[go: up one dir, main page]

 
 
nutrients-logo

Journal Browser

Journal Browser

Functional Evaluation of Edible Mushrooms and Their Active Materials

A special issue of Nutrients (ISSN 2072-6643). This special issue belongs to the section "Phytochemicals and Human Health".

Deadline for manuscript submissions: 30 November 2024 | Viewed by 6556

Special Issue Editors


E-Mail Website
Guest Editor
1. Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
2. School of Life Sciences, Jilin University, Changchun 130012, China
Interests: fungi; natural compounds; pharmacological efficacy
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
Interests: fungi; natural compounds; pharmacological efficacy; brain diseases

E-Mail Website
Guest Editor
Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China
Interests: active polysaccharide; fungi

Special Issue Information

Dear Colleagues,

In recent years, edible mushrooms have been increasingly studied due to the various effects they can have on human health. Mushrooms, such as Ganoderma sichuanense and Lentinula edodes, have been applied in clinical settings. However, many edible mushrooms are not well-known, and their properties have not yet been thoroughly studied. Additionally, the mechanisms involved in their effects, including their target cells and molecules, remain unclear.

The goal of this Special Issue is to explore the function of mushrooms and their active compounds and to clarify their mechanism of action and targets. Submitted manuscripts should adhere to the following requirements and scope:

  1. The studied active materials must include an edible mushroom (including wild or cultivated fungi) or its active ingredient(s), which should have strong application prospects;
  2. The pharmacological effects of the mushroom(s) or their active material(s) should be described, which includes, but is not limited to, immune-regulatory, anti-inflammatory, anti-neurodegeneration, antioxidative and antitumor properties. Data obtained from in vitro experiments must be confirmed using animal models or, preferably, through clinical trials;
  3. The study must clarify the mechanisms of action of the studied compounds, with a clear delineation of the targets, such as cells and cytokines associated with the identified efficacies.

Prof. Dr. Di Wang
Dr. Chunyue Wang
Dr. Yang Liu
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Nutrients is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2900 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • edible mushroom
  • active materials
  • efficacies
  • animal models
  • clinical trials
  • target cells/molecules

Published Papers (5 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

17 pages, 69108 KiB  
Article
Ganoderma lucidum Polysaccharides Ameliorate Acetaminophen-Induced Acute Liver Injury by Inhibiting Oxidative Stress and Apoptosis along the Nrf2 Pathway
by Nan Zhang, Zhongming Han, Rui Zhang, Linling Liu, Yanliang Gao, Jintao Li and Meixia Yan
Nutrients 2024, 16(12), 1859; https://doi.org/10.3390/nu16121859 - 13 Jun 2024
Viewed by 667
Abstract
The excessive employment of acetaminophen (APAP) is capable of generating oxidative stress and apoptosis, which ultimately result in acute liver injury (ALI). Ganoderma lucidum polysaccharides (GLPs) exhibit hepatoprotective activity, yet the protective impact and potential mechanism of GLPs in relation to APAP-induced ALI [...] Read more.
The excessive employment of acetaminophen (APAP) is capable of generating oxidative stress and apoptosis, which ultimately result in acute liver injury (ALI). Ganoderma lucidum polysaccharides (GLPs) exhibit hepatoprotective activity, yet the protective impact and potential mechanism of GLPs in relation to APAP-induced ALI remain ambiguous. The intention of this research was to scrutinize the effect of GLPs on APAP-induced ALI and to shed light on their potential mechanism. The results demonstrated that GLPs were capable of notably alleviating the oxidative stress triggered by APAP, as shown through a significant drop in the liver index, the activities of serum ALT and AST, and the amounts of ROS and MDA in liver tissue, along with an increase in the levels of SOD, GSH, and GSH-Px. Within these, the hepatoprotective activity at the high dose was the most conspicuous, and its therapeutic efficacy surpassed that of the positive drug (bifendate). The results of histopathological staining (HE) and apoptosis staining (TUNEL) indicated that GLPs could remarkably inhibit the necrosis of hepatocytes, the permeation of inflammatory cells, and the occurrence of apoptosis induced by APAP. Moreover, Western blot analysis manifested that GLPs enhanced the manifestation of Nrf2 and its subsequent HO-1, GCLC, and NQO1 proteins within the Nrf2 pathway. The results of qPCR also indicated that GLPs augmented the expression of antioxidant genes Nrf2, HO-1, GCLC, and NQO1. The results reveal that GLPs are able to set off the Nrf2 signaling path and attenuate ALI-related oxidative stress and apoptosis, which is a potential natural medicine for the therapy of APAP-induced liver injury. Full article
(This article belongs to the Special Issue Functional Evaluation of Edible Mushrooms and Their Active Materials)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Flowchart of ultrasonic-assisted hot water extraction of GLPs; (<b>B</b>) polysaccharides’ molecular weight (The red line represents the multi angle laser light scattering signal, the blue line represents the differential signal, and the black line is the molecular weight fitted by the two signals); (<b>C</b>) standard; (<b>D</b>) monosaccharide composition.</p>
Full article ">Figure 1 Cont.
<p>(<b>A</b>) Flowchart of ultrasonic-assisted hot water extraction of GLPs; (<b>B</b>) polysaccharides’ molecular weight (The red line represents the multi angle laser light scattering signal, the blue line represents the differential signal, and the black line is the molecular weight fitted by the two signals); (<b>C</b>) standard; (<b>D</b>) monosaccharide composition.</p>
Full article ">Figure 2
<p>Effect of GLPs on APAP-induced liver injury in mice: (<b>A</b>) experimental design; (<b>B</b>) liver index; (<b>C</b>) ALT activity; (<b>D</b>) AST activity <span class="html-italic">(n</span> = 6). The results are expressed as mean ± standard error; compared with the normal group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Normal control: blank control group, model: model group, positive control: positive group, GLP-L: <span class="html-italic">Ganoderma lucidum</span> polysaccharide low-dose group, GLP-M: <span class="html-italic">Ganoderma lucidum</span> polysaccharide medium-dose group, GLP-H: <span class="html-italic">Ganoderma lucidum</span> polysaccharide high-dose group.</p>
Full article ">Figure 3
<p>(<b>A</b>) HE staining image; (<b>B</b>,<b>C</b>) TUNEL staining was utilized to observe the apoptosis of liver tissue cells in each group <span class="html-italic">(n</span> = 6). The magnification in H&amp;E was ×20, and scale bars = 100 μm; compared with the normal control group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. The green arrow represents the liver lobules, the black arrow represents the nucleus, the red arrow represents inflammatory cells, and the blue arrow represents cell apoptosis.</p>
Full article ">Figure 3 Cont.
<p>(<b>A</b>) HE staining image; (<b>B</b>,<b>C</b>) TUNEL staining was utilized to observe the apoptosis of liver tissue cells in each group <span class="html-italic">(n</span> = 6). The magnification in H&amp;E was ×20, and scale bars = 100 μm; compared with the normal control group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. The green arrow represents the liver lobules, the black arrow represents the nucleus, the red arrow represents inflammatory cells, and the blue arrow represents cell apoptosis.</p>
Full article ">Figure 3 Cont.
<p>(<b>A</b>) HE staining image; (<b>B</b>,<b>C</b>) TUNEL staining was utilized to observe the apoptosis of liver tissue cells in each group <span class="html-italic">(n</span> = 6). The magnification in H&amp;E was ×20, and scale bars = 100 μm; compared with the normal control group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. The green arrow represents the liver lobules, the black arrow represents the nucleus, the red arrow represents inflammatory cells, and the blue arrow represents cell apoptosis.</p>
Full article ">Figure 4
<p>GLPs improve liver antioxidant status: (<b>A</b>) SOD activity; (<b>B</b>) GSH-Px activity; (<b>C</b>) GSH level; (<b>D</b>) MDA level; (<b>E</b>) ROS level (<span class="html-italic">n</span> = 6). The results are expressed as mean ± standard error; compared with the normal control group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Normal control: blank control group, model: model group, positive control: positive group, GLP-L: <span class="html-italic">Ganoderma lucidum</span> polysaccharide low-dose group, GLP-M: <span class="html-italic">Ganoderma lucidum</span> polysaccharide medium-dose group, GLP-H: <span class="html-italic">Ganoderma lucidum</span> polysaccharide high-dose group).</p>
Full article ">Figure 4 Cont.
<p>GLPs improve liver antioxidant status: (<b>A</b>) SOD activity; (<b>B</b>) GSH-Px activity; (<b>C</b>) GSH level; (<b>D</b>) MDA level; (<b>E</b>) ROS level (<span class="html-italic">n</span> = 6). The results are expressed as mean ± standard error; compared with the normal control group, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Normal control: blank control group, model: model group, positive control: positive group, GLP-L: <span class="html-italic">Ganoderma lucidum</span> polysaccharide low-dose group, GLP-M: <span class="html-italic">Ganoderma lucidum</span> polysaccharide medium-dose group, GLP-H: <span class="html-italic">Ganoderma lucidum</span> polysaccharide high-dose group).</p>
Full article ">Figure 5
<p>Effect of GLP treatment on the expression of Nrf2 pathway-related genes and proteins: (<b>A</b>) Nrf2, HO-1, GCLC, and NQO1 mRNA expression levels in liver tissue; (<b>B</b>) quantitative analysis of protein expression (<span class="html-italic">n</span> = 6); (<b>C</b>) protein imprinting analysis; compared with the normal group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Normal control: blank control group, model: model group, positive control: positive group, GLP-L: <span class="html-italic">Ganoderma lucidum</span> polysaccharide low-dose group, GLP-M: <span class="html-italic">Ganoderma lucidum</span> polysaccharide medium-dose group, GLP-H: <span class="html-italic">Ganoderma lucidum</span> polysaccharide high-dose group.</p>
Full article ">Figure 5 Cont.
<p>Effect of GLP treatment on the expression of Nrf2 pathway-related genes and proteins: (<b>A</b>) Nrf2, HO-1, GCLC, and NQO1 mRNA expression levels in liver tissue; (<b>B</b>) quantitative analysis of protein expression (<span class="html-italic">n</span> = 6); (<b>C</b>) protein imprinting analysis; compared with the normal group, ** <span class="html-italic">p</span> &lt; 0.01; compared with the model, # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01. Normal control: blank control group, model: model group, positive control: positive group, GLP-L: <span class="html-italic">Ganoderma lucidum</span> polysaccharide low-dose group, GLP-M: <span class="html-italic">Ganoderma lucidum</span> polysaccharide medium-dose group, GLP-H: <span class="html-italic">Ganoderma lucidum</span> polysaccharide high-dose group.</p>
Full article ">
15 pages, 3116 KiB  
Article
Evaluation of Physicochemical Properties and Prebiotics Function of a Bioactive Pleurotus eryngii Aqueous Extract Powder Obtained by Spray Drying
by Jianqiu Chen, Mengling Zhou, Liding Chen, Chengfeng Yang, Yating Deng, Jiahuan Li and Shujing Sun
Nutrients 2024, 16(11), 1555; https://doi.org/10.3390/nu16111555 - 21 May 2024
Viewed by 701
Abstract
A bioactive Pleurotus eryngii aqueous extract powder (SPAE) was obtained by spray drying and its performance in terms of physicochemical properties, in vitro digestion, inflammatory factors, and modulation of the intestinal microbiota was explored. The results indicated that the SPAE exhibited a more [...] Read more.
A bioactive Pleurotus eryngii aqueous extract powder (SPAE) was obtained by spray drying and its performance in terms of physicochemical properties, in vitro digestion, inflammatory factors, and modulation of the intestinal microbiota was explored. The results indicated that the SPAE exhibited a more uniform particle size distribution than P. eryngii polysaccharide (PEP). Meanwhile, a typical absorption peak observed at 843 cm−1 in the SPAE FTIR spectra indicated the existence of α-glycosidic bonds. SPAE exhibited higher antioxidant abilities and superior resistance to digestion in vitro. In addition, SPAE supplementation to mice significantly reduced the release of factors that promote inflammation, enhanced the secretion of anti-inflammatory factors, and sustained maximum production of short-chain fatty acids (SCFAs). Additionally, it significantly enhanced the relative abundance of SCFAs-producing Akkermansia and reduced the abundance of Ruminococcus and Clostridiides in intestines of mice. These results show the potential of SPAE as a novel material with prebiotic effects for the food and pharmaceutical industries. Full article
(This article belongs to the Special Issue Functional Evaluation of Edible Mushrooms and Their Active Materials)
Show Figures

Figure 1

Figure 1
<p>Characterization of the SPAE and PEP. (<b>A</b>) Scanning electron micrographs (<b>a1</b>–<b>a4</b>: SPAE, <b>b1</b>–<b>b4</b>: PEP). (<b>B</b>) FTIR spectra. (<b>C</b>) TGA–DTG curves. (<b>D</b>) DSC curves.</p>
Full article ">Figure 2
<p>ABTS (<b>A</b>) and DPPH (<b>B</b>) free radical scavenging effects of SPAE and PEP at different pH. The use of lowercase letters indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Effects of SPAE and PEP on inflammatory parameters in serum of mice. The use of lowercase letters indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>Concentrations of SCFAs in intestinal tract of mice with SPAE and PEP. (<b>A</b>) pH value. (<b>B</b>) and (<b>C</b>) intestinal SCFAs. The use of lowercase letters indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 5
<p>Effects of SPAE and PEP on gut microbiota composition analysis. (<b>A</b>) α diversity analyzed by Shannon index. (<b>B</b>) β diversity analyzed by PCoA. (<b>C</b>) OTU level of gut microbiota. (<b>D</b>) Phylum level of gut microbiota. (<b>E</b>) The ratio of <span class="html-italic">Bacteroidetes</span> and <span class="html-italic">Firmicutes</span>. (<b>F</b>–<b>H</b>) Genus level of gut microbiota. The use of “**” indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.01. The use of lowercase letters indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Relationship between inflammatory parameters, SCFAs, and gut microbiota. (<b>A</b>) Correlation analysis of gut microbiota at genus level. (<b>B</b>) Heatmap of the correlation coefficients between inflammatory parameters and gut microbiota at genus level. (<b>C</b>) Heatmap of correlation coefficients between SCFAs and gut microbiota at genus levels. The use of “*” indicates significant differences among the treatment groups at a significance level of <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">
15 pages, 6560 KiB  
Article
Intestinal Microbiota and Metabolomics Reveal the Role of Auricularia delicate in Regulating Colitis-Associated Colorectal Cancer
by Lanzhou Li, Honghan Liu, Jinqi Yu, Zhen Sun, Ming Jiang, Han Yu and Chunyue Wang
Nutrients 2023, 15(23), 5011; https://doi.org/10.3390/nu15235011 - 4 Dec 2023
Cited by 1 | Viewed by 1694
Abstract
Background: The edible fungus Auricularia delicate (ADe) is commonly employed in traditional medicine for intestinal disorders; however, its inhibitory effect on colitis-associated colorectal cancer (CAC) and the underlying mechanisms remain unexplored. (2) Methods: The inhibitory effect of ADe on CAC was investigated using [...] Read more.
Background: The edible fungus Auricularia delicate (ADe) is commonly employed in traditional medicine for intestinal disorders; however, its inhibitory effect on colitis-associated colorectal cancer (CAC) and the underlying mechanisms remain unexplored. (2) Methods: The inhibitory effect of ADe on CAC was investigated using a mouse model induced by azoxymethane/dextran sulfate sodium. Results: ADe effectively suppressed the growth and number of intestinal tumors in mice. Intestinal microbiota analyses revealed that ADe treatment increased Akkermansia and Parabacteroides while it decreased Clostridium, Turicibacter, Oscillospira, and Desulfovibrio. ADe regulated the levels of 2′-deoxyridine, creatinine, 1-palmitoyl lysophosphatidylcholine, and choline in serum. Furthermore, the levels of these metabolites were associated with the abundance of Oscillospira and Paraacteroides. ADe up-regulated the free fatty acid receptor 2 and β-Arrestin 2, inhibited the nuclear factor kappa B (NF-κB) pathway, and significantly attenuated the levels of inflammatory cytokines, thereby mitigating the inflammatory in CAC mice. Conclusions: The protective effect of ADe in CAC mice is associated with the regulation of intestinal microbiota, which leads to the inhibition of NF-kB pathway and regulation of inflammation. Full article
(This article belongs to the Special Issue Functional Evaluation of Edible Mushrooms and Their Active Materials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The protective effect of ADe in CAC mice. (<b>A</b>) A simplified flowchart depicting the animal experimental protocol. (<b>B</b>) Body weights of mice (<span class="html-italic">n</span> = 10). (<b>C</b>) Colorectum index measurements (<span class="html-italic">n</span> = 10). (<b>D</b>) Representative colorectal tissues of each group. (<b>E</b>) H&amp;E pathological sections showing colorectal tumors at different magnifications (40× scale bar: 500 μm; 400× scale bar: 50 μm) (<span class="html-italic">n</span> = 3). <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 vs. control group; red arrows: tumor tissue.</p>
Full article ">Figure 2
<p>The intestinal microbiota of CAC mice (<span class="html-italic">n</span> = 4) is regulated by ADe treatment. (<b>A</b>) A Venn diagram illustrated the overlap between different microbial taxa. (<b>B</b>) PCoA of unweighted UniFrac distance was executed to assess beta diversity. (<b>C</b>) Grouping box plots were generated to compare alpha diversity indices. (<b>D</b>) A heatmap was constructed to display the composition of the top 20 dominant genera. (<b>E</b>) LEfSe analysis. The logarithmic score threshold for LDA analysis was set at 2.0.</p>
Full article ">Figure 3
<p>The levels of serum metabolites in CAC mice were regulated by ADe treatment. (<b>A</b>) Venn diagram illustrated the overlap of altered metabolites. (<b>B</b>) Heatmap displaying 36 significantly altered metabolites. (<b>C</b>) Heatmap showing the associated alterations in metabolite levels. (<b>D</b>) Heatmap presenting the associations between significantly altered metabolites and microbiota species. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The regulatory effect of ADe on colorectal cytokines in colorectal tumor of CAC mice. Specifically, (<b>A</b>) IL-1α, (<b>B</b>) IL-1β, (<b>C</b>) IL-6, (<b>D</b>) IL-12, (<b>E</b>) IL-17A, (<b>F</b>) IL-22, (<b>G</b>) IL-27, (<b>H</b>) GM-CSF, (<b>I</b>) IFN-γ, (<b>J</b>) IFN-β, (<b>K</b>) MCP-1, and (<b>L</b>) TNF-α. (<span class="html-italic">n</span> = 6). <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. control group; * <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 vs. model group.</p>
Full article ">Figure 5
<p>The proteins associated with the NF-κB pathway and inflammation in colorectal tumors of CAC mice. Quantification data were standardized by employing GAPDH as a reference, and fold change values were reported relative to control mice (<span class="html-italic">n</span> = 3). <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 vs. control group; * <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 vs. model group.</p>
Full article ">
16 pages, 5683 KiB  
Article
Chroogomphus rutilus Regulates Bone Metabolism to Prevent Periodontal Bone Loss during Orthodontic Tooth Movement in Osteoporotic Rats
by Ying Zhou, Yanfeng Zhu, Xinghui Jin, Yongfeng Zhang, Jiyu Song, Zhina Wu, Yutong Li, Jingzheng Yi, Di Wang and Min Hu
Nutrients 2023, 15(23), 4906; https://doi.org/10.3390/nu15234906 - 24 Nov 2023
Viewed by 1341
Abstract
Osteoporosis (OP) leads to the acceleration of tooth movement and aggravation of periodontal bone loss during orthodontic treatment. Chroogomphus rutilus (CR) is abundant in nutrients and demonstrates remarkable antioxidant and anti-inflammatory properties. In the present study, the components of CR, including 35.00% total [...] Read more.
Osteoporosis (OP) leads to the acceleration of tooth movement and aggravation of periodontal bone loss during orthodontic treatment. Chroogomphus rutilus (CR) is abundant in nutrients and demonstrates remarkable antioxidant and anti-inflammatory properties. In the present study, the components of CR, including 35.00% total sugar, 0.69% reducing sugar, 14.40% crude protein, 7.30% total ash, 6.10% crude fat, 0.51% total flavonoids, 1.94% total triterpenoids, 0.32% total sterol, 1.30% total saponins, 1.69% total alkaloids, and 1.02% total phenol, were first systematically examined, followed by an investigation into its regulatory effects on bone metabolism in order to mitigate bone loss during orthodontic tooth movement in osteoporotic rats. The results of the imaging tests revealed that CR treatment reduced periodontal bone loss and normalized tooth movement in the OP. In conjunction with analyses of intestinal flora and metabolomics, CR enhances the prevalence of anti-inflammatory genera while reducing the production of inflammatory metabolites. Meanwhile, CR reduced the levels of periodontal inflammatory factors, including TNF-α, IL-1β, and IL-6, by activating Wnt/β-catenin signaling, and promoted periodontal bone formation. These findings imply that CR is a potent supplementary therapy for controlling periodontal bone remodeling in patients with OP undergoing orthodontic treatment. Full article
(This article belongs to the Special Issue Functional Evaluation of Edible Mushrooms and Their Active Materials)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>CR promoted bone remodeling in osteoporotic alveolar bone during OTM. (<b>A</b>) X-rays of femur and tibia in experimental rats (<span class="html-italic">n</span> = 5/groups). (<b>B</b>) Intraoral schematic of the OTM model. (<b>C</b>) CT scan of the left maxilla (<span class="html-italic">n</span> = 3/groups): 3D reconstructions of maxillary samples are shown on the left. The red dotted lines indicate the cementoenamel junction (CEJ; upper) and the alveolar crest (lower). Red solid line indicates the mesial, middle, and distal alveolar bone loss of the first molar. The white arrow points to the direction of tooth movement. The right side is the sagittal view of the alveolar bone; the a and b points represent the OTM distance, and the red box represents ROI of the alveolar bone. Quantitative analyses of (<b>D</b>) BMD, (<b>E</b>) BV/TV, (<b>F</b>) Tb.Th, (<b>G</b>) Tb.Sp, (<b>H</b>) OTM distance, and (<b>I</b>) middle bone loss among groups (<span class="html-italic">n</span> = 3/group). (<b>J</b>) H&amp;E staining of alveolar bone (<span class="html-italic">n</span> = 3/group, 200×, scale bar: 100 µm). M1: first molar; PDL: periodontal ligament; AB: alveolar bone. Data are expressed as mean ± SEM, and were analyzed via one-way ANOVA. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05, <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01, <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 <span class="html-italic">versus</span> CTRL rats; * <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">versus</span> vehicle-treated OP rats. (CR: <span class="html-italic">Chroogomphus rutilus</span>; OTM: orthodontic tooth movement; CT: computed tomography; ROI: the region of interest; BMD: bone mineral density; BV/TV: bone volume/total volume; Tb.Th: trabecular thickness; Tb.Sp: trabecular separation/spacing; H&amp;E: hematoxylin and eosin).</p>
Full article ">Figure 2
<p>CR preserved the balance of intestinal flora in osteoporosis (OP) rats (<span class="html-italic">n</span> = 3/group). (<b>A</b>) Venn diagram. The top 10 phylum (<b>B</b>) and top 20 genera (<b>C</b>) in terms of intestinal abundance. (<b>D</b>) α-diversity analysis. (<b>E</b>) PCoA plots represented as β-diversity relying on the Jaccard distance algorithm. (<b>F</b>) Heatmap of the top 20 genera in abundance. Group 1: CTRL rats; Group 2: vehicle-treated OP rats; Group 3: CR-treated OP rats. <sup>#</sup> <span class="html-italic">p</span> &lt; 0.05 <span class="html-italic">versus</span> CTRL rats. (CR: <span class="html-italic">Chroogomphus rutilus</span>).</p>
Full article ">Figure 3
<p>CR modified the metabolite levels in serum of osteoporosis (OP) rats (<span class="html-italic">n</span> = 3/group). (<b>A</b>) PCA score plots among three groups. (<b>B</b>) OPLS-DA plots between Group 2 and Group 3. (<b>C</b>) KEGG pathway analysis predicts alterations in metabolic pathways between vehicle-treated OP rats and CR-treated OP rats. (<b>D</b>) Heatmap of significantly changed serum metabolites. (<b>E</b>) Joint analysis of serum metabolites and gut microbiota. Group 1: CTRL rats; Group 2: vehicle-treated OP rats; Group 3: CR-treated OP rats. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01 between serum metabolites and intestinal flora. (CR: <span class="html-italic">Chroogomphus rutilus</span>; PCA: principal component analysis; OPLS-DA: orthogonal partial least squares discriminant analysis; KEGG: Kyoto Encyclopedia of Genes and Genomes).</p>
Full article ">Figure 4
<p>CR promoted the expression of periodontal osteogenic differentiation marker factors during OTM in OP rats. IHC staining of (<b>A</b>) Runx2, (<b>B</b>) osterix, (<b>C</b>) OPG, and (<b>D</b>) RANKL in periodontal tissue on the tension side of OTM (<span class="html-italic">n</span> = 3/group, 200×, scale bar: 100 µm). Quantification of MOD of (<b>E</b>) Runx2, (<b>F</b>) Osterix, (<b>G</b>) OPG, and (<b>H</b>) RANKL in the tension area (<span class="html-italic">n</span> = 3/group). Data are expressed as the mean ± SEM and were analyzed via one-way ANOVA. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 <span class="html-italic">versus</span> CTRL rats; ** <span class="html-italic">p</span> &lt; 0.01 <span class="html-italic">versus</span> vehicle-treated OP rats. (CR: <span class="html-italic">Chroogomphus rutilus</span>; IHC: immunohistochemical; Runx2: runt-related transcription factor 2; OPG: osteoprotegerin; RANKL: receptor activator of NF-κB ligand; MOD: mean optical density).</p>
Full article ">Figure 5
<p>CR regulated Wnt/β-catenin signaling and reduced periodontal inflammation during OTM in OP rats. IHC staining of (<b>A</b>) Wnt1, (<b>B</b>) GSK-3β, (<b>C</b>) β-catenin, (<b>D</b>) TNF-α, (<b>E</b>) IL-1β, and (<b>F</b>) IL-6 in periodontal tissue on the tension side of OTM (n = 3/group, 200×, scale bar: 100 µm). Quantification of MOD of (<b>G</b>) Wnt1, (<b>H</b>) GSK-3β, (<b>I</b>) β-catenin, (<b>J</b>) TNF-α, (<b>K</b>) IL-1β, and (<b>L</b>) IL-6 in the tension area (<span class="html-italic">n</span> = 3/group). Data are expressed as the mean ± SEM and were analyzed via one-way ANOVA. <sup>##</sup> <span class="html-italic">p</span> &lt; 0.01 and <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 <span class="html-italic">versus</span> CTRL rats; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 <span class="html-italic">versus</span> vehicle-treated OP rats. (CR: <span class="html-italic">Chroogomphus rutilus</span>; OTM: orthodontic tooth movement; IHC: immunohistochemical; GSK-3β: glycogen synthase kinase-3β; TNF-α: tumor necrosis factor-alpha; IL: interleukin; MOD: mean optical density).</p>
Full article ">
17 pages, 5806 KiB  
Article
Pleurotus abieticola Polysaccharide Alleviates Hyperlipidemia Symptoms via Inhibition of Nuclear Factor-κB/Signal Transducer and Activator of Transcription 3-Mediated Inflammatory Responses
by Yongfeng Zhang, Yingjie Lin, Keyi Wu, Ming Jiang, Lanzhou Li and Yang Liu
Nutrients 2023, 15(23), 4904; https://doi.org/10.3390/nu15234904 - 23 Nov 2023
Cited by 1 | Viewed by 1277
Abstract
Hyperlipidemia (HLP) is a metabolic syndrome induced by obesity, which has been widely recognized as a significant threat to human health. Pleurotus abieticola, an edible lignin-degrading fungus, remains relatively understudied in terms of its bioactivity and medicinal properties. In this study, the [...] Read more.
Hyperlipidemia (HLP) is a metabolic syndrome induced by obesity, which has been widely recognized as a significant threat to human health. Pleurotus abieticola, an edible lignin-degrading fungus, remains relatively understudied in terms of its bioactivity and medicinal properties. In this study, the lipid-lowering effect of Pleurotus abieticola polysaccharide (PAPS1) was systematically explored in high-fat diet (HFD)-induced HLP mice. The findings demonstrated that the administration of PAPS1 significantly inhibited bodyweight gain, ameliorated blood glucose and lipid levels, reduced fat accumulation, and mitigated hepatic injury in HLP mice. In addition, PAPS1 demonstrated the capability to increase the levels of three distinct fecal metabolites while simultaneously reducing the levels of eight other fecal metabolites in HLP mice. According to biological detection, PAPS1 reduced the hepatic level of reactive oxygen species (ROS) and pro-inflammatory factors, such as tumor necrosis factor (TNF)-α and interleukin (IL)-1β, -6, -17A, -22, and -23, and increased the expression of anti-inflammatory factor IL-10. Combined with proteomics, Western blot and immunohistochemistry analysis showed that PAPS1 exerted suppressive effects on inflammation and oxidative damage by inhibiting the nuclear factor-κB (NF-κB)/signal transducer and activator of transcription 3 (STAT3) signaling pathway in HLP mice. These findings offer evidence supporting the effectiveness of PAPS1 as a therapeutic agent in reducing lipid levels through its targeting of chronic inflammation. Full article
(This article belongs to the Special Issue Functional Evaluation of Edible Mushrooms and Their Active Materials)
Show Figures

Figure 1

Figure 1
<p>PAPS1 ameliorated hyperlipidemia symptoms in HFD-induced HLP mice. (<b>A</b>) The HLP model establishment and administration process. PAPS1 treatment suppressed the body weight (<b>B</b>) and plasma glucose (<b>C</b>) gain; decreased the serum levels of TG (<b>D</b>), TC (<b>E</b>), and LDL-C (<b>F</b>); and increased the serum level of HDL-C (<b>G</b>) in HFD-induced HLP mice. Data are expressed as the mean ± S.E.M. (n = 6). # <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 vs. NCD group; * <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 vs. HFD group; ^ <span class="html-italic">p</span> &lt; 0.05 and ^^ <span class="html-italic">p</span> &lt; 0.01 vs. SV group. NCD: normal chow diet; HFD: high-fat diet; SV: simvastatin; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide; TG: triglyceride; TC: total cholesterol; LDL-C: low-density lipoprotein cholesterol; HDL-C: high-density lipoprotein cholesterol.</p>
Full article ">Figure 2
<p>PAPS1 inhibited fat accumulation in HFD-induced HLP mice. (<b>A</b>) Histopathological analysis of eWAT, iWAT, pWAT, and BAT was performed using H&amp;E staining (200×; scale bar: 100 μm) (n = 3). PAPS1 treatment had no significant effect on eWAT index (<b>B</b>) but reduced the indexes of iWAT (<b>C</b>) and pWAT (<b>D</b>) and enhanced the BAT index (<b>E</b>) in HFD-induced HLP mice. Data are expressed as the mean ± S.E.M. (n = 6). # <span class="html-italic">p</span> &lt; 0.05 and ### <span class="html-italic">p</span> &lt; 0.001 vs. NCD group; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. HFD group; ^^ <span class="html-italic">p</span> &lt; 0.01 vs. SV group. NCD: normal chow diet; HFD: high-fat diet; SV: simvastatin; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide; eWAT: epididymal white adipose tissue; iWAT: inguinal white adipose tissue; pWAT: perirenal white adipose tissue; BAT: brown adipose tissue.</p>
Full article ">Figure 3
<p>PAPS1 improved liver injury in HFD-induced HLP mice. Histopathological analysis of liver was performed using H&amp;E staining (<b>A</b>) and oil red O staining (<b>B</b>) (200×; scale bar: 100 μm) (n = 3). PAPS1 treatment reduced the levels of AST (<b>C</b>,<b>D</b>) and ALT (<b>E</b>,<b>F</b>) in serum and liver of HLP mice caused by HFD intake. Data are expressed as the mean ± S.E.M. (n = 6). ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. NCD group; ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001 vs. HFD group. NCD: normal chow diet; HFD: high-fat diet; SV: simvastatin; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide; H&amp;E: hematoxylin and eosin; ALT: alanine aminotransferase; AST: aspartate transaminase.</p>
Full article ">Figure 4
<p>PAPS1 regulated the fecal metabolite level in HFD-induced HLP mice. (<b>A</b>) OPLS-DA score plot of fecal metabolites among NCD, HFD, and PAPS1 groups. (<b>B</b>) KEGG-enriched metabolic pathways of significantly altered fecal metabolites between HFD and PAPS1 groups. Heatmap (<b>C</b>), the associated heatmap (<b>D</b>), and boxplots (<b>E</b>–<b>O</b>) of significantly differential metabolites in intestinal feces among three groups. Data are expressed as the mean ± S.E.M. (n = 4). # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, and ### <span class="html-italic">p</span> &lt; 0.001 vs. NCD group; * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001 vs. HFD group. KEGG: Kyoto Encyclopedia of Genes and Genomes; VEGF: vascular endothelial growth factor; NCD: normal chow diet; HFD: high-fat diet; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide.</p>
Full article ">Figure 5
<p>Proteomics of analysis of the liver in HFD-induced HLP mice. Heatmap (<b>A</b>), GO (<b>B</b>), and KEGG (<b>C</b>) enrichment analysis of significantly altered proteins among NCD, HFD, and PAPS1 groups. (<b>D</b>) The WB results showed PAPS1 treatment increased hepatic levels of Creg1, Gls2, Gsta4, and Gpx4 and decreased the hepatic level of 4-HNE in HFD-induced HLP mice. (<b>E</b>) PAPS1 treatment reduced the hepatic level of ROS in HFD-induced HLP mice. Data are expressed as the mean ± S.E.M. (n = 6). ### <span class="html-italic">p</span> &lt; 0.001 vs. NCD group; ** <span class="html-italic">p</span> &lt; 0.01 vs. HFD group. The IHC analysis showed PAPS1 treatment enhanced the hepatic expression of Creg1 (<b>F</b>) and reduced the expression of 4-HNE (<b>G</b>) in HFD-induced HLP mice. NCD: normal chow diet; HFD: high-fat diet; SV: simvastatin; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide; Creg1: cellular repressor of E1A-stimulated genes 1; Gls2: glutaminase 2; Gsta4: glutathione S-transferase alpha 4; Gpx4: glutathione peroxidase 4; 4-HNE: 4-hydroxynonenal.</p>
Full article ">Figure 6
<p>PAPS1 treatment inhibited NF-κB/STAT3-mediated inflammatory response in HFD-induced HLP mice. (<b>A</b>) PAPS1 treatment suppressed NF-κB/STAT3 signaling pathway and its related downstream proteins of liver in HFD-induced HLP mice. PAPS1 treatment down-regulated the hepatic levels of TNF-α (<b>B</b>), IL-1β (<b>C</b>), IL-6 (<b>D</b>), IL-17A (<b>F</b>), IL-22 (<b>G</b>), and IL-23 (<b>H</b>), and up-regulated the hepatic level of IL-10 (<b>E</b>) in HFD-induced HLP mice. Data are expressed as the mean ± S.E.M. (n = 6). ## <span class="html-italic">p</span> &lt; 0.01 and ### <span class="html-italic">p</span> &lt; 0.001 vs. NCD group; * <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 vs. HFD group; ^^ <span class="html-italic">p</span> &lt; 0.01 vs. SV group. (<b>I</b>) PAPS1 treatment suppressed the hepatic level of P-NF-κB in HFD-induced HLP mice, as analyzed using IHC. NF-κB: nuclear factor-κB; P-NF-κB: phosphorylated NF-κB; STAT3: signal transducer and activator of transcription 3; P-STAT3: phosphorylated STAT3; Saa1: serum amyloid A protein; Hp: haptoglobin; IFN-γ: interferon-γ; TNF-α: tumor necrosis factor; IL-1β: interleukin-1β; HFD: high-fat diet; NCD: normal chow diet; PAPS1: <span class="html-italic">Pleurotus abieticola</span> polysaccharide; SV: simvastatin.</p>
Full article ">
Back to TopTop