp). Results represent the means ± SEM of 10 samples. Statistical analysis was performed with one-way ANOVA. 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.</p>
Full article ">Figure 3
<p>Representative histological observations and analyses of markers related to bone metabolism. TRAP staining of osteoclast infiltration in the periodontium, where osteoclasts are depicted in red, at both low and high magnifications (<b>a</b>). Immunohistochemical expression of OPG and RANKL in the interdental periodontal tissues (<b>b</b>). Immunohistochemical expression of OCN, BSP, and OSX in the interdental periodontal tissues (<b>c</b>). Immunohistochemical analysis (<b>d</b>). Means and standard deviations of the number of TRAP-positive cells, RANKL-positive cells/OPG-positive cells, OCN-positive cells, BSP-positive cells, and OSX-positive cells. Results represent the means ± SEM of four samples. Statistical analysis was performed with one-way ANOVA. 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; 0.0001.</p>
Full article ">Figure 4
<p>Effects of experimental periodontitis on the inflammatory status of the gingival tissues. Immunohistochemical expression of IL-6, IL-1β, and TNF-α in the interdental gingiva and periodontal ligament (<b>a</b>). Immunohistochemical analysis of IL-6, IL-1β, and TNF-α (<b>b</b>). Means and standard deviations of the number of positive cells. Results represent the means ± SEM of four samples. The expression of inflammation-related genes in rat gingival tissues (<b>c</b>). Total RNA was extracted from the gingival tissue around the first molars, and the mRNA levels of <span class="html-italic">TNF-α</span>, <span class="html-italic">IL-6</span>, <span class="html-italic">IL-1β</span>, <span class="html-italic">iNOS</span>, <span class="html-italic">COX2</span>, <span class="html-italic">MMP-1</span>, <span class="html-italic">MMP-3</span>, and <span class="html-italic">MMP-13</span> were determined by qRT-PCR. Results represent the means ± SEM of three samples. Statistical analysis was performed with one-way ANOVA. 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.</p>
Full article ">Figure 5
<p>Effects of experimental periodontitis on the inflammatory status of the serum and systemic organs. Myeloperoxidase (MPO) activity in the serum of rats (<b>a</b>). Results represent the means ± SEM of three samples. Inflammatory mediator production (TNF-α, IL-6, IL-1β) in the serum of the rats (<b>b</b>). ELISA kits were employed to detect the protein levels of the inflammatory cytokines. Results represent the means ± SEM of three samples. H&amp;E staining of major organs (heart, liver, spleen, lung, and kidney) in experimental rats (<b>c</b>). No obvious damage was observed in the visceral organs (n = 6/group). Statistical analysis was performed with one-way ANOVA. 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.</p>
Full article ">Figure 6
<p>Anti-inflammatory effects of CLH gel on <span class="html-italic">P. gingivalis</span>-infected RAW264.7 cells. (<b>a</b>) iNOS and IL-1β mRNA levels in RAW264.7 cells after <span class="html-italic">P. gingivalis</span> (MOI = 100) stimulation for 6 h, with 1 µg/mL CLH treatment. (<b>b</b>) iNOS protein expression under the same conditions. (<b>c</b>) Gene set enrichment analysis (GSEA) line graph of functional gene set enrichment scores. (<b>d</b>) Heatmap of differentially expressed genes in the Type II interferon network, ranked by enrichment score. (<b>e</b>) mRNA levels of irf8, stat2, socs1, psmb9, tap1, irf1, IL-1β, and stat1 under the same conditions. (<b>f</b>) Protein expressions of IRF8, STAT2, SOCS1, IRF1, IL-1β, and STAT1 in RAW264.7 cells after stimulation with <span class="html-italic">P. gingivalis</span> combined with 1 µg/mL CLH for 6 h. with 1 µg/mL CLH. Statistical analysis was performed with one-way ANOVA. ns, no significance. ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">
21 pages, 7918 KiB  
Article
Genotoxic and Anti-Genotoxic Assessments of Fermented Houttuynia cordata Thunb. Leaf Ethanolic Extract and Its Anti-Cancer Effect in a Dual-Organ Carcinogenesis Model of Colon and Liver in Rats
by Chonikarn Singai, Pornsiri Pitchakarn, Sirinya Taya, Rawiwan Wongpoomchai and Ariyaphong Wongnoppavich
Foods 2024, 13(22), 3645; https://doi.org/10.3390/foods13223645 - 15 Nov 2024
Viewed by 583
Abstract
The incidence of multiple-organ cancers has recently increased due to simultaneous exposure to various environmental carcinogens. Houttuynia cordata Thunb. (H. cordata) is recognized for its many health benefits, including its anti-cancer properties. The fermentation of its leaves has been shown to [...] Read more.
The incidence of multiple-organ cancers has recently increased due to simultaneous exposure to various environmental carcinogens. Houttuynia cordata Thunb. (H. cordata) is recognized for its many health benefits, including its anti-cancer properties. The fermentation of its leaves has been shown to significantly enhance the bioflavonoid content and its bioactivities. This study aimed to evaluate the effectiveness of fermented H.cordata leaf (FHCL) extracts against combined carcinogens and investigate the underlying mechanisms. The crude ethanolic extract of FHCL was partitioned to obtain hexane- (HEX), dichloromethane- (DCM), ethyl acetate- (ETAC), butanol- (nBA), and residue fractions. The crude ethanolic extract (200–250 μg/mL) and the DCM fraction (50 μg/mL) significantly reduced NO production in RAW264.7 macrophages. In addition, the crude extract and the DCM and ETAC fractions showed anti-genotoxicity against aflatoxin B1 and 2-amino-3,4-dimethylimidazo [4,5-f]quinoline (MeIQ) in Salmonella typhimurium assays (S9+). Despite demonstrating genotoxicity in the Salmonella mutation assay (with and without S9 activation), oral administration of the crude extract at 500 mg/kg of body weight (bw) for 40 days in rats did not induce micronucleated hepatocytes, indicating that the extract is non-genotoxic in vivo. Moreover, the crude extract significantly decreased Phase I but increased Phase II xenobiotic-metabolizing enzyme activities in the rats. Next, the anti-cancer effects of FHCL were evaluated in a dual-organ carcinogenesis model of the colon and liver in rats induced by 1,2-dimethylhydrazine (DMH) and diethylnitrosamine (DEN), respectively. The crude extract significantly reduced not only the number and size of glutathione S-transferase placental form positive foci in the liver (at doses of 100 and 500 mg/kg bw) but also the number of aberrant crypt foci in rat colons (at 500 mg/kg bw). Furthermore, FHCL significantly reduced the expression of proliferating cell nuclear antigen (PCNA) in the colon (at 100 and 500 mg/kg bw) and liver (at 500 mg/kg bw) of the treated rats. In conclusion, FHCL exhibits significant preventive properties against colon and liver cancers in this dual-organ carcinogenesis model. Its mechanisms of action may involve anti-inflammatory effects, the prevention of genotoxicity, the modulation of xenobiotic-metabolizing enzymes, and the inhibition of cancer cell proliferation. These findings support the use of FHCL as a natural supplement for preventing cancer. Full article
Show Figures

Figure 1

Figure 1
<p>The effect of FHCL crude ethanolic extract (<b>A</b>) and its fractions (<b>B</b>) on nitric oxide (NO) production in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages was assessed using Griess reagents. The results are presented as the mean ± standard deviation from three independent experiments. Significance levels are indicated as *** <span class="html-italic">p</span> ≤ 0.001 and **** <span class="html-italic">p</span> ≤ 0.0001. HEX: hexane fraction; DCM: dichloromethane fraction; ETAC: ethyl acetate fraction; nBA: butanol fraction.</p>
Full article ">Figure 2
<p>The genotoxicity protocol. (<b>A</b>) The effect of crude FHCL ethanolic extract on micronucleated hepatocytes (<b>B</b>) and the activities of xenobiotic-metabolizing enzymes were evaluated in rat liver. Deionized (DI) water was used as the negative control. The enzymes studied included Phase I enzymes CYP1A1 (<b>C</b>), CYP1A2 (<b>D</b>), CYP2E1 (<b>E</b>), and CYP3A2 (<b>F</b>) and Phase II enzymes glutathione S-transferases (GST) (<b>G</b>) and UDP-glucuronosyltransferase (UGT) (<b>H</b>). Specific assays were performed for each enzyme. The results are presented as the mean ± standard deviation. Statistical significance was indicated by * (<span class="html-italic">p</span> ≤ 0.05) when compared to the vehicle control group. “ns” denotes non-significant differences (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 3
<p>The study protocol for the dual-organ carcinogenesis model, including the timeline for administering carcinogens (DEN and DMH) and crude FHCL extract in rats (<b>A</b>). The serum alanine aminotransferase (ALT) levels in rats (<b>B</b>). Normal saline (NSS) was used as the control for the carcinogen-treated group, while deionized (DI) water served as the control for the crude extract treatment. The data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 8). Statistical significance is indicated by * (<span class="html-italic">p</span> ≤ 0.05) and ** (<span class="html-italic">p</span> ≤ 0.01).</p>
Full article ">Figure 4
<p>The effects of a ten-week administration period of crude FHCL extract on preneoplastic lesions induced by dual carcinogens in rat colons and livers. The analysis of preneoplastic lesions in the colon includes the number (<b>A</b>) and area (<b>B</b>) of aberrant crypt foci (ACF), identified with 0.2% methylene blue staining. For the liver, preneoplastic lesions are evaluated by the number (<b>C</b>) and size (<b>D</b>) of glutathione <span class="html-italic">S</span>-transferase placental form (GST-P)-positive foci, detected through immunohistochemistry. The figure shows representative images of preneoplastic lesions (ACF and GST-P-positive foci indicated by arrows) in the colons (40× magnification) and livers (100× magnification) (<b>E</b>). The data are presented as the mean ± standard deviation (<span class="html-italic">n</span> = 8). Statistical significance is indicated by * (<span class="html-italic">p</span> ≤ 0.05) and **** (<span class="html-italic">p</span> ≤ 0.0001). “ns” indicates non-significant differences (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 5
<p>Effect of crude FHCL extract on early-stage carcinogenesis in rats: modulation of cell proliferation. Immunohistochemical staining was used to quantify proliferating cell nuclear antigen (PCNA)-positive cells in colon (<b>A</b>) and liver (<b>B</b>). Figure displays representative immunohistochemical images of PCNA-positive cells in liver sections (400× magnification) and colon sections (400× magnification) (<b>C</b>). In colon, PCNA-positive cells exhibit brownish staining, while PCNA-positive hepatic cells are indicated by arrows. Data are presented as mean ± standard deviation. Statistical significance is denoted by ** (<span class="html-italic">p</span> ≤ 0.01) and **** (<span class="html-italic">p</span> ≤ 0.0001). “ns” indicates non-significant differences (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 6
<p>Effect of crude FHCL extract on early-stage carcinogenesis in rats: modulation of cell apoptosis. Immunohistochemical staining was used to quantify TUNEL-positive cells in colon (<b>A</b>) and liver (<b>B</b>). Figure displays representative immunohistochemical images of TUNEL-positive cells in colon sections (400× magnification) and liver sections (200× magnification) (<b>C</b>). TUNEL-positive cells in colon exhibit brownish staining, while TUNEL-positive hepatic cells are indicated by arrows. Data are presented as mean ± standard deviation. “ns” indicates non-significant differences (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">
15 pages, 4962 KiB  
Article
Anti-Inflammatory Effects of Cordyceps Cs-HK1 Fungus Exopolysaccharide on Lipopolysaccharide-Stimulated Macrophages via the TLR4/MyD88/NF-κB Pathway
by Yan-Yu Zhu, Yu-Han Dong, Fang-Ting Gu, Zi-Chen Zhao, Lin-Xi Huang, Wai-Yin Cheng and Jian-Yong Wu
Nutrients 2024, 16(22), 3885; https://doi.org/10.3390/nu16223885 - 14 Nov 2024
Viewed by 575
Abstract
Chronic inflammation is a common factor in the pathological processes of multiple human diseases. EPS-LM, an exopolysaccharide (EPS) from the Cordyceps sinensis fungus Cs-HK1, has shown notable anti-inflammatory activities in previous studies. This study aimed to investigate the major signaling events mediating the [...] Read more.
Chronic inflammation is a common factor in the pathological processes of multiple human diseases. EPS-LM, an exopolysaccharide (EPS) from the Cordyceps sinensis fungus Cs-HK1, has shown notable anti-inflammatory activities in previous studies. This study aimed to investigate the major signaling events mediating the anti-inflammatory effects of EPS-LM in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cell culture. EPS-LM treatment significantly reduced LPS-induced production of pro-inflammatory mediators, including nitric oxide (NO) and reactive oxygen species (ROS). It also suppressed the expression levels of Toll-like receptor 4 (TLR4) and myeloid differentiation primary response gene 88 (MyD88), subsequently delaying the translocation of nuclear factor-kappa B (NF-κB) to the nucleus. Additionally, co-immunoprecipitation (Co-IP) experiments demonstrated that EPS-LM inhibited the binding of TLR4 to MyD88. The ability of EPS-LM to inhibit the TLR4/MyD88/NF-κB pathway, coupled with its capacity to reduce oxidative stress, underscores its multifaceted anti-inflammatory effects. These effects render EPS-LM as a promising candidate for the comprehensive management of various inflammatory and oxidative stress-related conditions, protecting against cell damage. Full article
(This article belongs to the Section Nutrition and Metabolism)
Show Figures

Figure 1

Figure 1
<p>Major experimental steps for isolation of EPS-LM from Cs-HK1 mycelial fermentation and purification process.</p>
Full article ">Figure 2
<p>FT-IR spectrum of EPS-LM isolated from Cs-HK1 mycelial fermentation medium.</p>
Full article ">Figure 3
<p>Effects of EPS-LM treatment on cell viability and proliferation in RAW 264.7 cells. (Data as mean ± SEM, <span class="html-italic">n</span> = 6. One-way ANOVA followed by Tukey’s multiple comparison test: * <span class="html-italic">p</span> &lt; 0.1, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 versus Ctrl).</p>
Full article ">Figure 4
<p>Effects of EPS-LM on LPS-induced ROS production in RAW 264.7 cells. (Data as mean ± SEM, <span class="html-italic">n</span> = 6. One-way ANOVA followed by Tukey’s multiple comparison test: *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 versus LPS).</p>
Full article ">Figure 5
<p>Effects of LPS and EPS-LM on RAW 264.7 cells: morphology of (<b>A</b>) control; (<b>B</b>) LPS-treated; (<b>C</b>) LPS + 200 µg/mL EPS-treated group; (<b>D</b>) NO production. (Data as mean ± SEM, <span class="html-italic">n</span> = 6; One-way ANOVA followed by Tukey’s multiple comparison test: ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 versus LPS).</p>
Full article ">Figure 6
<p>Western blotting and immunofluorescence analysis of the effect of EPS-LM on LPS-induced inflammatory protein expression in RAW 264.7 cells: (<b>A</b>) the representative bands of IL-18, NF-κB p65, TLR4, and MyD88 were determined by Western blotting. Actin were used as internal control; (<b>B</b>–<b>D</b>) TLR4, MyD88, NF-κB p65 expression levels in RAW 264.7 cells; (<b>E</b>–<b>G</b>) statistical analysis of the ratio of green to blue fluorescence for images corresponding to TLR4, MyD88, and NF-κB p65, respectively. (Data as mean ± SEM, <span class="html-italic">n</span> = 3. One-way ANOVA followed by Tukey’s multiple comparison test: *** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001 versus LPS).</p>
Full article ">Figure 7
<p>EPS-LM reduces the interaction between TLR4 and MyD88 in RAW 264.7 cells. RAW 264.7 cells were treated with LPS in the presence or absence of EPS-LM (50 and 200 µg/mL). Co-IP was performed to assess the interaction between TLR4 and MyD88. TLR4 and MyD88 were detected by immunoblotting (IB) in the input and TLR4-immunoprecipitated (IP: TLR4) samples. GAPDH was used as a loading control.</p>
Full article ">Figure 8
<p>A hypothesized signal mechanism for the protective effects of EPS-LM against LPS-induced inflammatory injury and ROS via the TLR4/MyD88/NF-κB pathway: (1) LPS binding to TLR4 activates MyD88, leading to a signaling cascade that degrades IκB, releasing and phosphorylating p50 and p65 subunits; (2) these subunits form an active NF-κB complex that moves to the nucleus to transcribe pro-inflammatory cytokines like TNF-α and IL-1β; (3) LPS also induces ROS and NO production in mitochondria, contributing to inflammation and damage; (4) EPS-LM inhibits TLR4-MyD88 interaction, reducing ROS and NO generation and providing anti-inflammatory protection.</p>
Full article ">
10 pages, 2767 KiB  
Article
Uricase-Expressing Engineered Macrophages Alleviate Murine Hyperuricemia
by Yu-Zhong Feng, Hao Cheng, Guo-Qing Xiong, Jia-Zhen Cui, Zhi-Li Chen, Yuan-Yuan Lu, Zhi-Xin Meng, Chen Zhu, Hao-Long Dong, Xiang-Hua Xiong, Gang Liu, Qing-Yang Wang and Hui-Peng Chen
Biomedicines 2024, 12(11), 2602; https://doi.org/10.3390/biomedicines12112602 - 14 Nov 2024
Viewed by 443
Abstract
Background: Uricase, or urate oxidase (Uox) is a key enzyme in uric acid (UA) metabolism and has been applied in clinical treatment of human hyperuricemia (HUA). However, the current clinically applied uricases, despite their potent urate-lowering capacity, tend to form anti-drug antibodies [...] Read more.
Background: Uricase, or urate oxidase (Uox) is a key enzyme in uric acid (UA) metabolism and has been applied in clinical treatment of human hyperuricemia (HUA). However, the current clinically applied uricases, despite their potent urate-lowering capacity, tend to form anti-drug antibodies because of their immunogenicity, leading to increased risk of anaphylaxis, faster drug clearance and reduced or even complete loss of therapeutic effect, limiting their clinical application. In this study, we constructed engineered macrophages that stably expressed uricase, which might serve as a promising alternative to the direct injection of uricases. Materials and Methods: Engineered macrophages RAW264.7 cells were injected intravenously to treat hyperuricemic KM mice. Serum uric acid and bio-indicators for renal and hepatic functions were detected by an automatic biochemical analyzer; inflammatory cytokines were determined by ELISA; the livers and kidneys of the mice were sectioned for histological examination. Results: The uricase-expressing macrophages reduced UA levels from 300 ± 1.5 μmol/L to 101 ± 8.3 μmol/L in vitro. And in an HUA mouse model established by gavage with yeast extract, intravenous injection of the engineered macrophages could reduce the serum uric acid (sUA) of mice to normal level on the 14th day of modeling, with a decrease of 48.6%, and the urate-lowering effect was comparable to that of the first-line clinical drug allopurinol. In terms of safety, engineered macrophages did not cause liver or kidney dysfunction in mice, nor did they induce systemic immune response. Conclusions: Using macrophages as a chassis to deliver uricase might be a new, safe and effective strategy for the treatment and control of hyperuricemia. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Figure 1

Figure 1
<p><span class="html-italic">mus</span>-Uri expression in the engineered macrophage RAW264.7. (<b>A</b>) Positive rate after lentiviral transfection, detected by flow cytometry; (<b>B</b>) uricase relative expression determined by qPCR; (<b>C</b>) <span class="html-italic">mus</span>-Uri protein expression confirmed by Western blotting.</p>
Full article ">Figure 2
<p>Engineered macrophage reduces UA level in vitro. Comparison was performed between RAW-mus Uri and RAW-Ctrl groups at each time point. (n = 10, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 3
<p>(<b>A</b>) Rate of EGFP-positive RAW264.7 after cytometry sorting; P1 represents all viable cells, while P2 represents EGFP-positive cells. (<b>B</b>) Uric acid levels in each group 0, 7, and 14 days after intervention. The levels between HUA + RAW-Ctrl and HUA + RAW-Uri were compared. (n = 10, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 4
<p>Safety evaluation of engineered macrophage RAW-<span class="html-italic">mus</span>-Uri. (<b>A</b>) Hepatic function indicators AST, ALT, TBIL and DBIL and renal function indicators urea and crea collected from serum are assessed. (n = 10, *** <span class="html-italic">p</span> &lt; 0.001) (<b>B</b>,<b>C</b>) H&amp;E staining and histological examination of liver (<b>B</b>) and kidney (<b>C</b>) in each group.</p>
Full article ">
19 pages, 99468 KiB  
Article
2-Acetamidophenol (2-AAP) Suppresses the Progression of Atherosclerosis by Alleviating Hyperlipidemia and Attenuating the Ferroptosis Pathway
by Xiaohan Zang, Yongcheng Wang, Cong Han, Lishuang Cui, Haojie Liu, Shuimiao Tian, Kechun Liu, Peihai Li, Chen Sun, Qing Xia and Yun Zhang
Mar. Drugs 2024, 22(11), 513; https://doi.org/10.3390/md22110513 - 13 Nov 2024
Viewed by 577
Abstract
Hyperlipidemia and consequent endothelial inflammation, along with foam cell generation, promote the progression of atherosclerosis (AS). Here, we aimed to investigate the effects of 2-acetamidophenol (2-AAP), which was selected by zebrafish phenotypic screening, in alleviating AS by relieving hyperlipidemia and inhibiting foam cell [...] Read more.
Hyperlipidemia and consequent endothelial inflammation, along with foam cell generation, promote the progression of atherosclerosis (AS). Here, we aimed to investigate the effects of 2-acetamidophenol (2-AAP), which was selected by zebrafish phenotypic screening, in alleviating AS by relieving hyperlipidemia and inhibiting foam cell formation, as well as the underlying mechanisms. In a zebrafish hyperlipidemia model, 2-AAP increased lipid-lowering efficacy; alleviated TC, TG, LDL-C, and MDA levels; elevated HDL-C and T-SOD levels; significantly improved intravascular macrophage aggregation; and improved blood flow. In an ox-LDL-induced RAW264.7 model, 2-AAP inhibited lipid phagocytosis in RAW264.7 cells; reduced the intracellular TC, TG, FC, and CE contents; and decreased the CE/TC ratio, thus slowing foam cell generation. In addition, 2-AAP alleviated intracellular ROS and ferrous ion accumulation in RAW264.7 cells, reduced the MDA content, and increased GPX4 viability. Furthermore, transcriptome analyses and gene expression validation showed 2-AAP treatment upregulates genes related to GSH synthesis and transport, such as gclc, gclm, gss, and gpx4a, and enhanced the expression levels of genes involved in the storage and transportation of iron ions, such as fpn1, fth, and g6pd, indicating that 2-AAP dramatically regulated the ferroptosis and glutathione metabolic pathways. Overall, our study demonstrated that 2-AAP demonstrated potential in AS by alleviating hyperlipidemia and attenuating the ferroptosis pathway and provided evidence supporting the future application of 2-AAP in AS treatment. Full article
(This article belongs to the Special Issue Zebrafish Models in Marine Drug Discovery)
Show Figures

Figure 1

Figure 1
<p>Effects of 2-AAP treatment on lipid metabolism in zebrafish after egg yolk powder feeding. (<b>A</b>) Typical images of zebrafish stained with Oil Red O. The whole fish was shown on the left, and the enlarged fish tails were shown on the right. (<b>B</b>) IOD values of Oil Red O staining in the tail, <span class="html-italic">n</span> = 10. (<b>C</b>) The lipid lowering rate of 2-AAP treatment, <span class="html-italic">n</span> = 10. Effects of 2-AAP treatment on TC (<b>D</b>), TG (<b>E</b>), LDL-C (<b>F</b>), and HDL-C (<b>G</b>) levels. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 2
<p>2-AAP improves blood flow velocity and stability. (<b>A</b>) Schematic diagram of the location of blood flow velocity detection. (<b>B</b>) Real-time detection of zebrafish blood flow velocity waveforms. (<b>C</b>) Mean blood flow velocity in the tail vessels of zebrafish, <span class="html-italic">n</span> = 7. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 3
<p>2-AAP improves vascular inflammation. (<b>A</b>) Phenotypic diagram of decreased macrophage density caused by 2-AAP. The whole fish was shown on the left, and the enlarged fish tails were shown on the right. (<b>B</b>) IOD values of immune cells that accumulated in the tail, <span class="html-italic">n</span> = 12. (<b>C</b>) Phenotypic diagram of alleviation of intravascular macrophage aggregation by 2-AAP. The red arrow indicates intravascular macrophages. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 4
<p>2-AAP improves oxidative stress. (<b>A</b>) Phenotypic diagram of reduced ROS fluorescence intensity caused by 2-AAP. The whole fish was shown on the left, and the enlarged fish tails were shown on the right. (<b>B</b>) IOD values of ROS accumulated in the tail, <span class="html-italic">n</span> = 16. Effects of 2-AAP treatment on T-SOD (<b>C</b>), MDA (<b>D</b>), CAT (<b>E</b>), and T-AOC (<b>F</b>) levels. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>2-AAP reduces macrophage phagocytosis of oxidized lipids. (<b>A</b>) Oil Red O staining of macrophages. (<b>B</b>) Viability of RAW264.7 cells treated with 2-AAP. TG (<b>C</b>), TC (<b>D</b>), FC (<b>E</b>), CE (<b>F</b>), and CE/TC (<b>G</b>) contents in macrophages. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 6
<p>2-AAP improves oxidative stress and ferroptosis. (<b>A</b>) Typical diagram of ROS probe binding. (<b>B</b>) Statistics of the average fluorescence intensity of ROS. (<b>C</b>) MDA contents in the macrophages. (<b>D</b>) A typical FerroOrange diagram. (<b>E</b>) GPX4 vitality in macrophages. (<b>F</b>) Statistics of the average fluorescence intensity of FerroOrange. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 7
<p>Effects of 2-AAP on genes in zebrafish. (<b>A</b>) Venn diagram. (<b>B</b>) Differential expression gene statistics. (<b>C</b>) Volcano plots of 2-AAP-vs.-M. (<b>D</b>) Differential gene radar map of 2-AAP-vs.-M. The first circle from outside to inside represents upregulated genes (red) and downregulated genes (blue), and the size of the circle varies according to the ∣Log2FC∣ value. The data in the second outer circle represent the average expression of the 2-AAP group. Genes that were upregulated in expression after drug administration are shown as yellow spikes in the graph.</p>
Full article ">Figure 8
<p>Enrichment analysis. (<b>A</b>) Top 30 GO terms in AAP-vs.-M. (<b>B</b>) GO analysis radar chart in AAP-vs.-M. The first circle represents the items of GO enrichment, and the outside of the circle represents a ruler with respect to gene number. Different colors represent different GO classifications. The second circle represents the number of genes enriched in the GO terms, and the color indicates the <span class="html-italic">p</span> value; the greater the number of genes, the longer the bar, and the smaller the <span class="html-italic">p</span> value, the redder the color. The third circle represents gene expression trends. (<b>C</b>) Top 20 pathways enriched in the AAP-vs.-M comparison. (<b>D</b>) KEGG analysis radar chart of the AAP-vs.-M comparison.</p>
Full article ">Figure 9
<p>GSEA. (<b>A</b>) ES distribution map in ferroptosis. (<b>B</b>) ES distribution map of glutathione metabolism. (<b>C</b>) Differential gene-clustering heatmap for ferroptosis. (<b>D</b>) Differential gene-clustering heatmap of glutathione metabolism. The green line shows the distribution of ES for all genes. As depicted in the figure, when ES &gt; 0, the gene on the left side of the dashed line is the core gene, which contributed significantly to the enriched pathways.</p>
Full article ">Figure 10
<p>Expression of ferroptosis-, glutathione metabolism-, lipid metabolism-, inflammation-, and antioxidant-related genes following 2-AAP exposure. In the column chart, brown represents the blank group, blue represents the model group, and red represents the administration group. Compared with the control group, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Compared with the model group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">
13 pages, 2988 KiB  
Article
Hot-Melt Extrusion Drug Delivery System-Formulated Haematococcus pluvialis Extracts Regulate Inflammation and Oxidative Stress in Lipopolysaccharide-Stimulated Macrophages
by Tae-Young Gil, Ha-Yeon Sim, Ha-Yeon Lee, Suji Ryu, Jong-Suep Baek, Dae Geun Kim, Jaehoon Sim and Hyo-Jin An
Mar. Drugs 2024, 22(11), 512; https://doi.org/10.3390/md22110512 - 13 Nov 2024
Viewed by 595
Abstract
Haematococcus pluvialis contains valuable bioactive compounds, including astaxanthin, proteins, and fatty acids. Astaxanthin is known for its various health benefits, such as preserving the redox balance and reducing inflammation. However, its low stability and poor water solubility present challenges for various applications. Hot-melt [...] Read more.
Haematococcus pluvialis contains valuable bioactive compounds, including astaxanthin, proteins, and fatty acids. Astaxanthin is known for its various health benefits, such as preserving the redox balance and reducing inflammation. However, its low stability and poor water solubility present challenges for various applications. Hot-melt extrusion (HME) technology enhances the aqueous solubility of H. pluvialis extracts, increasing the usable astaxanthin content through nanoencapsulation (HME-DDS-applied extracts, ASX-60F and ASX-100F). This study compared the effects of HME-DDS-derived extracts (ASX-60F and ASX-100F) and the non-applied extract (ASX-C) under inflammatory and oxidative stress conditions. In animal models of sepsis, 60F and 100F treatment exhibited higher survival rates and a lower expression of pro-inflammatory biomarkers compared to those treated with C. In lipopolysaccharide-stimulated RAW 264.7 macrophages, nitric oxide (NO) production and the expression of pro-inflammatory mediators such as cyclooxygenase-2 and inducible NO synthase were reduced by 60F or 100F treatments via ERK/p-38 mitogen-activated protein kinase (MAPK) signaling. Moreover, 60F or 100F inhibited reactive oxygen species production regulated by nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling. Collectively, these findings suggest that HME-DDS-derived H. pluvialis extracts exert anti-inflammatory and antioxidant effects by inhibiting MAPK phosphorylation and activating Nrf2/HO-1 expression. Full article
(This article belongs to the Section Marine Pharmacology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>ASX-C, ASX-60F, or ASX-100F reduced mortality and inhibited pro-inflammatory production in mouse models of LPS-induced septic shock. Effects of ASX-C, ASX-60F, or ASX-100F on mortality and inflammatory mediators in LPS-induced sepsis mice model. Mice (<span class="html-italic">n</span> = 5~6 per group) were orally administered (per os, p.o.) with ASX-C, ASX-60F, ASX-100F, or Dexa for 1 h followed by 25 mg/kg of LPS i.p. Dexa (5mg/kg, p.o.) stands for dexamethasone (positive control). (<b>A</b>) Survival rates of mice monitored for 120 h. (<b>B</b>–<b>E</b>) Liver tissue obtained from mice 4 h after LPS injection. Production of pro-inflammatory mediators, NO (<b>B</b>) and PGE<sub>2</sub> (<b>D</b>); production was measured using EIA enzyme immune assay (EIA). Pro-inflammatory markers, iNOS (<b>C</b>) and COX2 (<b>E</b>), were determined by using Western blotting. β-actin served as an internal control. Data are shown as mean ± S.D. ### <span class="html-italic">p</span> &lt; 0.001 vs. normal; *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS; + <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 vs. ASX-C.</p>
Full article ">Figure 2
<p>ASX-C, ASX-60F, or ASX-100F reduced pro-inflammatory cytokines and phosphorylation of pro-inflammatory signaling protein expression in mouse models of LPS-induced septic shock. Effects of ASX-C, ASX-60F, or ASX-100F on pro-inflammatory biomarkers in LPS-induced sepsis mice model. Mice (<span class="html-italic">n</span> = 5~6 per group) were orally administered with ASX-C, ASX-60F, ASX-100F, or Dexa. for 1 h followed by 25 mg/kg of LPS i.p. (<b>A</b>) Pro-inflammatory cytokines were evaluated using enzyme immune assay EIA. (<b>B</b>,<b>C</b>) Proteins from liver tissue of septic shock mice were determined by using Western blotting assay with β-actin or total form of ERK and p38 as internal controls. Data are shown as mean ± S.D. ### <span class="html-italic">p</span> &lt; 0.001 vs. normal; ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS; + <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 vs. ASX-C.</p>
Full article ">Figure 3
<p>ASX-C, ASX-60F, or ASX-100F suppressed the expression of inducible enzyme and mediators via Akt-ERK/p38 MAPK signaling pathways in LPS-stimulated RAW264.7 macrophages. Effects of ASX-C, ASX-60F, or ASX-100F on Akt-ERK/p38 MAPK pathways in LPS-stimulated RAW264.7 macrophages model. (<b>A</b>) Levels of NO were determined using Griess reagent. Positive control was incubated with l-NIL (20 μM). Western blots of iNOS and COX2 (<b>B</b>); phosphorylation of Akt (<b>C</b>), ERK, or p-38 (<b>D</b>) was determined as protein expressions with β-actin or total form of Akt, ERK, or p38 as internal controls. Data are shown as mean ± S.D. ### <span class="html-italic">p</span> &lt; 0.001 vs. normal; * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS; ++ <span class="html-italic">p</span> &lt; 0.01, +++ <span class="html-italic">p</span> &lt; 0.001 vs. ASX-C.</p>
Full article ">Figure 4
<p>Effects of ASX-C, ASX-60F, or ASX-100F on ROS-generation in LPS-stimulated RAW264.7 macrophages model. (<b>A</b>) Cells were pre-treated with or without indicated materials (ASX-C, ASX-60F, or ASX-100F) for 1 h and incubated with LPS for 2 h, and the levels of ROS were measured using FACS. (<b>B</b>) Graph indicates relative percentages of ROS-positive cells. Data are shown as mean ± S.D. ### <span class="html-italic">p</span> &lt; 0.001 vs. normal; * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 vs. LPS; ++ <span class="html-italic">p</span> &lt; 0.01, +++ <span class="html-italic">p</span> &lt; 0.001 vs. ASX-C.</p>
Full article ">Figure 5
<p>Effects of ASX-C, ASX-60F, or ASX-100F on Nrf2 and HO-1 protein expressions in LPS-stimulated RAW264.7 macrophage model. Cells were pre-treated with or without indicated materials (ASX-C, ASX-60F, or ASX-100F) for 1 h and incubated with LPS for 12 h. (<b>A</b>) Nrf2 and (<b>B</b>) HO-1 protein expressions were measured by using Western blotting assay. β-actin and PARP-1 served as internal controls for cytosolic fraction (C) and nuclear fraction (N), respectively. Data are shown as mean ± S.D. ### <span class="html-italic">p</span> &lt; 0.001 vs. normal; *** <span class="html-italic">p</span> &lt; 0.001 vs. LPS.</p>
Full article ">Figure 6
<p>A schematic illustration of <span class="html-italic">H. pluvialis</span> production via hot-melt extrusion (HME) process.</p>
Full article ">
12 pages, 1043 KiB  
Article
Nine New Glycosylated Compounds from the Leaves of the Medicinal Plant Malus hupehensis
by Lin-Lin Yuan, Yi Wang, Guo-Kai Wang and Ji-Kai Liu
Molecules 2024, 29(22), 5269; https://doi.org/10.3390/molecules29225269 - 7 Nov 2024
Viewed by 460
Abstract
Nine new compounds (19), including four dihydrochalcone glycosides, two dibenzofuran glycosides, and two biphenyl glycosides, were isolated from the leaves of the medicinal plant Malus hupehensis collected in Shennongjia Forestry District (Hubei, China). Their structures were elucidated by comprehensive [...] Read more.
Nine new compounds (19), including four dihydrochalcone glycosides, two dibenzofuran glycosides, and two biphenyl glycosides, were isolated from the leaves of the medicinal plant Malus hupehensis collected in Shennongjia Forestry District (Hubei, China). Their structures were elucidated by comprehensive spectroscopic techniques, including HRESIMS and NMR spectra. All compounds were tested by preliminary biological evaluation for their α-glucosidase inhibitory and NO production activities. Compound 4 was found to show significant inhibitory activity against NO production in LPS-activated RAW 264.7 macrophage cells with an IC50 value of 29.60 μM, and compounds 3 and 4 were found to exhibit potent α-glucosidase inhibition with IC50 values of 44.17 and 60.15 μM, respectively. This work represents the first report of the diverse glycosides from the plant Malus hupehensis. It expands our understanding of the secondary metabolites of this medicinal plant and lays the foundation for the study of the bioactive principles of the ethnic hypoglycemic medicinal plant. Full article
(This article belongs to the Section Bioorganic Chemistry)
Show Figures

Figure 1

Figure 1
<p>Key HMBC, <sup>1</sup>H-<sup>1</sup>H COSY, and ROESY correlations of <b>1</b>–<b>9</b>.</p>
Full article ">Figure 2
<p>Structures of compounds <b>1</b>–<b>9</b>.</p>
Full article ">Figure 3
<p>Molecular docking of <b>3</b> and <b>4</b> and α-glucosidase. The 3D structure of α-glucosidase is shown in blue slate color, ligands are shown in cyan, side chain amino acids are shown in violet, and distances (Angstrom) are shown in black; (<b>A</b>) interactions of <b>3</b>; (<b>B</b>) interactions of <b>4</b>.</p>
Full article ">
17 pages, 4942 KiB  
Article
Anti-Inflammatory Effects and Metabolomic Analysis of Ilex Rotunda Extracted by Supercritical Fluid Extraction
by Duc Dat Le, Young Su Jang, Vinhquang Truong, Thientam Dinh, Thinhulinh Dang, Soojung Yu and Mina Lee
Int. J. Mol. Sci. 2024, 25(22), 11965; https://doi.org/10.3390/ijms252211965 - 7 Nov 2024
Viewed by 432
Abstract
Ilex rotunda is a famous medicinal plant with many ethnopharmacological uses. It is traditionally employed for treating inflammation and cardiovascular diseases. In this study, we established green technology to extract the leaves and twigs of I. rotunda. The obtained extracts and [...] Read more.
Ilex rotunda is a famous medicinal plant with many ethnopharmacological uses. It is traditionally employed for treating inflammation and cardiovascular diseases. In this study, we established green technology to extract the leaves and twigs of I. rotunda. The obtained extracts and their fractions were evaluated for their anti-inflammatory potential. In cytokine assays, the extract, n-hexane (H), methylene chloride (MC), and EtOAc (E) fractions of the twigs of I. rotunda significantly inhibited lipopolysaccharide (LPS)-induced nitric oxide (NO), interleukin (IL)-6, and tumor necrosis factor (TNF)-α production in RAW264.7 macrophages. Furthermore, the extract, H, and MC fractions of the leaves of I. rotunda modulated cytokine expression by downregulating LPS-induced NO, IL-6, and TNF-α production in RAW264.7 macrophages. Western blotting analysis revealed that the extracts and fractions of the leaves and twigs of I. rotunda inhibited inflammatory cytokines by inactivating nuclear factor kappa B (NFκB) action by reducing the phosphorylation of transcript factor (p65) and nuclear factor-kappa B inhibitor alpha (IκBα) degradation, or by inactivating mitogen-activated protein kinase (MAPK) through the p38 or ERK signaling pathways via the active ingredients of the leaves and twigs of I. rotunda. Ultra-high-resolution liquid chromatography–Orbitrap mass analysis (UHPLC–ESI-Orbitrap-MS/MS)-based molecular networking, in cooperation with social open platform-guided isolation and dereplication, led to the identification of metabolites in this plant. Our findings indicate that the leaves and twigs of I. rotunda could be promising candidates for developing therapeutic strategies to treat anti-inflammatory diseases. Full article
(This article belongs to the Special Issue Molecular Pharmacology of Medicinal Plants)
Show Figures

Figure 1

Figure 1
<p>Antioxidative effects of leaf extract and fractions (<b>A</b>,<b>C</b>) and twig extract and fractions (<b>B</b>,<b>D</b>) were accessed by scavenging activity of DPPH and ABTS radicals. Samples were assayed at 10 and 100 µg/mL in triplicates. Values are expressed as means ± SD. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to positive control (Ascorbic acid, AA).</p>
Full article ">Figure 2
<p>Cytotoxic effects of leaf (<b>A</b>,<b>B</b>) and twig (<b>C</b>,<b>D</b>) extracts and their fractions on cell survival at different concentrations. Cell viability was assessed using the MTT assay. RAW264.7 cells were treated with LPS or samples in a dose-dependent manner at concentrations of 10, 20, 30, 50, and 100 µg/mL and repeated three times. Values are expressed as means ± SD. Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01, compared to the control (CTL).</p>
Full article ">Figure 3
<p>The inhibitory effects of the leaf and twig extracts and their fractions on NO (<b>A</b>,<b>B</b>), IL-6 (<b>C</b>,<b>D</b>), and TNF-α (<b>E</b>,<b>F</b>) production induced by LPS-activated RAW264.7 cells. Cells were pretreated with tested samples (10, 20, and 30 µg/mL) for 1 h and then stimulated with LPS (5 ng/mL) for 24 h. NO, IL-6, and TNF-α production in the culture media were quantified using the Griess assay and an enzyme immunoassay (EIAISA) kit, respectively. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to controls.</p>
Full article ">Figure 4
<p>Western blot analysis of the effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on iNOS, COX-2 expression levels. LPS-induced RAW264.7 macrophages were pretreated with samples at concentrations of 30 μg/mL and incubated for 6 h. Total proteins were then isolated, separated by SDS-PAGE, and immunoblotted using specific p-p65, p-IκBα, and IκBα antibodies. β-Actin served as an internal control. Relative optical density ratio vs. β-actin or total form was determined using a densitometric analysis program (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA), normalized to internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 compared to non-sample in LPS stimulation.</p>
Full article ">Figure 5
<p>Inhibitory effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on LPS-induced NF-κB signaling pathway in RAW264.7 macrophages. Cells were pretreated with MC fraction from twigs (20 μg/mL) and other samples (30 μg/mL) for 1 h, followed by LPS stimulation for 15 min. Proteins were isolated, separated using SDS-PAGE, and immunoblotted with antibodies specific to p-p65, p-IκBα, IκBα. β-actin was served as internal control. Relative optical density ratio versus β-actin or the total form was determined using densitometric analysis software (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA) and normalized against internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, compared to non-sample under LPS stimulation.</p>
Full article ">Figure 6
<p>The inhibitory effects of leaf extract and fractions (<b>A</b>) and twig extract and fractions (<b>B</b>) on the LPS-induced MAPK signaling pathway in RAW264.7 macrophages. After pretreating cells with the MC fraction of twigs (20 μg/mL) and other samples (30 μg/mL) for 1 h, cells were stimulated with LPS for 15 min. Total proteins were then isolated, separated by SDS-PAGE, and immunoblotted using specific antibodies for p-p38, p-ERK, and ERK. β-Actin was served as an internal control. The relative optical density ratio vs. β-actin or total form was determined using a densitometric analysis program (Bio-Rad Quantity One Software, version 4.6.3 (Basic), Bio-Rad Laboratories Inc., Hercules, CA, USA) normalized to the internal control. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the control in LPS stimulation.</p>
Full article ">Figure 7
<p>The cell viability (<b>A</b>,<b>B</b>) and inhibitory effects of leaf and twig extracts and their fractions on IL-2 production (<b>C</b>,<b>D</b>) induced by CD3-CD28 Jurkat T cells. Cells were pretreated with the samples (10, 50, and 100 µg/mL) for 1 h and subsequently stimulated with CD3-CD28 (7 µg/mL and 2 µg/mL CD3 and CD28, respectively) for 24 h. IL-2 production in the culture media was quantified using an enzyme-linked immunosorbent assay (ELISA) kit. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 vs. non-treated group (CTL). Differences were significant at * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared to the non-sample control in CD3-CD28 stimulation.</p>
Full article ">Figure 8
<p>The chromatograms of H (A, black), E (B, red), B (C, green), MC (D, blue), and W fractions (E, yellow) and extracts (F, magenta) of leaves of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span> detected in TIC for negative ion mode.</p>
Full article ">Figure 9
<p>Feature-based molecular network of extract (Red) and fractions [H: Blue; MC: Orange; E: Violet; B: Dark Green; W: Yellow] of twigs (GNPS molecular network with job ID = 96ad1b5bbed84c8bb073d8c882f3897b) of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span>.</p>
Full article ">Figure 10
<p>Feature-based molecular network of extract (Neon Blue) and fractions [H: Black; MC: Purple; E: Magenta; B: Yellow; W: Blue] of leaves (GNPS molecular network with job ID = cfa6443e13824edf8132a3f391ad91f3) of <span class="html-italic">I</span>. <span class="html-italic">rotunda</span>.</p>
Full article ">
13 pages, 3667 KiB  
Article
Structural Characteristics and Immunological Function of a New Non-Starch Polysaccharide from Red Sprout Taro
by Sha Luo, Yao Xiao, Asjad Ali, Qianglong Zhu, Nan Shan, Jingyu Sun, Shenglin Wang, Jianhui Xiao, Yingjin Huang and Qinghong Zhou
Foods 2024, 13(22), 3531; https://doi.org/10.3390/foods13223531 - 5 Nov 2024
Viewed by 562
Abstract
Taro is a tuber crop that is used for nutritional and medicinal purposes due to its abundance of non-starch polysaccharides (NSPs). Red Sprout taro is a local variety in Southern China, but the characteristics and bioactivities of its NSPs are currently unknown. In [...] Read more.
Taro is a tuber crop that is used for nutritional and medicinal purposes due to its abundance of non-starch polysaccharides (NSPs). Red Sprout taro is a local variety in Southern China, but the characteristics and bioactivities of its NSPs are currently unknown. In this study, NSPs were isolated from the corms of Red Sprout taro using hot-water extraction, ion-exchange chromatography, and ethanol precipitation; their molecular weight, monosaccharide composition, structural formulae, and immunomodulatory effects were examined. A novel NSP named Colocasia esculenta polysaccharide 1 (CEP1) was purified and characterized and was shown to mainly consist of glucose (60.49%) and galactose (25.92%) and have a molecular weight of 4556.272 kDa. The backbone of CEP1 consisted primarily of →4)-α-D-Glcp-(1→, →4,6)-β-D-Galp-(1→, and →3)-β-D-Galp-(1→ residues, with a branch consisting of the β-D-Glcp-(1→ residue. In addition, 25–400 µg/mL CEP1 was shown to have immunomodulatory effects on RAW264.7 macrophages. CEP1 not only increased cell viability, phagocytic capacity, inducible nitric oxide synthase secretion, and nitric oxide generation in RAW264.7 cells, but it also activated M1 and M2 macrophages to generate tumor necrosis factor α, interleukin 6, transforming growth factor β, and interleukin 10. These findings could lead to the use of CEP1 from Red Sprout taro as a possible immunomodulatory polysaccharide in functional foods. Full article
(This article belongs to the Section Food Physics and (Bio)Chemistry)
Show Figures

Figure 1

Figure 1
<p>The elution profile of the crude polysaccharides of <span class="html-italic">C. esculenta</span> cv. Red Sprout on a DEAE-52 cellulose column. CEP1, CEP2, and CEP3 were collected from tube 3 to tube 19, from tube 32 to tube 42, and from tube 55 to tube 56, respectively.</p>
Full article ">Figure 2
<p>The FT-IR spectra of the polysaccharide CEP1.</p>
Full article ">Figure 3
<p>Nuclear magnetic resonance (NMR) spectral analysis of CEP1 from C. esculenta cv. Red Sprout. (<b>A</b>) 1H-NMR of CEP1; (<b>B</b>) 13C-NMR of CEP1; (<b>C</b>) 1H-1H correlation spectroscopy (COSY) of CEP1; (<b>D</b>) heteronuclear single quantum correlation (HSQC) of CEP1; (<b>E</b>) heteronuclear multiple-bond correlation (HMBC) of CEP1; (<b>F</b>) nuclear Overhauser effect spectroscopy (NOESY) of CEP1.</p>
Full article ">Figure 4
<p>Projected structure of CEP1.</p>
Full article ">Figure 5
<p>Impacts of CEP1 on the viability of RAW264.7 macrophages. RAW264.7 macrophages were cultured for 6 h and treated with 5 μg/mL LPS (positive control), DMEM (control), and 25, 50, 100, 200, 400, and 800 μg/mL CEP1 for 24 h at 37 °C in 5% CO<sub>2</sub>. Cell viability was examined using the methyl thiazole tetrazolium (MTT) method. The experiments were repeated six times. The data are expressed as the mean ± standard deviation. * <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 by one-way ANOVA and Dunnett’s test.</p>
Full article ">Figure 6
<p>Impacts of CEP1 on the phagocytic activity of RAW264.7 macrophages. RAW264.7 macrophages were cultured for 6 h and treated with 1 μg/mL LPS (positive control), DMEM (control), and 25, 100, and 400 μg/mL CEP1 for 24 h at 37 °C and 5% CO<sub>2</sub>. The phagocytic activity of the RAW264.7 macrophages was assessed with a Vybrant™ Phagocytosis test kit. The experiments were performed six times. The data are expressed as the mean ± standard deviation. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA and Dunnett’s test.</p>
Full article ">Figure 7
<p>Activation impacts of CEP1 on M1 macrophages. Impact of CEP1 on the phagocytic activity of RAW264.7 macrophages. RAW264.7 macrophages were cultured for 6 h and treated with 1 μg/mL LPS (positive control), DMEM (control), and 25, 100, and 400 μg/mL CEP1 for 24 h at 37 °C and 5% CO<sub>2</sub>. The protein levels of IL-6 (<b>A</b>), TNF-α (<b>B</b>), and INOS (<b>C</b>) as well as NO production (<b>D</b>) were analyzed via ELISA kits. The experiments were repeated three times. The data are expressed as the mean ± standard deviation. * <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 by one-way ANOVA and Dunnett’s test.</p>
Full article ">Figure 8
<p>Activating impacts of CEP1 on M2 macrophages RAW264.7 macrophages were cultured for 6 h and treated with 20 ng/mL IL-4 (positive control), DMEM (control), and 25, 100, and 400 μg/mL CEP1 for 24 h at 37 °C and 5% CO<sub>2</sub>. The protein levels of IL-10 (<b>A</b>) and TGF-β (<b>B</b>) were quantified using ELISA kits. The experiments were triplicated. The data are expressed as the mean ± standard deviation. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA and Dunnett’s test.</p>
Full article ">
14 pages, 3513 KiB  
Article
Digital Holographic Microscopy in Veterinary Medicine—A Feasibility Study to Analyze Label-Free Leukocytes in Blood and Milk of Dairy Cows
by Sabine Farschtschi, Manuel Lengl, Stefan Röhrl, Christian Klenk, Oliver Hayden, Klaus Diepold and Michael W. Pfaffl
Animals 2024, 14(21), 3156; https://doi.org/10.3390/ani14213156 - 3 Nov 2024
Viewed by 898
Abstract
For several years, the determination of a differential cell count of a raw milk sample has been proposed as a more accurate tool for monitoring the udder health of dairy cows compared with using the absolute somatic cell count. However, the required sample [...] Read more.
For several years, the determination of a differential cell count of a raw milk sample has been proposed as a more accurate tool for monitoring the udder health of dairy cows compared with using the absolute somatic cell count. However, the required sample preparation and staining process can be labor- and cost-intensive. Therefore, the aim of our study was to demonstrate the feasibility of analyzing unlabeled blood and milk leukocytes from dairy cows by means of digital holographic microscopy (DHM). For this, we trained three different machine learning methods, i.e., k-Nearest Neighbor, Random Forests, and Support Vector Machine, on sorted leukocyte populations (granulocytes, lymphocytes, and monocytes/macrophages) isolated from blood and milk samples of three dairy cows by using fluorescence-activated cell sorting. Afterward, those classifiers were applied to differentiate unlabeled blood and milk samples analyzed by DHM. A total of 70 blood and 70 milk samples were used. Those samples were collected from five clinically healthy cows at 14-time points within a study period of 26 days. The outcome was compared with the results of the same samples analyzed by flow cytometry and (in the case of blood samples) also to routine analysis in an external laboratory. Moreover, a standard vaccination was used as an immune stimulus during the study to check for changes in cell morphology or cell counts. When applied to isolated leukocytes, Random Forests performed best, with a specificity of 0.93 for blood and 0.84 for milk cells and a sensitivity of 0.90 and 0.81, respectively. Although the results of the three analytical methods differed, it could be demonstrated that a DHM analysis is applicable for blood and milk leukocyte samples with high reliability. Compared with the flow cytometric results, Random Forests showed an MAE of 0.11 (SD = 0.04), an RMSE of 0.13 (SD = 0.14), and an MRE of 1.00 (SD = 1.11) for all blood leukocyte counts and an MAE of 0.20 (SD = 0.11), an RMSE of 0.21 (SD = 0.11) and an MRE of 1.95 (SD = 2.17) for all milk cell populations. Further studies with larger sample sizes and varying immune cell compositions are required to establish method-specific reference ranges. Full article
(This article belongs to the Section Cattle)
Show Figures

Figure 1

Figure 1
<p>Schematic overview of the workflow.</p>
Full article ">Figure 2
<p>Sampling scheme of blood and milk samples. Blood and milk samples were collected from each of the five cows at 14 time points. All cows were vaccinated on day 8 after sampling.</p>
Full article ">Figure 3
<p>Representative false-color phase images of different populations of unlabeled leukocytes, analyzed in DHM. (<b>A</b>) Blood granulocyte; (<b>B</b>) Blood lymphocyte; (<b>C</b>) Blood monocyte; (<b>D</b>) Milk granulocyte; (<b>E</b>) Milk lymphocyte; (<b>F</b>) Milk macrophage.</p>
Full article ">Figure 4
<p>Exemplary scatter plots of flow cytometric and digital holographic microscopy analyses. (<b>A</b>) Blood leukocyte populations analyzed by FACS; (<b>B</b>) Blood leukocyte populations analyzed by DHM; (<b>C</b>) Milk leukocyte populations analyzed by FACS; (<b>D</b>) Milk leukocyte populations analyzed by DHM.</p>
Full article ">Figure 5
<p>Confusion matrix showing the results of Random Forest classification of sorted blood cells.</p>
Full article ">Figure 6
<p>Confusion matrix showing the results of Random Forest classification of sorted milk cells.</p>
Full article ">Figure 7
<p>Cell count progression over time, DHM results obtained using k-Nearest Neighbor. (<b>A</b>) Blood cells of cow #963, analyzed by DHM, FACS and external laboratory; (<b>B</b>) Milk cells of cow #963, analyzed by DHM and FACS.</p>
Full article ">
15 pages, 2945 KiB  
Article
Selective PPARδ Agonist GW501516 Protects Against LPS-Induced Macrophage Inflammation and Acute Liver Failure in Mice via Suppressing Inflammatory Mediators
by Hyun-Joung Lim and Hyun Jeong Kwak
Molecules 2024, 29(21), 5189; https://doi.org/10.3390/molecules29215189 - 2 Nov 2024
Viewed by 798
Abstract
Inflammation is critical in the development of acute liver failure (ALF). Peroxisome proliferator-activated receptor delta (PPARδ) regulates anti-inflammatory responses and is protective in several diseases such as obesity and cancer. However, the beneficial effects and underlying mechanisms of PPARδ agonist GW501516 in ALF [...] Read more.
Inflammation is critical in the development of acute liver failure (ALF). Peroxisome proliferator-activated receptor delta (PPARδ) regulates anti-inflammatory responses and is protective in several diseases such as obesity and cancer. However, the beneficial effects and underlying mechanisms of PPARδ agonist GW501516 in ALF remain unclear. This study investigated the molecular mechanisms underlying the anti-inflammatory effects of GW501516 in macrophages and assessed its protective potential against lipopolysaccharide (LPS)/galactosamine (GalN)-induced ALF. In vivo administration of GW501516 significantly reduced LPS/GalN-induced hepatotoxicity, as evidenced by lower mortality, decreased liver damage, and attenuated secretion of IL-1β, IL-6, and TNF-α. GW501516 treatment also decreased LPS-induced nitric oxide synthase 2 (NOS2) expression and nitric oxide (NO) production in RAW264.7 cells, an effect reversed by PPARδ siRNA. Additionally, GW501516 inhibited LPS-induced phosphorylation of p38 and c-Jun N-terminal kinase (JNK), suggesting that inactivation of these MAPKs contributes to its effects. The secretion of IL-6, TNF-α, and NF-κB DNA-binding activity were also suppressed by GW501516, while the nuclear translocation of the NF-κB p65 subunit was unaffected. In conclusion, our findings suggest that GW501516 exerts protective effects in ALF by inhibiting the production of inflammatory mediators. Therefore, GW501516 may act as a potential agent for developing anti-inflammatory therapies for ALF. Full article
Show Figures

Figure 1

Figure 1
<p>In vivo effects of GW501516 in mice with ALF induced by LPS/D-GalN. Mice were pretreated with GW501516 (2 mg/kg, ip) at 6 h prior to LPS/D-GalN (20 mg/700 mg/kg) challenge. Survival rate of mice were monitored for 5 h after LPS/D-GalN challenge (<b>A</b>). Serum levels of ALT and AST were measured (<b>B</b>,<b>C</b>). Representative gross liver morphology (<b>D</b>), representative images of H&amp;E-stained liver section (<b>E</b>), and liver injury scores were shown (<b>F</b>). Data shown are means ± SD at least 10 different animals per group. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal mice; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-GalN-treated mice.</p>
Full article ">Figure 2
<p>Effects of GW501516 on pro-inflammatory cytokines production in LPS/D-GalN-induced ALF mice. Mice were pretreated with GW501516 (2 mg/kg, ip) at 6 h prior to LPS/D-GalN (20 mg/700 mg/kg) challenge. Serum levels of (IL-6), IL-1β, TNF-α, (<b>A</b>–<b>C</b>) and mRNA levels of IL-6, IL-1β, and NOS2 in the hepatic tissue (<b>D</b>–<b>F</b>) were measured. Hepatic tissues performed immunohistochemistry against NOS2 antibody (red arrows). (<b>G</b>) % of NOS2-positive areas of were calculated using Image J 1.53. All values represent the mean ± SD from three or more independent experiments; <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal mice; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-GalN-treated mice.</p>
Full article ">Figure 3
<p>PPARδ activation decreases NOS2 expression and subsequent NO production in LPS-activated RAW264.7 cells. Relative expression levels of NOS2 mRNA (<b>A</b>) and protein (<b>B</b>); the nitrite levels (<b>C</b>) were measured in RAW264.7 cells activated with LPS (100 ng/mL), with or without pretreatment with GW501516. RAW264.7 cell transfected with PPARδ-specific siRNA were treated with LPS in the presence or absence of GW501516 pretreatment. Expression levels of PPARδ and NOS2 were detected by Western blot (<b>D</b>). Csi: scrambled control siRNA, Psi: PPARδ-specific siRNA. PPARδ-overexpressed cells were treated with LPS with or without GW501516 pretreatment. PPARδ and NOS2 expressions were detected by Western blot (<b>E</b>). Ad/C: control adenovirus, Ad/P: PPARδ adenovirus. All quantifications were analyzed using Image J. Data shown are means ± SD of at least three experiments (each performed in duplicates). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal control; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-treated cells.</p>
Full article ">Figure 4
<p>Effect of GW501516 on the regulation of MAPkinase in LPS-activated RAW264.7 cells. NOS2 expressions with p38, JNK, and ERK1/2 inhibitor treatment (<b>A</b>). Expressions of p-p38, p-JNK, and p-ERK1/2 after LPS activation with or without GW501516 pretreatment. SB203580 (<b>B</b>): p38 inhibitor, PD98059: ERK1/2 inhibitor, SP600125: JNK inhibitor. All of quantifications were analyzed by Image J1.53. Data shown are means ± SD of at least three experiments (each performed in duplicates). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal control; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-treated cells.</p>
Full article ">Figure 5
<p>PPARδ agonist GW501516 significantly inhibits expressions of pro-inflammatory cytokines in LPS-activated RAW264.7 cells. mRNA expression of TNF-α (<b>A</b>) and IL-6 (<b>B</b>) in LPS (100 ng/mL)-stimulated cells with or without GW501516 (100 nM) pretreatment. The amount of TNF-α (<b>C</b>) and IL-6 (<b>D</b>) secreted into culture medium was measured by using ELISA kit specific for respective cytokines. All quantifications were analyzed using Image J1.53. Data shown are means ± SD of at least three experiments (each performed in duplicates). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal control; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-treated cells.</p>
Full article ">Figure 6
<p>Treatment of PPARδ agonist GW501516 decreases NF-κB binding to DNA, not the nuclear translocation of p65 subunit, in LPS-stimulated RAW264.7 cells. Cells were pretreated with 3 μM BAY-117082 for 30 min and then incubated with LPS for 24 h. NOS2 expression was determined by Western blot (<b>A</b>). Cells were pretreated with GW501516 for 12 h prior to incubation of LPS for 30 min. Cytoplasmic or nuclear proteins were extracted and detected via Western blot against p65 (<b>B</b>). Quantification data of nuclear translocation of p65 subunit expressed as p65 (nucleus)/p65 (cytosol) (<b>C</b>). Representative IF-staining images of p65 subunit (<b>D</b>), and the nuclear proteins were extracted and subjected to EMSA to analyzed the p65 DNA-binding activity (<b>E</b>). Scale bar = 100 μm. Data shown are means ± SD of at least three experiments (each performed in duplicates). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 versus normal control; * <span class="html-italic">p</span> &lt; 0.05 vs. LPS-treated cells.</p>
Full article ">
Back to TopTop