[go: up one dir, main page]

 
 

Topic Editors

Dr. Pujie Shi
Center for Engineered Therapeutics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
Dr. Tiantian Lin
Department of Food Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
Food, Nutrition and Health, Faculty of Land and Food Systems, The University of British Columbia, Vancouver, BC, Canada
Department of Food Science and Technology, National University of Singapore, 2 Science Drive 2, Singapore 117542, Singapore
Food Science and Technology Center, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
Department of Food Science and Technology, Korea National University of Transportation, Jeungpyeong-gun, Chungbuk, Republic of Korea
Institute of Bioprocess Engineering and Pharmaceutical Technology, University of Applied Sciences Mittelhessen, Giessen, Germany

Bioactive Compounds with Application Potentials in Nutraceuticals and Nutricosmetics: Focus on Mechanism of Action and Application Science

Abstract submission deadline
30 September 2024
Manuscript submission deadline
31 December 2024
Viewed by
59493

Topic Information

Dear Colleagues,

As biological compounds isolated from natural plants, nutraceuticals offer numerous benefits, such as preventing chronic disease, delaying aging, extending lifespan, improving health, and supporting body structure and function. Nutricosmetics can currently be used as nutritional supplements to provide adequate nutrients for nails, hair, and skin, and work from within the body to add beauty, while future trends in the health and beauty industry will revolve around nutraceuticals and nutricosmetics. Bioactive compounds are key factors for the development of nutraceuticals or nutricosmetics, as well as some functional foods. This topic invites recent studies covering, but not limited to, the following subtopics:

(1) Bioactivities of compounds from edible natural products, including ethnopharmacology and folk medicine with long histories of ingestion;
(2) Functional mechanism studies of nutraceuticals and nutricosmetics via biochemical analysis, in vitro/vivo/silico or evidence-based clinical trials;
(3) Process for biosynthesis and formulation of nutraceuticals, nutricosmetics, or biocompatible cosmetic materials;
(4) Advanced technologies for improving the stability and functionality of bioactive products for application.

Dr. Pujie Shi
Dr. Tiantian Lin
Dr. Lin Chen
Dr. Xin Yang
Prof. Dr. Caili Fu
Prof. Dr. Hyun-Gyun Yuk
Dr. Rong Fan
Topic Editors

Keywords

  • nutraceuticals
  • nutricosmetic
  • biocompatible
  • stability
  • extending lifespan
  • chronic disease
  • skin health
  • antioxidant

Participating Journals

Journal Name Impact Factor CiteScore Launched Year First Decision (median) APC
Antioxidants
antioxidants
6.0 10.6 2012 15.5 Days CHF 2900 Submit
BioChem
biochem
- - 2021 24.1 Days CHF 1000 Submit
Biomolecules
biomolecules
4.8 9.4 2011 16.3 Days CHF 2700 Submit
Cells
cells
5.1 9.9 2012 17.5 Days CHF 2700 Submit
International Journal of Molecular Sciences
ijms
4.9 8.1 2000 18.1 Days CHF 2900 Submit
Nutrients
nutrients
4.8 9.2 2009 17.5 Days CHF 2900 Submit
Pharmaceutics
pharmaceutics
4.9 7.9 2009 14.9 Days CHF 2900 Submit
Foods
foods
4.7 7.4 2012 14.3 Days CHF 2900 Submit

Preprints.org is a multidiscipline platform providing preprint service that is dedicated to sharing your research from the start and empowering your research journey.

MDPI Topics is cooperating with Preprints.org and has built a direct connection between MDPI journals and Preprints.org. Authors are encouraged to enjoy the benefits by posting a preprint at Preprints.org prior to publication:

  1. Immediately share your ideas ahead of publication and establish your research priority;
  2. Protect your idea from being stolen with this time-stamped preprint article;
  3. Enhance the exposure and impact of your research;
  4. Receive feedback from your peers in advance;
  5. Have it indexed in Web of Science (Preprint Citation Index), Google Scholar, Crossref, SHARE, PrePubMed, Scilit and Europe PMC.

Published Papers (26 papers)

Order results
Result details
Journals
Select all
Export citation of selected articles as:
22 pages, 1503 KiB  
Article
Effect of Chickpea (Cicer arietinum L.) Flour Incorporation on Quality, Antioxidant Properties, and Bioactive Compounds of Shortbread Cookies
by Katarzyna Felisiak, Sylwia Przybylska, Grzegorz Tokarczyk, Małgorzata Tabaszewska, Jacek Słupski and Joanna Wydurska
Foods 2024, 13(15), 2356; https://doi.org/10.3390/foods13152356 - 26 Jul 2024
Viewed by 596
Abstract
High nutritional value and antioxidant properties make chickpea flour a valuable substitute for wheat flour, although its texture-forming abilities are different. The aim of this study was to investigate the possibility of increasing the content of bioactive compounds and antioxidant properties of shortbread [...] Read more.
High nutritional value and antioxidant properties make chickpea flour a valuable substitute for wheat flour, although its texture-forming abilities are different. The aim of this study was to investigate the possibility of increasing the content of bioactive compounds and antioxidant properties of shortbread cookies by simple partial or complete replacement of wheat flour with chickpea flour without considerable changes in texture, color, sensory properties, or acceptability. Shortbread cookies were made from wheat flour (0% of chickpea flour), wheat flour and chickpea flour (replacement of 25%, 50%, and 75%), and chickpea flour (100%). Generally, the increase in chickpea flour share resulted in an increase in protein, fat, and ash content, as well as antioxidant properties. Polyphenol content, flavonoid content, and antioxidant activities increased three- to sixfold in shortbread cookies containing chickpea flour in comparison to wheat cookies. The level of proteins increased about 50% and the antioxidant properties were three to six times higher than in wheat cookies. Cookies containing up to 75% chickpea flour were assessed as very good or good quality, while only cookies without wheat flour were assessed as sufficient quality. It could be concluded that part of the wheat flour content in shortbread cookies can be replaced by chickpea flour. Application of a 25% proportion of chickpea flour increases physicochemical properties without changes in sensory properties. Sensory quality was up to 75% lower, but antioxidant properties were increased. However, complete replacement of wheat flour in shortbread cookies without changing the recipe resulted in a product of slightly lower sensory quality. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of chickpea flour on the appearance and color of shortbread cookies (share of chickpea flour: S0—0%, S25—25%, S50—50%, S75—75%, S100—100%). Photo J. Wydurska.</p>
Full article ">Figure 2
<p>Taste sensory profile of shortbread cookies without and with chickpea flour.</p>
Full article ">Figure 3
<p>Odor sensory profile of shortbread cookies without and with chickpea flour.</p>
Full article ">Figure 4
<p>PCA biplot of the first two principal components for antioxidant activity, sensory assessment, hardness, color parameters, and shortbread cookie components and component distribution. TPC—total phenolic compound; TFC—total flavonoid compound; TC—total carotenoids; TEAC—Trolox equivalent antioxidant capacity; FRAP—ferric reducing antioxidant power; RSA—radical scavenging ability; FCA—ferrous chelating ability; Sens. a.—sensory assessment; <span class="html-italic">L</span>*—lightness, BI—browning index. * additional variables.</p>
Full article ">
14 pages, 3215 KiB  
Article
The Use of Polysaccharide AOP30 from the Rhizome of Alpinia officinarum Hance to Alleviate Lipopolysaccharide-Induced Intestinal Epithelial Barrier Dysfunction and Inflammation via the TLR4/NfκB Signaling Pathway in Caco-2 Cell Monolayers
by Xuejing Jia, Yun Huang, Guanghuo Liu, Zipeng Li, Qiwei Tan and Saiyi Zhong
Nutrients 2024, 16(13), 2151; https://doi.org/10.3390/nu16132151 - 5 Jul 2024
Viewed by 691
Abstract
Alpinia officinarum Hance is rich in carbohydrates and is flavored by natives. The polysaccharide fraction 30 is purified from the rhizome of A. officinarum Hance (AOP30) and shows excellent immunoregulatory ability when administered to regulate immunity. However, the effect of AOP30 on the [...] Read more.
Alpinia officinarum Hance is rich in carbohydrates and is flavored by natives. The polysaccharide fraction 30 is purified from the rhizome of A. officinarum Hance (AOP30) and shows excellent immunoregulatory ability when administered to regulate immunity. However, the effect of AOP30 on the intestinal epithelial barrier is not well understood. Therefore, the aim of this study is to investigate the protective effect of AOP30 on the intestinal epithelial barrier using a lipopolysaccharide (LPS)-induced intestinal epithelial barrier dysfunction model and further explore its underlying mechanisms. Cytotoxicity, transepithelial electrical resistance (TEER) values, and Fluorescein isothiocyanate (FITC)–dextran flux are measured. Simultaneously, the protein and mRNA levels of tight junction (TJ) proteins, including zonula occludens-1 (ZO-1), Occludin, and Claudin-1, are determined using Western blotting and reverse-transcription quantitative polymerase chain reaction methods, respectively. The results indicate that AOP30 restores the LPS-induced decrease in the TEER value and cell viability. Furthermore, it increases the mRNA and protein expression of ZO-1, Occludin, and Claudin-1. Notably, ZO-1 is the primary tight junction protein altered in response to LPS-induced intestinal epithelial dysfunction. Additionally, AOP30 downregulates the production of TNFα via the Toll-like receptor 4 (TLR4)/NF-κB signaling pathway. Collectively, the findings of this study indicate that AOP30 can be developed as a functional food ingredient or natural therapeutic agent for addressing intestinal epithelial barrier dysfunction. It sheds light on the role of AOP30 in improving intestinal epithelial function. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL,), incubated alone (<b>A</b>) and co-incubated (<b>B</b>) with 1 μg/mL of LPS for 24 h, on cell viability in Caco-2 cells. Data are presented as mean ± SD. Bars with different superscript letters indicate significantly differences among the groups (<span class="html-italic">p</span> &lt;  0.05).</p>
Full article ">Figure 1 Cont.
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL,), incubated alone (<b>A</b>) and co-incubated (<b>B</b>) with 1 μg/mL of LPS for 24 h, on cell viability in Caco-2 cells. Data are presented as mean ± SD. Bars with different superscript letters indicate significantly differences among the groups (<span class="html-italic">p</span> &lt;  0.05).</p>
Full article ">Figure 2
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL) co-incubated with 1 μg/mL of LPS for 24 h on TEER value in Caco-2 cells. Data are presented as mean ± SD bars. with different superscript letters indicating significantly differences among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL), co-incubated with 1 μg/mL of LPS for 24 h, on FITC–dextran paracellular transport; briefly, FITC–dextran (4 kD) was added into the upper chamber for 3.5 h after being replaced with HBSS solution. Thus, Caco-2 cell monolayer permeability was evaluated by measuring FITC–dextran paracellular transport. Data are presented as mean ± SD bars, with different superscript letters indicating significantly difference among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL), co-incubated with 1 μg/mL of LPS for 24 h, on mRNA expression of ZO-1, Occludin, and Claudin-1 in Caco-2 cells. GAPDH is used as an internal reference, data are presented as mean ± SD, and bars with differing superscript letters indicate significant differences among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/mL), co-incubated with 1 μg/mL of LPS for 24 h, on tight junction protein expression in Caco-2 cells. Western blotting analysis is conducted on the expression level of ZO-1, Occludin, and Claudin-1 (<b>A</b>,<b>B</b>), and GAPDH is used as an internal reference. Data are presented as mean ± SD; bars with differing superscript letters indicate significant differences among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Effect of different concentrations of AOP30 (200, 400, and 800 μg/m) co-incubated with 1 μg/mL of LPS for 24 h on the secretion of TNFα in Caco-2 cells, data are presented as mean ± SD, and bars with differing superscript letters indicate significant differences among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>The effect of different concentrations of AOP30 (200, 400, and 800 μg/mL co-incubated with 1 μg/mL of LPS for 24 h on TLR4/NF-κB signaling pathway in Caco-2 cells. Western blotting analysis is conducted on the expression level of TLR4 (<b>A</b>,<b>B</b>), and phospho-NF-κB p65 (<b>A</b>,<b>C</b>). GAPDH is used as an internal reference, data are presented as mean ± SD, and bars with differing superscript letters indicate significant differences among the groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
18 pages, 1962 KiB  
Article
Revealing the Hypoglycemic Effect of Red Yeast Rice: Perspectives from the Inhibition of α-Glucosidase and the Anti-Glycation Capability by Ankaflavin and Monascin
by Shufen Wu, Changyan Dong, Meihui Zhang, Yi Cheng, Xiaobo Cao, Benxu Yang, Chao Li and Xin Peng
Foods 2024, 13(10), 1573; https://doi.org/10.3390/foods13101573 - 17 May 2024
Viewed by 986
Abstract
Red yeast rice dietary supplements have been proven to ameliorate hyperglycemia, but the mechanism was unclear. In this work, ankaflavin (AK) and monascin (MS), as typical pigments derived from red yeast rice, were found to exert noteworthy inhibitory ability against α-glucosidase, with an [...] Read more.
Red yeast rice dietary supplements have been proven to ameliorate hyperglycemia, but the mechanism was unclear. In this work, ankaflavin (AK) and monascin (MS), as typical pigments derived from red yeast rice, were found to exert noteworthy inhibitory ability against α-glucosidase, with an IC50 of 126.5 ± 2.5 and 302.6 ± 2.5 μM, respectively, compared with acarbose (IC50 = 341.3 ± 13.6 μM). They also exhibited mixed-type inhibition of α-glucosidase in vitro and caused fluorescence quenching through the static-quenching process. Molecular-docking studies indicated that AK and MS bind to amino acid residues outside the catalytic center, which induces structural changes in the enzyme, thus influencing its catalytic activity. The anti-glycation ability of Monascus-fermented products was evaluated, and they exhibited a high inhibition rate of 87.1% in fluorescent advanced glycation end-product formation at a concentration of 0.2 mg mL−1, while aminoguanidine showed a rate of 75.7% at the same concentration. These results will be significant in broadening the application scope of Monascus pigments, especially AK and MS, in treating type 2 diabetes. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) The inhibitory activities of AK, MS and acarbose towards α-Glu. Conditions: C<sub>α-Glu</sub> = 0.10 U/mL, C<span class="html-italic"><sub>p</sub></span><sub>NPG</sub> = 2.08 mM, pH = 6.8, T = 310 K. Plots of <span class="html-italic">v</span> versus the concentration of α-Glu in the presence of AK (<b>B</b>) and MS (<b>C</b>). Lineweaver–Burk plots for AK (<b>D</b>) and MS (<b>E</b>), and the secondary plots (in the inset) represent the slope and <span class="html-italic">Y</span>-intercept versus the concentration of inhibitor, respectively.</p>
Full article ">Figure 2
<p>Fluorescence spectra of α-Glu in the presence of AK (<b>A</b>) and MS (<b>B</b>). Conditions: C<sub>α-Glu</sub> = 2.0 μM, pH = 6.8, T = 298 K, <span class="html-italic">λ<sub>ex</sub></span> = 280 nm. Impact of AK (<b>C</b>) and MS (<b>D</b>) on the synchronous fluorescence spectra of α-Glu at ∆<span class="html-italic">λ</span> = 15 and 60 nm. C<sub>α-Glu</sub> = 2.0 μM, C<sub>AK</sub> = 0, 4.0, 8.0, 12.0, 16.0, 20.0, 24.0 and 28.0 μM, respectively, and C<sub>MS</sub> = 0, 3.0, 6.0, 9.0, 12.0, 15.0, 18.0 and 21.0 μM, respectively. The inset is the fluorescence spectra of α-Glu. Stern–Volmer plots for the quenching of α-Glu fluorescence by AK (<b>E</b>) and MS (<b>F</b>) at different temperatures. The plots of log(1/([Q<sub>t</sub>] − (F<sub>0</sub> − F)[P<sub>t</sub>]/[F<sub>0</sub>])) versus log((F<sub>0</sub> − F)/F) for the binding of AK (<b>G</b>) and MS (<b>H</b>) to α-Glu, respectively.</p>
Full article ">Figure 3
<p>Molecular-docking results of the AK-α-Glu system and MS-α-Glu systems in the presence of the substrate (<span class="html-italic">p</span>NPG). Structure of α-Glu shows the subdomains and binding sites of AK (<b>A</b>) and MS (<b>C</b>). The 2D detailed view shows the interactions between AK (<b>B</b>)/MS (<b>D</b>) and the neighboring residues.</p>
Full article ">Figure 4
<p>Effect of different concentrations of MPs on the formation of glycation products in the in vitro BSA–Fru reaction: fructosamine (<b>A</b>), α-dicarbonyl compounds (<b>B</b>) and total fluorescent AGEs (<b>C</b>). Carbonyl group content (<b>D</b>), sulphydryl (SH) group content (<b>E</b>), far-UV CD spectra (<b>F</b>), the surface hydrophobicity (<b>G</b>) and ThT fluorescence intensity (<b>H</b>) of native BSA, glycated BSA and glycated BSA treated with varying concentrations of MPs. AG was used as a positive control. Different lowercase letters above columns represent a significant difference (<span class="html-italic">p</span> &lt; 0.05) among glycated BSA treated with MPs, and different capital letters above columns represent a significant difference (<span class="html-italic">p</span> &lt; 0.05) among glycated BSA treated with AG.</p>
Full article ">
15 pages, 1022 KiB  
Article
Do Young Consumers Care about Antioxidant Benefits and Resveratrol and Caffeic Acid Consumption?
by Cristina Ștefania Gălbău, Mihaela Badea and Laura Elena Gaman
Nutrients 2024, 16(10), 1439; https://doi.org/10.3390/nu16101439 - 10 May 2024
Viewed by 858
Abstract
Resveratrol and caffeic acid are some of the most consumed antioxidants during the day, so their importance as sources and their benefits need to be evaluated and updated. This survey aimed not only to analyze whether young Romanian consumers are informed about the [...] Read more.
Resveratrol and caffeic acid are some of the most consumed antioxidants during the day, so their importance as sources and their benefits need to be evaluated and updated. This survey aimed not only to analyze whether young Romanian consumers are informed about the benefits of antioxidants in general, and resveratrol and caffeic acid in particular, but also to observe the degree of nutritional education of these participants. Young consumers know the concept of antioxidants relatively well; they managed to give examples of antioxidants and indicate their effects. The majority of those chosen drink wine and coffee, but many are unaware of their health advantages and antioxidant properties. Students are less familiar with the antioxidant chemicals resveratrol and caffeic acid. It is advised to have a thorough understanding of these significant antioxidants and their nutritional content as they are present in our regular diets, and further studies on different kinds of antioxidants are required to increase the awareness of people concerning their importance in daily life. Full article
Show Figures

Figure 1

Figure 1
<p>Chemical structure of trans-resveratrol and cis-resveratrol.</p>
Full article ">Figure 2
<p>Sources of resveratrol and its health benefits.</p>
Full article ">Figure 3
<p>Chemical structure of caffeic acid.</p>
Full article ">Figure 4
<p>Sources of caffeic acid and its health benefits.</p>
Full article ">
17 pages, 2355 KiB  
Article
Sustainable Valorization of Coffee Silverskin: Extraction of Phenolic Compounds and Proteins for Enzymatic Production of Bioactive Peptides
by Wilasinee Jirarat, Tanyawat Kaewsalud, Kamon Yakul, Pornchai Rachtanapun and Thanongsak Chaiyaso
Foods 2024, 13(8), 1230; https://doi.org/10.3390/foods13081230 - 17 Apr 2024
Viewed by 1072
Abstract
Coffee silverskin (CS), a by-product of the coffee roasting process, has high protein content (16.2−19.0%, w/w), making it a potential source for plant protein and bioactive peptide production. This study aims to develop innovative extraction methods for phenolic compounds and [...] Read more.
Coffee silverskin (CS), a by-product of the coffee roasting process, has high protein content (16.2−19.0%, w/w), making it a potential source for plant protein and bioactive peptide production. This study aims to develop innovative extraction methods for phenolic compounds and proteins from CS. The conditions for hydrothermal (HT) extraction of phenolic compounds from CS were optimized by varying CS loading (2.5−10%, w/v), temperature (110−130 °C), and time (5−30 min) using a one-factor-at-a-time (OFAT) approach. The highest TPC of 55.59 ± 0.12 µmole GAE/g CS was achieved at 5.0% (w/v) CS loading and autoclaving at 125 °C for 25 min. Following hydrothermal extraction, CS protein was extracted from HT-extracted solid fraction by microwave-assisted alkaline extraction (MAE) using 0.2 M NaOH at 90 W for 2 min, resulting in a protein recovery of 12.19 ± 0.39 mg/g CS. The CS protein was then subjected to enzymatic hydrolysis using protease from Bacillus halodurans SE5 (protease_SE5). Protease_SE5-derived CS protein hydrolysate had a peptide concentration of 0.73 ± 0.09 mg/mL, with ABTS, DPPH, and FRAP values of 15.71 ± 0.10, 16.63 ± 0.061, and 6.48 ± 0.01 µmole TE/mL, respectively. Peptide identification by LC-MS/MS revealed several promising biological activities without toxicity or allergenicity concerns. This study’s integrated approach offers a sustainable and efficient method for extracting valuable compounds from CS, with potential applications in the food and pharmaceutical industries. Full article
Show Figures

Figure 1

Figure 1
<p>The antioxidant activity and TPC of the HT-extracted CS liquid fraction. The effects of (<b>a</b>) CS loading (% <span class="html-italic">w</span>/<span class="html-italic">v</span>), (<b>b</b>) temperature (°C), and (<b>c</b>) time (min) on antioxidant activity and TPC were investigated. The experiments are performed in triplicate (<span class="html-italic">n</span> = 3). The results are reported as mean  ±  SD. Different letters (a–e) indicate significant differences at <span class="html-italic">p</span>  &lt;  0.05 according to analysis by Duncan’s multiple range test.</p>
Full article ">Figure 2
<p>Enzymatic production of bioactive peptides from CS. Protein recovery, decrease in protein concentration, and increase in peptide concentration by CAE at 50 °C for 240 min (<b>a</b>) and 90 °C for 30 min (<b>b</b>). The increase in antioxidant activity of CS protein hydrolysate derived from CAE at 50 °C for 240 min (<b>c</b>) and 90 °C for 30 min (<b>d</b>). The protein from CS was extracted by CAE, followed by enzymatic hydrolysis using protease_SE5 (200,000 U/g protein) at pH 9.5 and 55 °C for 12 h. Different letters (a–e) indicate significant differences at <span class="html-italic">p</span>  &lt;  0.05 according to analysis by Duncan’s multiple range test.</p>
Full article ">Figure 3
<p>The molecular weight distribution of CS protein hydrolysate by tris-tricine SDS-PAGE analysis. Electrophoresis was carried out under denaturing conditions in 16.5% polyacrylamide gel. The CS protein was extracted by 0.2 M NaOH at 50 °C for 240 min (CAE), ultrasound-assisted alkaline extraction (UAE) for 10 min, and microwave-assisted alkaline extraction (MAE) at 90 W for 2 min. The enzymatic hydrolysis was performed at pH 9.5 and 55 °C for 12 h using protease_SE5 (200,000 U/g protein).</p>
Full article ">Figure 4
<p>Peptide concentration and antioxidant activity of CS protein hydrolysate. The antioxidant activity of CS protein hydrolysate by ABTS, DPPH, and FRAP assay. The CS protein was extracted by MAE at 90 W for 2 min using 0.2 M NaOH and subjected to hydrolysis by either protease_SE5 or Alcalase (200,000 U/g protein) at pH 9.5 and 55 °C for 12 h.</p>
Full article ">Figure 5
<p>Size exclusion chromatography of &lt;3 kDa from protease_SE5 (<b>a</b>) and Alcalase (<b>b</b>). Each fraction (2.5 mL) was eluted by deionized water at a flow rate of 23 mL/h. The active fractions were lyophilized before analysis by LC-MS/MS and <span class="html-italic">de novo</span> peptide sequencing. Antioxidant activity of active fractions from protease_SE5 (<b>c</b>) and Alcalase (<b>d</b>) obtained from the Sephadex G-25 column. The experiments are performed in triplicate (<span class="html-italic">n</span> = 3). Different letters (a–d) indicate significant differences at <span class="html-italic">p</span>  &lt;  0.05 according to analysis by Duncan’s multiple range test.</p>
Full article ">
12 pages, 1232 KiB  
Article
A Low FODMAP Diet Supplemented with L-Tryptophan Reduces the Symptoms of Functional Constipation in Elderly Patients
by Cezary Chojnacki, Marta Mędrek-Socha, Aleksandra Błońska, Janusz Błasiak, Tomasz Popławski, Jan Chojnacki and Anita Gąsiorowska
Nutrients 2024, 16(7), 1027; https://doi.org/10.3390/nu16071027 - 1 Apr 2024
Viewed by 1578
Abstract
(1) Background: The elderly suffer from functional constipation (FC), whose causes are not fully known, but nutritional factors may play a role. The aim of the present study was to assess the effect of a low FODMAP diet supplemented with L-tryptophan (TRP) on [...] Read more.
(1) Background: The elderly suffer from functional constipation (FC), whose causes are not fully known, but nutritional factors may play a role. The aim of the present study was to assess the effect of a low FODMAP diet supplemented with L-tryptophan (TRP) on its metabolism and symptoms of functional constipation in elderly patients. (2) Methods: This study included 40 people without abdominal complaints (Group I, controls) and 60 patients with FC, diagnosed according to the Rome IV Criteria (Group II). Two groups were randomly selected: Group IIA (n = 30) was qualified for administration of the low FODMAP diet, and the diet of patients of Group IIB (n = 30) was supplemented with 1000 mg TRP per day. The severity of abdominal symptoms was assessed with an abdominal pain index ranging from 1 to 7 points (S-score). The concentration of TRP and its metabolites, 5-hydroxyindoleacetic acid (5-HIAA), kynurenine (KYN), and 3-indoxyl sulfate (3-IS) in urine were determined using the LC-MS/MS method. (3) Results: In Group II, 5-HIAA concentration in urine was lower, and KYN and 3-IS concentrations were higher than in the control group. A negative correlation was found between the S-score and urinary concentration of 5-HIAA (p < 0.001), and 3-IS concentration was positively correlated with the S-score. However, the correlation between the S-score and 3-IS concentration was negative (p < 0.01). After a dietary intervention, 5-HIAA concentration increased in both groups, and the severity of symptoms decreased, but the decrease was more pronounced in Group IIB. (4) Conclusion: A low FODMAP diet supplemented with L-tryptophan has beneficial effects in elderly patients suffering from functional constipation. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>). Correlation between the severity of symptoms (S-score, points) and 5-hydroxyindoleacetic acid (5-HIAA, mg/gCr) in urine; <span class="html-italic">r =</span> −0.5266). (<b>B</b>). Correlation between symptom severity and urinary levels of 3-indoxyl sulfate (3-IS, _g/gCr); <span class="html-italic">r</span> = 0.3616. The correlations were analyzed with the Spearman rank test with the rho rank correlation coefficient (r).</p>
Full article ">Figure 2
<p>Urine concentration of tryptophan (TRP; (<b>A</b>)), 5-hydroxyindoleacetic acid (5-HIAA; (<b>B</b>)), kynurenine (KYN; (<b>C</b>)), indican (3-IS; (<b>D</b>)), and severity of symptoms in patients with functional constipation before (dark blue) and after nutritional treatment (green); the Wilcoxon matched-pair signed-rank test was used to compare before and after groups; <span class="html-italic">* p &lt;</span> 0.05.</p>
Full article ">Figure 3
<p>Comparison of symptom severity (S-score) in Group IIA (dark blue) and IIB (light blue) before (<b>A</b>) and after (<b>B</b>) nutritional intervention; differences in both groups before and after treatment were evaluated by the Wilcoxon signed-rank test, **** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
13 pages, 2153 KiB  
Article
The Mechanism of the Anti-Obesity Effects of a Standardized Brassica juncea Extract in 3T3-L1 Preadipocytes and High-Fat Diet-Induced Obese C57BL/6J Mice
by June-Seok Lim, Ji-Hyun Im, Xionggao Han, Xiao Men, Geon Oh, Xiaolu Fu, Woonsang Hwang, Sun-Il Choi and Ok-Hwan Lee
Nutrients 2024, 16(6), 846; https://doi.org/10.3390/nu16060846 - 15 Mar 2024
Cited by 1 | Viewed by 1409
Abstract
Obesity is a global health concern. Recent research has suggested that the development of anti-obesity ingredients and functional foods should focus on natural products without side effects. We examined the effectiveness and underlying mechanisms of Brassica juncea extract (BJE) in combating obesity via [...] Read more.
Obesity is a global health concern. Recent research has suggested that the development of anti-obesity ingredients and functional foods should focus on natural products without side effects. We examined the effectiveness and underlying mechanisms of Brassica juncea extract (BJE) in combating obesity via experiments conducted in both in vitro and in vivo obesity models. In in vitro experiments conducted in a controlled environment, the application of BJE demonstrated the ability to suppress the accumulation of lipids induced by MDI in 3T3-L1 adipocytes. Additionally, it downregulated adipogenic-related proteins peroxisome proliferator-activated receptor-γ (PPAR-γ), CCAAT/enhancer-binding protein-α (C/EBP-α), adipocyte protein 2 (aP2), and lipid synthesis-related protein acetyl-CoA carboxylase (ACC). It also upregulated the heat generation protein peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α) and fatty acid oxidation protein carnitine palmitoyltransferase-1 (CPT-1). The oral administration of BJE decreased body weight, alleviated liver damage, and inhibited the accumulation of lipids in mice with diet-induced obesity resulting from a high-fat diet. The inhibition of lipid accumulation by BJE in vivo was associated with a decreased expression of adipogenic and lipid synthesis proteins and an increased expression of heat generation and fatty acid oxidation proteins. BJE administration improved obesity by decreasing adipogenesis and activating heat generation and fatty acid oxidation in 3T3-L1 cells and in HFD-induced obese C57BL/6J mice. These results suggest that BJE shows potential as a natural method for preventing metabolic diseases associated with obesity. Full article
Show Figures

Figure 1

Figure 1
<p>HPLC chromatogram of BJE. The monitoring wavelength for sinigrin was set at 228 nm.</p>
Full article ">Figure 2
<p>Effect of BJE on cell viability (<b>A</b>) and lipid accumulation (<b>B</b>) on different 3T3-L1 adipocytes. All results are presented as the mean ± SD of three independent in triplicate. Bars with different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 using Duncan’s multiple range test.</p>
Full article ">Figure 3
<p>Effect of BJE on the expression of proteins related to adipogenic, lipid synthesis, fatty acid oxidation, and heat generation in 3T3-L1. PPAR-γ (<b>A</b>). C/EBP-α (<b>B</b>). aP2 (<b>C</b>). p-ACC/ACC (<b>D</b>). PGC-1α (<b>E</b>). CPT-1 (<b>F</b>). All results are presented as the mean ± SD of three independent in triplicate. Bars with different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 using Duncan’s multiple range test.</p>
Full article ">Figure 4
<p>Effect of BJE on body weight, food efficiency, adipose tissue mass, organ mass, and adipose tissue size in HFD-induced obese C57BL/6J mice. eWAT were subjected to staining with H&amp;E and subsequently examined using a microscope. Changes in total body weight (<b>A</b>). Food efficiency ratio (<b>B</b>). eWAT weight (<b>C</b>). Other tissue weight (<b>D</b>). Histology of the eWAT (<b>E</b>). All values are expressed as the mean ± SD, and statistical analyses were performed using Dunnett’s <span class="html-italic">t</span>-test. ### <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. HFD group.</p>
Full article ">Figure 4 Cont.
<p>Effect of BJE on body weight, food efficiency, adipose tissue mass, organ mass, and adipose tissue size in HFD-induced obese C57BL/6J mice. eWAT were subjected to staining with H&amp;E and subsequently examined using a microscope. Changes in total body weight (<b>A</b>). Food efficiency ratio (<b>B</b>). eWAT weight (<b>C</b>). Other tissue weight (<b>D</b>). Histology of the eWAT (<b>E</b>). All values are expressed as the mean ± SD, and statistical analyses were performed using Dunnett’s <span class="html-italic">t</span>-test. ### <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. HFD group.</p>
Full article ">Figure 5
<p>Effect of BJE on the expression of proteins related to adipogenesis, lipid synthesis, fatty acid oxidation, and heat generation in HFD-induced obese C57BL/6J mice. PPAR-γ (<b>A</b>). C/EBP-α (<b>B</b>). aP2 (<b>C</b>). p-ACC/ACC (<b>D</b>). PGC-1α (<b>E</b>). CPT-1 (<b>F</b>). All results are presented as the mean ± SD of 3 independent in triplicate. Bars with different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 using Duncan’s multiple range test.</p>
Full article ">Figure 5 Cont.
<p>Effect of BJE on the expression of proteins related to adipogenesis, lipid synthesis, fatty acid oxidation, and heat generation in HFD-induced obese C57BL/6J mice. PPAR-γ (<b>A</b>). C/EBP-α (<b>B</b>). aP2 (<b>C</b>). p-ACC/ACC (<b>D</b>). PGC-1α (<b>E</b>). CPT-1 (<b>F</b>). All results are presented as the mean ± SD of 3 independent in triplicate. Bars with different letters indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 using Duncan’s multiple range test.</p>
Full article ">
22 pages, 1642 KiB  
Review
Sensory Nutrition and Bitterness and Astringency of Polyphenols
by Naomi Osakabe, Takafumi Shimizu, Yasuyuki Fujii, Taiki Fushimi and Vittorio Calabrese
Biomolecules 2024, 14(2), 234; https://doi.org/10.3390/biom14020234 - 17 Feb 2024
Cited by 4 | Viewed by 2364
Abstract
Recent studies have demonstrated that the interaction of dietary constituents with taste and olfactory receptors and nociceptors expressed in the oral cavity, nasal cavity and gastrointestinal tract regulate homeostasis through activation of the neuroendocrine system. Polyphenols, of which 8000 have been identified to [...] Read more.
Recent studies have demonstrated that the interaction of dietary constituents with taste and olfactory receptors and nociceptors expressed in the oral cavity, nasal cavity and gastrointestinal tract regulate homeostasis through activation of the neuroendocrine system. Polyphenols, of which 8000 have been identified to date, represent the greatest diversity of secondary metabolites in plants, most of which are bitter and some of them astringent. Epidemiological studies have shown that polyphenol intake contributes to maintaining and improving cardiovascular, cognitive and sensory health. However, because polyphenols have very low bioavailability, the mechanisms of their beneficial effects are unknown. In this review, we focused on the taste of polyphenols from the perspective of sensory nutrition, summarized the results of previous studies on their relationship with bioregulation and discussed their future potential. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Chemical structure of flavonoid.</p>
Full article ">Figure 2
<p>Hromesis (<b>a</b>) and the mechanism of stress response (<b>b</b>) induced by astringent polyphenols. Stress stimuli input to the nucleus fasciculus solitarius (NTS) project norepinephrine via the locus coeruleus (NTS) to the hypothalamus, brainstem and other brain regions. The stimulation increases sympathetic nerve activity (sympathetic nervous -adrenal medulla axis), which causes changes in various organs via adrenergic receptors; adrenaline is also secreted into the blood from the adrenal glands, inducing browning in adipose and synthesizing skeletal muscle proteins. The paraventricular nucleus (PVN) of the hypothalamus synthesizes corticotropin-releasing hormone (CRH), which stimulates the secretion of adrenocorticotropic hormone (ACTH) from proopiomelanocortin (PONC) in the pituitary gland into the blood. ACTH promotes the secretion of glucocorticoids from the adrenal glands into the blood, and glucocorticoids provide negative feedback to the hypothalamus-pituitary-adrenocortical (HPA) axis via receptors expressed CNS.</p>
Full article ">Figure 3
<p>Hypothetical recognition mechanism of astringent polyphenols by TRP channel.</p>
Full article ">Figure 4
<p>Illustrations of two hypotheses for the mechanisms underlying the observed effects of a single (<b>a</b>) and repeated oral administration (<b>b</b>) of astringent polyphenols on micro-and systemic circulation.</p>
Full article ">
16 pages, 7096 KiB  
Review
A Literature Review of the Pharmacological Effects of Jujube
by Deqi Zhu, Ning Jiang, Ning Wang, Yufen Zhao and Xinmin Liu
Foods 2024, 13(2), 193; https://doi.org/10.3390/foods13020193 - 6 Jan 2024
Cited by 1 | Viewed by 3182
Abstract
Jujube is a plant native to China that could be used in medicine and food. Its dried fruit is a superior herb commonly used in traditional Chinese medicine formulations for its calming effect and for nourishing the blood and strengthening the spleen and [...] Read more.
Jujube is a plant native to China that could be used in medicine and food. Its dried fruit is a superior herb commonly used in traditional Chinese medicine formulations for its calming effect and for nourishing the blood and strengthening the spleen and stomach. Jujube contains numerous active components including polysaccharides, phenols, and triterpene acids, which show a diverse array of pharmacological activities such as neuroprotection and the prevention and treatment of cardiovascular diseases. In this paper, the research status of jujube over the past two decades has been statistically evaluated. Meanwhile, by tracking the latest research advances, the pharmacological efficacy and molecular mechanisms of jujube are exhaustively expounded to provide specific and systematic references for further research on the pharmacological effects of jujube and its application in the food and pharmaceutical industries. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The growth, distribution and co-occurrence analysis of jujube research from 2003 to 2022. (<b>A</b>) Growth of jujube research; (<b>B</b>) Co-occurrence analysis of article keywords; (<b>C</b>) Distribution of jujube research.</p>
Full article ">Figure 2
<p>The pharmacological effects and the potential active ingredients of jujube.</p>
Full article ">
19 pages, 6617 KiB  
Article
Effects of Long-Term Administration of Bovine Bone Gelatin Peptides on Myocardial Hypertrophy in Spontaneously Hypertensive Rats
by Songmin Cao, Xinyu Wang, Lujuan Xing and Wangang Zhang
Nutrients 2023, 15(24), 5021; https://doi.org/10.3390/nu15245021 - 6 Dec 2023
Viewed by 1259
Abstract
The research purpose was to investigate the effects and the underlying molecular mechanisms of bovine bone gelatin peptides (BGP) on myocardial hypertrophy in spontaneously hypertensive rats (SHR). BGP relieved myocardial hypertrophy and fibrosis in SHR rats in a dose-dependent manner by reducing the [...] Read more.
The research purpose was to investigate the effects and the underlying molecular mechanisms of bovine bone gelatin peptides (BGP) on myocardial hypertrophy in spontaneously hypertensive rats (SHR). BGP relieved myocardial hypertrophy and fibrosis in SHR rats in a dose-dependent manner by reducing the left ventricular mass index, myocardial cell diameter, myocardial fibrosis area, and levels of myocardial hypertrophy markers (atrial natriuretic and brain natriuretic peptide). Label-free quantitative proteomics analysis showed that long-term administration of BGP changed the left ventricle proteomes of SHR. The 37 differentially expressed proteins in the high-dose BGP group participated in multiple signaling pathways associated with cardiac hypertrophy and fibrosis indicating that BGP could play a cardioprotective effect on SHR rats by targeting multiple signaling pathways. Further validation experiments showed that a high dose of BGP inhibited the expression of phosphoinositide 3-kinase (Pi3k), phosphorylated protein kinase B (p-Akt), and transforming growth factor-beta 1 (TGF-β1) in the myocardial tissue of SHR rats. Together, BGP could be an effective candidate for functional nutritional supplements to inhibit myocardial hypertrophy and fibrosis by negatively regulating the TGF-β1 and Pi3k/Akt signaling pathways. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effects of long-term oral BGP on myocardial hypertrophy and fibrosis in SHR rats. Note: (<b>A</b>) Heart weight index (HMI) and left ventricular mass index (LVWI) of rats; (<b>B</b>) The levels of ANP and BNP of rats; (<b>C</b>) HE and Masson staining of the left ventricle of rats with the magnification of 1×, 400×, 400× respectively; (<b>D</b>) Cardiomyocyte diameter of rats; (<b>E</b>) Percentage of myocardial fibrosis area of rats. WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6); * and ** indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from SHR-NCG group. # and ## indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from SHR-PCG group. ++ indicates an extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from the WKY-NCG group. ns indicates no significant difference between different groups (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 1 Cont.
<p>Effects of long-term oral BGP on myocardial hypertrophy and fibrosis in SHR rats. Note: (<b>A</b>) Heart weight index (HMI) and left ventricular mass index (LVWI) of rats; (<b>B</b>) The levels of ANP and BNP of rats; (<b>C</b>) HE and Masson staining of the left ventricle of rats with the magnification of 1×, 400×, 400× respectively; (<b>D</b>) Cardiomyocyte diameter of rats; (<b>E</b>) Percentage of myocardial fibrosis area of rats. WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6); * and ** indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from SHR-NCG group. # and ## indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from SHR-PCG group. ++ indicates an extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from the WKY-NCG group. ns indicates no significant difference between different groups (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 2
<p>Volcano plots (<b>A</b>) and hierarchical cluster analysis (<b>B</b>) of differentially expressed proteins in rats identified by label-free proteomics. Note: Volcano plots: Compared with SHR-NCG, the differentially expressed proteins in WKY-NCG (<b>A-1</b>), SHR-LPG (<b>A-2</b>), SHR-HPG (<b>A-3</b>), and SHR-PCG (<b>A-4</b>) groups identified by label-free proteomics; The gray points in image (<b>A</b>)(<b>A-1</b>–<b>A-4</b>) represent proteins with insignificant differences. WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2 Cont.
<p>Volcano plots (<b>A</b>) and hierarchical cluster analysis (<b>B</b>) of differentially expressed proteins in rats identified by label-free proteomics. Note: Volcano plots: Compared with SHR-NCG, the differentially expressed proteins in WKY-NCG (<b>A-1</b>), SHR-LPG (<b>A-2</b>), SHR-HPG (<b>A-3</b>), and SHR-PCG (<b>A-4</b>) groups identified by label-free proteomics; The gray points in image (<b>A</b>)(<b>A-1</b>–<b>A-4</b>) represent proteins with insignificant differences. WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 2 Cont.
<p>Volcano plots (<b>A</b>) and hierarchical cluster analysis (<b>B</b>) of differentially expressed proteins in rats identified by label-free proteomics. Note: Volcano plots: Compared with SHR-NCG, the differentially expressed proteins in WKY-NCG (<b>A-1</b>), SHR-LPG (<b>A-2</b>), SHR-HPG (<b>A-3</b>), and SHR-PCG (<b>A-4</b>) groups identified by label-free proteomics; The gray points in image (<b>A</b>)(<b>A-1</b>–<b>A-4</b>) represent proteins with insignificant differences. WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6).</p>
Full article ">Figure 3
<p>GO annotation analysis and signal pathway analysis of differentially abundant proteins in the high-dose BGP group. Note: (<b>A</b>) Go annotation analysis, (<b>B</b>) KEGG pathway analysis, (<b>C</b>) Protein-protein interaction analysis (PPI).</p>
Full article ">Figure 3 Cont.
<p>GO annotation analysis and signal pathway analysis of differentially abundant proteins in the high-dose BGP group. Note: (<b>A</b>) Go annotation analysis, (<b>B</b>) KEGG pathway analysis, (<b>C</b>) Protein-protein interaction analysis (PPI).</p>
Full article ">Figure 4
<p>Expression levels of differentially expressed proteins of Ppp1cc (<b>B</b>), Gys1 (<b>C</b>), and Eef2 (<b>D</b>) in rat myocardial tissue. Note: (<b>A</b>) The protein bands (Eef2, Gys1, and Pp1cc) in Western blot analysis; WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6); * and ** indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from SHR-NCG group. # and ## indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) compared to SHR-PCG group. ++ indicates an extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from the WKY-NCG group. ns indicates no significant difference between different groups (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 5
<p>Effect of long-term oral administration of BGP on Pi3k/Akt signaling pathway and TGF-β1 level in SHR rats. Note: (<b>A</b>) The protein bands (Pi3k, Akt, and p-Akt) in Western blot analysis; (<b>B</b>) expression levels of TGF-β1; (<b>C</b>) expression levels of Pi3K; (<b>D</b>) expression levels of Akt; (<b>E</b>) expression levels of p-Akt; WKY-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-NCG: 0.9% saline (<span class="html-italic">n</span> = 6), SHR-LPG: low dose BGP group-100 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-HPG: high dose BGP group-200 mg/kg BW (<span class="html-italic">n</span> = 6), SHR-PCG: captopril-10.0 mg/kg BW (<span class="html-italic">n</span> = 6); * and ** indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) compared to SHR-NCG group. # and ## indicate significant difference (<span class="html-italic">p</span> &lt; 0.05) and extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) compared to SHR-PCG group. ++ indicates an extremely significant difference (<span class="html-italic">p</span> &lt; 0.01) from the WKY-NCG group. ns indicates no significant difference between different groups (<span class="html-italic">p</span> &gt; 0.05).</p>
Full article ">Figure 6
<p>The signal pathways of BGP preventing myocardial hypertrophy and myocardial fibrosis in SHR rats.</p>
Full article ">
19 pages, 33396 KiB  
Article
Probiotic and Muscadine Grape Extract Interventions Shift the Gut Microbiome and Improve Metabolic Parameters in Female C57BL/6 Mice
by Tiffany M. Newman, Adam S. Wilson, Kenysha Y. J. Clear, E. Ann Tallant, Patricia E. Gallagher and Katherine L. Cook
Cells 2023, 12(22), 2599; https://doi.org/10.3390/cells12222599 - 10 Nov 2023
Cited by 1 | Viewed by 1656
Abstract
Obesity and Western-like diet consumption leads to gut microbiome dysbiosis, which is associated with the development of cardio-metabolic diseases and poor health outcomes. The objective of this study was to reduce Western diet-mediated gut microbial dysbiosis, metabolic dysfunction, and systemic inflammation through the [...] Read more.
Obesity and Western-like diet consumption leads to gut microbiome dysbiosis, which is associated with the development of cardio-metabolic diseases and poor health outcomes. The objective of this study was to reduce Western diet-mediated gut microbial dysbiosis, metabolic dysfunction, and systemic inflammation through the administration of a novel combined intervention strategy (oral probiotic bacteria supplements and muscadine grape extract (MGE)). To do so, adult female C57BL/6 mice were fed a low-fat control or Western-style diet and sub-grouped into diet alone, probiotic intervention, antibiotic treatments, MGE supplementation, a combination of MGE and probiotics, or MGE and antibiotics for 13 weeks. Mouse body weight, visceral adipose tissue (VAT), liver, and mammary glands (MG) were weighed at the end of the study. Fecal 16S rRNA sequencing was performed to determine gut bacterial microbiome populations. Collagen, macrophage, and monocyte chemoattractant protein-1 (MCP-1) in the VAT and MG tissue were examined by immunohistochemistry. Adipocyte diameter was measured in VAT. Immunohistochemistry of intestinal segments was used to examine villi length, muscularis thickness, and goblet cell numbers. We show that dietary interventions in Western diet-fed mice modulated % body weight gain, visceral adiposity, MG weight, gut microbial populations, and inflammation. Intervention strategies in both diets effectively reduced VAT and MG fibrosis, VAT and MG macrophages, adipocyte diameter, and VAT and MG MCP-1. Interventions also improved intestinal health parameters. In conclusion, dietary intervention with MGE and probiotics modulates several microbial, inflammatory, and metabolic factors reducing poor health outcomes associated with Western diet intake. Full article
Show Figures

Figure 1

Figure 1
<p>Dietary intervention modulates body condition and tissue morphology. (<b>A</b>) Body weight of female C57BL/6 mice following 13 weeks of exposure to diets and intervention strategies. (<b>B</b>) % change in body weight. (<b>C</b>) Visceral adipose tissue weight at study completion. (<b>D</b>) Normalized visceral adipose tissue weight at the end of the study. (<b>E</b>) Weight of the right lower (4/5) mammary gland at study completion. (<b>F</b>) Normalized inguinal mammary gland weight. <span class="html-italic">n</span> = 7–8. * <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 2
<p>Diet composition and intervention strategies modulate gut microbiome composition. (<b>A</b>) Shannon diversity index of fecal samples collected after 13 weeks of diet and intervention exposure. (<b>B</b>) Principal component analysis of fecal microbial composition. (<b>C</b>) Proportional abundance of bacterial phyla identified in fecal samples. Each bar represents data collected from one mouse. (<b>D</b>) Proportional abundance of fecal Bacteroidetes. (<b>E</b>) Proportional abundance of fecal Firmicutes. (<b>F</b>) The ratio of Bacteroidetes to Firmicutes. <span class="html-italic">n</span> = 7–8, * <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>Dietary intake and MGE consumption mediate gut colonization of probiotic bacterial species. (<b>A</b>) Proportional abundance of bacterial species were identified in murine feces following 13 weeks of diet and intervention exposure. Each bar represents the fecal bacterial composition of a single mouse. (<b>B</b>–<b>E</b>) Proportional abundance of probiotic bacterial species identified in murine feces. (<b>B</b>) <span class="html-italic">Bifidobacterium</span>. (<b>C</b>) <span class="html-italic">Lactobacillus unclassified</span>. (<b>D</b><span class="html-italic">) Lactobacillus brevis</span>. (<b>E</b>) <span class="html-italic">Lactobacillus plantarum</span>. *** <span class="html-italic">p</span> &lt; 0.001. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 4
<p>Diet and intervention strategies mediate changes in visceral adipose tissue fibrosis. Representative 20× images of visceral adipose tissue stained with Picrosirius Red. Percentage of pixels positive for PicRed staining. * <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 5
<p>Diet and intervention strategies mediate changes in visceral adipose tissue immune cell infiltration. Representative 20× images of visceral adipose tissue stained with anti-F4/80. Number of F4/80-positive macrophages identified per million pixels. * <span class="html-italic">p</span> &lt; 0.05. *** <span class="html-italic">p</span> &lt; 0.001. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 6
<p>Diet and intervention strategies mediate changes in visceral adipose tissue physiology and inflammation. (<b>A</b>) Representative 20× images of visceral adipose tissue stained with anti-MCP-1. (<b>B</b>) Percentage of pixels positive for anti-MCP-1 staining. (<b>C</b>) Average adipocyte diameter calculated from three representative adipocytes per image. * <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 7
<p>Diet and intervention strategies mediate factors associated with intestinal inflammation. (<b>A</b>) Images of intestinal sections stained with H&amp;E and Alcian Blue. (<b>B</b>) Average villus length measured in H&amp;E images. (<b>C</b>) Alcian Blue-positive goblet cells counted per villus. * <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 ">
15 pages, 1928 KiB  
Article
Screening and Activity Analysis of α-Glucosidase Inhibitory Peptides Derived from Coix Seed Prolamins Using Bioinformatics and Molecular Docking
by Zhiming Li, Shu Zhang, Weihong Meng, Jiayu Zhang and Dongjie Zhang
Foods 2023, 12(21), 3970; https://doi.org/10.3390/foods12213970 - 30 Oct 2023
Cited by 1 | Viewed by 1545
Abstract
Hydrolysates of coix seed prolamins (CHPs) have an excellent hypoglycemic effect and can effectively inhibit α-glucosidase, which is the therapeutic target enzyme for type 2 diabetes mellitus. However, its hypoglycemic components and molecular mechanisms remain unclear, and its stability in food processing needs [...] Read more.
Hydrolysates of coix seed prolamins (CHPs) have an excellent hypoglycemic effect and can effectively inhibit α-glucosidase, which is the therapeutic target enzyme for type 2 diabetes mellitus. However, its hypoglycemic components and molecular mechanisms remain unclear, and its stability in food processing needs to be explored. In this study, four potential α-glucosidase inhibitory peptides (LFPSNPLA, FPCNPLV, HLPFNPQ, LLPFYPN) were identified and screened from CHPs using LC-MS/MS and virtual screening techniques. The results of molecular docking showed that the four peptides mainly inhibited α-glucosidase activity through hydrogen bonding and hydrophobic interactions, with Pro and Leu in the peptides playing important roles. In addition, CHPs can maintain good activity under high temperatures (40~100 °C) and weakly acidic or weakly alkaline conditions (pH 6.0~8.0). The addition of glucose (at 100 °C) and NaCl increased the inhibitory activity of α-glucosidase in CHPs. The addition of metal ions significantly decreased the inhibitory activity of α-glucosidase by CHPs, and their effects varied in magnitude with Cu2+ having the largest effect followed by Zn2+, Fe3+, K+, Mg2+, and Ca2+. These results further highlight the potential of CHPs as a foodborne hypoglycemic ingredient, providing a theoretical basis for the application of CHPs in the healthy food industry. Full article
Show Figures

Figure 1

Figure 1
<p>Effects of different proteases on the degree of hydrolysis and the inhibitory activity of α-glucosidase of the CHPs. Different letters Different letters (a–d) indicate significant difference between the mean values of different samples (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Peptides identified in the CHPs-Alc. (<b>A</b>) Peptides number of identified peptides from the parent proteins α-coixin, β-coixin, γ-coixin among all of the identified peptides in the CHPs-Alc; (<b>B</b>) molecular weight distribution of the identified peptides in the CHPs-Alc.</p>
Full article ">Figure 3
<p>Molecular docking results of potential alpha-glucosidase inhibitory peptide with α-glucosidase (2QMJ). (<b>A</b>) LFPSNPLA; (<b>B</b>) FPCNPLV; (<b>C</b>) HLPFNPQ; (<b>D</b>) LLPFYPN.</p>
Full article ">Figure 4
<p>The stability of α-glucosidase inhibitory activity of CHPs-Alc under different food processing conditions: (<b>A</b>) Temperature, (<b>B</b>) pH, (<b>C1</b>) edible sugar was added at 25 °C, (<b>C2</b>) edible sugar was added at 100 °C, (<b>D</b>) NaCl, (<b>E</b>) metal ions. Different letters Different letters (a–e) indicate significant difference between the mean values of different samples (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
15 pages, 333 KiB  
Article
Effects of Indonesian Shortfin Eel (Anguilla bicolor) By-Product Oil Supplementation on HOMA-IR and Lipid Profile in Obese Male Wistar Rats
by Ginna Megawati, Siti Shofiah Syahruddin, Winona Tjandra, Maya Kusumawati, Dewi Marhaeni Diah Herawati, Dida Achmad Gurnida and Ida Musfiroh
Nutrients 2023, 15(18), 3904; https://doi.org/10.3390/nu15183904 - 7 Sep 2023
Cited by 1 | Viewed by 1374
Abstract
The prevalence of people being overweight and obese has increased globally over the past decades. The use of omega-3 fatty acids—a compound usually primarily found in fish oil—has been known to improve the metabolic profile of obese patients. As the demand for eels [...] Read more.
The prevalence of people being overweight and obese has increased globally over the past decades. The use of omega-3 fatty acids—a compound usually primarily found in fish oil—has been known to improve the metabolic profile of obese patients. As the demand for eels increases, the number of waste products from the eels increases and creates environmental problems. This study was conducted to investigate the effect of a newly discovered Indonesian Shortfin eel by-product oil supplementation on the Homeostasis Model Assessment-Estimated Insulin Resistance (HOMA-IR) and lipid profiles of obese male (Lee index ≥ 0.3) Wistar rats (Rattus norvegicus). The oil was extracted from waste products (heads). Fifteen obese rats were divided into three groups and were administered NaCl (C), commercial fish oil (CO), and Indonesian shortfin eel by-product oil (EO). All groups had statistically significant differences in total cholesterol, LDL, and triglyceride levels (p < 0.05). The CO and EO group showed a significant decrease in total cholesterol, LDL, and triglyceride after treatment. However, no significant difference was found in HDL levels and HOMA-IR. The supplementation of Indonesian shortfin eel by-product oil significantly improved lipid profile while effectively mitigating environmental challenges. Full article
17 pages, 2787 KiB  
Protocol
Ketogenic-Mimicking Diet as a Therapeutic Modality for Bipolar Disorder: Biomechanistic Rationale and Protocol for a Pilot Clinical Trial
by Jeffrey L. B. Bohnen, Travis P. Wigstrom, Alexis M. Griggs, Stiven Roytman, Rebecca R. Paalanen, Hailemicael A. Andrews, Nicolaas I. Bohnen, Jacob J. H. Franklin and Melvin G. McInnis
Nutrients 2023, 15(13), 3068; https://doi.org/10.3390/nu15133068 - 7 Jul 2023
Cited by 2 | Viewed by 4244
Abstract
There is growing interest in the investigation of ketogenic diets as a potential therapy for bipolar disorder. The overlapping pharmacotherapies utilized for both bipolar disorder and seizures suggest that a mechanistic overlap may exist between these conditions, with fasting and the ketogenic diet [...] Read more.
There is growing interest in the investigation of ketogenic diets as a potential therapy for bipolar disorder. The overlapping pharmacotherapies utilized for both bipolar disorder and seizures suggest that a mechanistic overlap may exist between these conditions, with fasting and the ketogenic diet representing the most time-proven therapies for seizure control. Recently, preliminary evidence has begun to emerge supporting a potential role for ketogenic diets in treating bipolar disorder. Notably, some patients may struggle to initiate a strict diet in the midst of a mood episode or significant life stressors. The key question addressed by this pilot clinical trial protocol is if benefits can be achieved with a less restrictive diet, as this would allow such an intervention to be accessible for more patients. Recent development of so-called ketone esters, that once ingested is converted to natural ketone bodies, combined with low glycemic index dietary changes has the potential to mimic two foundational components of therapeutic ketosis: high levels of ketones and minimal spiking of glucose/insulin. This pilot clinical trial protocol thus aims to investigate the effect of a ‘ketogenic-mimicking diet’ (combining supplementation of ketone esters with a low glycemic index dietary intervention) on neural network stability, mood, and biomarker outcomes in the setting of bipolar disorder. Positive findings obtained via this pilot clinical trial protocol may support future target engagement studies of ketogenic-mimicking diets or related ketogenic interventions. A lack of positive findings, in contrast, may justify a focus on more strict dietary interventions for future research. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Protocol Flowchart: Visual Overview.</p>
Full article ">Figure 2
<p>Key Outcomes: Visual Overview.</p>
Full article ">Figure 3
<p>Neuronal Mechanisms Implicated in Bipolar Disorder: Hypothesized Conceptual Model. (<b>A</b>) Typical neuronal model as baseline comparison. With proper homeostatic regulation of electrochemical gradients, neuronal excitability is optimized. (<b>B</b>) Neuronal model with intracellular sodium accumulation secondary to secondary to Na<sup>+</sup>/K<sup>+</sup> ATPase hypofunctionality. In turn, this would be expected to raise neuronal membrane potential, resulting in hyperexcitability and inappropriate target engagement (i.e., excessive neurotransmitter release from vesicles)—thus, a working mechanistic model applicable to (hypo)manic episodes. Interestingly, individuals with bipolar disorder have been observed to exhibit decreased Na<sup>+</sup>/K<sup>+</sup> ATPase activity and intracellular sodium accumulation in a mood-related pattern [<a href="#B27-nutrients-15-03068" class="html-bibr">27</a>]. Moreover, stimulants and antidepressants would be capable of converging on synaptic hyperexcitability mechanisms and are widely known to trigger (hypo)mania in susceptible individuals. (<b>C</b>) Neuronal model illustrating synaptic vesicle depletion as a potential downstream consequence of hyperexcitability and inappropriate target engagement. Synaptic vesicle depletion may result in phasic hypoactive synaptic signaling until homeostasis is restored. This provides a working mechanistic model applicable to depressive episodes that helps account for the temporal relationship commonly observed between (hypo)manic and depressive episodes.</p>
Full article ">Figure 4
<p>Conceptual illustration of how resting membrane potential may be altered as a function of metabolic impairment and resultant Na<sup>+</sup>/K<sup>+</sup> ATPase hypofunctionality. With increasing severity of metabolic impairment, resting membrane potential may rise secondary to Na<sup>+</sup>/K<sup>+</sup> ATPase hypofunctionality. In turn, this may alter neuronal excitability. As a point of reference that speaks to the delicate nature of neuronal membrane homeostasis, complete inactivation of the Na<sup>+</sup>/K<sup>+</sup> ATPase would result in a resting membrane potential that is approximately 10 mV less electronegative compared to baseline [<a href="#B59-nutrients-15-03068" class="html-bibr">59</a>].</p>
Full article ">Figure 5
<p>Metabolic Dysfunction in Context: Biopsychosocial Model. Importantly, metabolic dysregulation should be conceptualized within the broader context of the biopsychosocial model. There are many upstream and downstream factors integrated with metabolism, including lifestyle factors (such as nutrition, exercise, and sleep), medical comorbidities, medications, substance use, trauma, and chronic stress. Accumulating evidence supports the critical role of mitochondrial function in bipolar illness, which may help explain how altered neuronal excitability represents just one component of a broader model connecting biological and metabolic mechanisms with psychological and social risk factors [<a href="#B60-nutrients-15-03068" class="html-bibr">60</a>]. As a pertinent example, stress and trauma have been linked to alterations in cortisol signaling and mitochondrial function [<a href="#B61-nutrients-15-03068" class="html-bibr">61</a>]. Moreover, this figure illustrates a critical link between altered neuronal excitability and altered patterns of neural network functional connectivity, as patterns of spreading activation throughout neural networks are inherently dependent on patterns of neuronal excitability. The foundation of the image depicts how these mechanisms may ultimately culminate in suffering experienced by the patients we serve—something that should not be overlooked for the sake of mechanistic granularity.</p>
Full article ">
13 pages, 6764 KiB  
Article
Preparation of Red Ginseng Marc-Derived Gintonin and Its Application as a Skin Nutrient
by Rami Lee, Ji-Hun Kim, Hongik Hwang, Hyewhon Rhim, Sung-Hee Hwang, Ik-Hyun Cho, Do-Geun Kim, Hyoung-Chun Kim and Seung-Yeol Nah
Nutrients 2023, 15(11), 2574; https://doi.org/10.3390/nu15112574 - 31 May 2023
Cited by 5 | Viewed by 1979
Abstract
Ginseng is one of the traditional herbal medicines for tonic. Gintonin is a new material derived from white/red ginseng and its lysophosphatidic acids (LPAs) play as a ligand for G protein-coupled LPA receptors. Korean red ginseng marc (KRGM) is a by-product after the [...] Read more.
Ginseng is one of the traditional herbal medicines for tonic. Gintonin is a new material derived from white/red ginseng and its lysophosphatidic acids (LPAs) play as a ligand for G protein-coupled LPA receptors. Korean red ginseng marc (KRGM) is a by-product after the KRG processes. We developed a low-cost/high-efficiency method for KRGM gintonin production. We further studied the KRGM gintonin-mediated anti-skin aging effects under UVB exposure using human dermal fibroblasts (HDFs). KRGM gintonin yield is about 8%. KRGM gintonin contains a high amount of LPA C18:2, lysophosphatidylcholine (LPC), and phosphatidylcholine (PC), which is similar to white ginseng gintonin. KRGM gintonin induced [Ca2+]i transient via LPA1/3 receptors and increased cell viability/proliferation under UVB exposure. The underlying mechanisms of these results are associated with the antioxidant action of KRGM gintonin. KRGM gintonin attenuated UVB-induced cell senescence by inhibiting cellular β-galactosidase overexpression and facilitated wound healing. These results indicate that KRGM can be a novel bioresource of KRGM gintonin, which can be industrially utilized as new material for skin nutrition and/or skin healthcare. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic methods for the preparation of KRGM gintonin. The method uses only edible ethanol and water. KRG extraction first takes place with distilled water for 12 h, 4 to 7 times continuously, to produce KRG extract, which is further processed for KRG product productions, leaving KRGM as a leftover. The KRGM is dried, and then the dry KRGM was refluxed with 70% ethanol for 8 h at 80 °C three times; the air-dried or freeze-dried ethanol extract is about 8% yield of solid content. The resulting powder obtained from the air-dry process was designated KRGM gintonin.</p>
Full article ">Figure 2
<p>Effects of KRGM gintonin (GT) on [Ca<sup>2+</sup>]<sub>i</sub> transient in HDFs. (<b>A</b>) Comparison of [Ca<sup>2+</sup>]<sub>i</sub> transient in human dermal fibroblasts (HDFs) by KRGM gintonin. Treatment concentrations of KRGM gintonin are 0, 1, 3, or 10 μg/mL. (<b>B</b>) Histograms of [Ca<sup>2+</sup>]<sub>i</sub> transient in HDF cells induced by KRGM gintonin at three representative concentrations (0, 1, 3, and 10 μg/mL). [Ca<sup>2+</sup>]<sub>i</sub> transient was elicited in a dose-dependent manner. The strongest [Ca<sup>2+</sup>]<sub>i</sub> transient was exhibited at 10 μg/mL. (<b>C</b>) Inhibitory effect of [Ca<sup>2+</sup>]<sub>i</sub> transient in HDFs induced by KRGM gintonin by Ki16425 (10 μM) after KRGM 10 μg/mL treatment. * <span class="html-italic">p</span> &lt; 0.05, compared with baseline (0 μg/mL); *** <span class="html-italic">p</span> &lt; 0.05, compared with 10 μg/mL treatment group. Statistical analysis was conducted by <span class="html-italic">t</span>-test. Each graph shows the mean ± SEM of three independent experiments.</p>
Full article ">Figure 3
<p>Cell cytotoxicity and cell protection assay by KRGM gintonin in HDFs. HDFs were seeded in a 96-well plate at 2 × 10<sup>4</sup> cells/well density for assay. (<b>A</b>) Cell viability assay using KRGM gintonin (GT) at 24 h. Each treatment group with indicated doses exhibited a significant increase. * <span class="html-italic">p</span> &lt; 0.05, compared with the untreated group. (<b>B</b>–<b>D</b>) Cell viability assay using KRGM gintonin at 24 h, 48 h, and 72 h under ultraviolet (UV) damage at a power of 50 mJ/cm<sup>2</sup>. HDFs were treated with KRGM gintonin. (<b>B</b>) KRGM gintonin treatment increased cell viability at a concentration of 1 and 3 μg/mL under UVB exposure at 24 h. (<b>C</b>) KRGM gintonin treatment increased cell viability in a dose-dependent manner from 0.1 to 10 μg/mL under UVB exposure at 48 h. (<b>D</b>) KRGM gintonin treatment increased cell viability at every dose under UVB exposure at 72 h. * <span class="html-italic">p</span> &lt; 0.05, compared with the control group. ** <span class="html-italic">p</span> &lt; 0.01, compared with untreated group. Each graph shows the mean ± SEM of three independent experiments. UT; untreated control group, Ki; Ki16425 10 μM treatment group.</p>
Full article ">Figure 4
<p>Antioxidant effect of KRGM gintonin (GT) and inhibition of NO production by KRGM gintonin (GT). (<b>A</b>) KRGM gintonin effects on ABTS<sup>+</sup> radical scavenging activity. ABTS<sup>+</sup> radical scavenging activity was measured using KRGM gintonin. KRGM gintonin showed the strongest radical scavenging activity in the 10 mg/mL group. The ABTS<sup>+</sup> radical scavenging effect of KRGM gintonin increased in a dose-dependent manner. Each bar indicates the mean ± S.D. (<span class="html-italic">n</span> = 4). Each graph shows the mean ± SD of four independent experiments. (<b>B</b>–<b>D</b>) HDFs were seeded in a 96-well plate at a 1 × 10<sup>4</sup> cells/well density for assay. HDFs were treated with KRGM gintonin. (<b>B</b>) Every dose of KRGM gintonin (from 0.1 to 10 μg/mL) showed a significant decrease in NO production at 24 h. * <span class="html-italic">p</span> &lt; 0.05, compared with the untreated control group). (<b>C</b>) Every dose of KRGM gintonin (from 0.1 to 10 μg/mL) showed a significant decrease at 48 h, showing the most decrease in NO production at 30 μg/mL. * <span class="html-italic">p</span> &lt; 0.05, compared with the untreated control group). (<b>D</b>) Every dose of KRGM gintonin (from 0.1 to 10 μg/mL) showed a significant decrease at 72 h, showing the most decrease in NO production at 30 μg/mL. * <span class="html-italic">p</span> &lt; 0.05, compared with the untreated control group). Each NO produced was shown as the concentration of NO vs. cell number ratio (μM). Each graph shows the mean ± SD of three independent experiments. UT; untreated control group, LPA; LPA treatment group (10 μM), GT; KRGM gintonin treated group.</p>
Full article ">Figure 5
<p>KRGM gintonin (GT) effect on reactive oxygen species (ROS) production in UVB-damaged HDFs. HDFs were seeded in an 8-well chamber at a 1 × 10<sup>5</sup> cells/well density for assay. (<b>A</b>) HDFs treated with/or without KRGM gintonin (10 μg/mL) or LPA (10 μM). (<b>B</b>) Protective effect of KRGM gintonin against UVB damage, which generates ROS. UVB irradiation caused HDFs damage, co-treatment of KRGM gintonin protected cells against UVB irradiation, and LPA as a positive control showed a cell-protective effect. * <span class="html-italic">p</span> &lt; 0.05, compared with non-treatment of UVB. ** <span class="html-italic">p</span> &lt; 0.001, compared with non-treatment of UVB. Each graph shows the mean ± SD of three independent experiments. UT; untreated control, LPA; LPA treated control (10 μM), UV; UVB 50 mJ/cm<sup>2</sup>, GT; KRGM gintonin treated group.</p>
Full article ">Figure 6
<p>Effects of KRGM gintonin (GT) on cellular senescence and on scratch wound healing. (<b>A</b>) SA-β–galactosidase assay in HDFs upon KRGM gintonin treatment. Untreated control, UVB 50 mJ/cm<sup>2</sup> treated, UVB 50 mJ/cm<sup>2</sup> + KRGM gintonin (0.3 μg/mL), UVB 50 mJ/cm<sup>2</sup> + KRGM gintonin (1 μg/mL), UVB 50 mJ/cm<sup>2</sup> + KRGM gintonin (3 μg/mL), and UVB 50 mJ/cm<sup>2</sup> + KRGM gintonin (10 μg/mL). The histograms are depicted as a percentage of the β–galactosidase positive cells among total cells. β–galactosidase positive cells are relatively low in the UVB-untreated group but high under UVB exposure. The restoring effects from UVB damage were observed at 1–3 μg/mL of KRGM gintonin. * <span class="html-italic">p</span> &lt; 0.05, compared with the control group. Each graph shows the mean ± SEM of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.001, compared with the control group. Each graph shows the mean ± SEM of three independent experiments. (<b>B</b>) Effects of gintonin on scratch wound healing of HDFs. After the serum was removed for 24 h, cells were scratched with a micropipette tip and then incubated with a serum-free medium for another 24 h containing KRGM gintonin (0.3–30 μg/mL) and lysophosphatidic acid (LPA, 10 μM) as a positive reference agent. Images were obtained by taking photos at 0 h and 24 h post-treatment with KRGM gintonin or LPA. Representative images of control cells, cells treated with KRGM gintonin (0.3 and 10 μg/mL), and LPA (10 μM). The scale bar is equivalent to 200 μm. LPA was used as reference material for a positive control. Wound closing response of untreated cells (UT) was calculated as 100%. Data are presented as the means ± S.E.M. (<span class="html-italic">n</span> = 9); * <span class="html-italic">p</span> &lt; 0.05 vs. control, ** <span class="html-italic">p</span> &lt; 0.01 vs. control. UT; untreated control, LPA; LPA treated group (10 μM), GT; KRGM gintonin treated group.</p>
Full article ">
15 pages, 984 KiB  
Review
Use of Dietary Fibers in Reducing the Risk of Several Cancer Types: An Umbrella Review
by Jun Hu, Junjing Wang, Yuxing Li, Kun Xue and Juntao Kan
Nutrients 2023, 15(11), 2545; https://doi.org/10.3390/nu15112545 - 30 May 2023
Cited by 14 | Viewed by 3854
Abstract
(1) Background: Numerous meta-analyses have shown that a high intake of dietary fiber plays a protective role in preventing the development of various types of cancer. However, previous studies have been limited by focusing on a single type of dietary fiber and variations [...] Read more.
(1) Background: Numerous meta-analyses have shown that a high intake of dietary fiber plays a protective role in preventing the development of various types of cancer. However, previous studies have been limited by focusing on a single type of dietary fiber and variations in outcome measures, which may not be effectively applied to provide dietary guidance for the general population. (2) Object: We summarized the meta-analysis of dietary fiber and cancer, and provided references for residents to prevent cancer. (3) Methods: Systematic search of relevant meta-analyses on the association between dietary fiber and cancer occurrence in PubMed, Web of Science and other databases was conducted from the time of database construction to February 2023. The method logical and evidence quality assessments were performed by applying the criteria in the “A Measurement Tool to Assess Systematic Reviews-2” (AMSTAR2) scale and the World Cancer Research Fund/American Institute for Cancer Research (WCRF/AICR) Expert Report, respectively. (4) Results: Our analysis included 11 meta-analyses, and the AMSTAR 2 assessment revealed that the overall methodological quality was suboptimal, with two key items lacking sufficient information. Nonetheless, our findings indicate that a high intake of dietary fiber is associated with a reduced risk of several types of cancer, including esophageal, gastric, colon, rectal, colorectal adenoma, breast, endometrial, ovarian, renal cell, prostate, and pancreatic cancers. The majority of these associations were supported by a “probable” level of evidence. (5) Conclusions: Dietary fiber intake has different protective effects on different cancers. Full article
Show Figures

Figure 1

Figure 1
<p>Flow diagram of the enrollment of systematic reviews/meta-analyses of the association of dietary fiber with cancer.</p>
Full article ">Figure 2
<p>Level of evidence for the association between dietary fiber and cancer.</p>
Full article ">
25 pages, 6153 KiB  
Article
Biomolecules of Fermented Tropical Fruits and Fermenting Microbes as Regulators of Human Hair Loss, Hair Quality, and Scalp Microbiota
by Wolfgang Mayer, Michaela Weibel, Chiara De Luca, Galina Ibragimova, Ilya Trakhtman, Zaira Kharaeva, Danny L. Chandler and Liudmila Korkina
Biomolecules 2023, 13(4), 699; https://doi.org/10.3390/biom13040699 - 20 Apr 2023
Cited by 2 | Viewed by 4022
Abstract
Plant-derived secondary metabolites (polyphenols/terpenes/alkaloids) and microbial exometabolites/membrane components of fermented tropical fruits are known as highly bioavailable biomolecules causing skin and hair improvement effects (wound healing, anti-inflammatory, antioxidant, antidiabetic, antiacne, skin/hair microbiota balancing, hair growth-promoting, and hair loss-inhibiting). Caffein is considered as a [...] Read more.
Plant-derived secondary metabolites (polyphenols/terpenes/alkaloids) and microbial exometabolites/membrane components of fermented tropical fruits are known as highly bioavailable biomolecules causing skin and hair improvement effects (wound healing, anti-inflammatory, antioxidant, antidiabetic, antiacne, skin/hair microbiota balancing, hair growth-promoting, and hair loss-inhibiting). Caffein is considered as a hair growth promoter. A randomized placebo- and caffein-controlled clinical trial on the efficacy of fermented papaya (FP) plus fermented mangosteen (FM) towards human hair quality and loss was conducted. Shampoo and lotion hair care products containing FP, FM, and caffein as active agents were developed and applied to 154 subjects of both sexes with clinically confirmed androgenic or diffuse alopecia for 3 months. Their clinical efficacy was assessed subjectively by questionnaires filled in by dermatologists/trichologists, and by the objective trichomicroscopical calculations. Hair and scalp skin quality was determined by microbiota pattern and ATP, SH-groups, protein, and malonyl dialdehyde quantification. Comparative clinical data showed that the experimental hair care cosmetics significantly inhibited hair loss, increased hair density/thickness, and improved hair follicle structure versus placebo and caffein controls. The cosmetics with FP and FM substantially normalized the microbiota pattern and increased ATP content in hair follicle, while inhibiting lipid peroxidation in the scalp skin, and SH-group formation in the hair shaft. Full article
Show Figures

Figure 1

Figure 1
<p>Determination of hair density. One cm<sup>2</sup> is bordered by the orange line. Magnification ×60.</p>
Full article ">Figure 2
<p>The determination of a single hair diameter was conducted under ×200 magnification lenses. The numbers in yellow squares indicate the hair diameter in μm.</p>
Full article ">Figure 3
<p>Determination of the hair follicle diameter and lipid/water mantel conditions: (<b>a</b>) hair root/follicle; (<b>b</b>) lipid mantel and hydration conditions were performed under ×200 magnification.</p>
Full article ">Figure 4
<p>Preparation of hair follicles for the ATP measurements in the hair follicles: (<b>a</b>) hair plucking; (<b>b</b>) separated hair follicles.</p>
Full article ">Figure 5
<p>Hair diameter (μm) distribution before and after the trial: (<b>a</b>) Experimental Group 1; (<b>b</b>) Placebo Control Group 2; (<b>c</b>) Caffeine Control Group 3. Small boxes—median values; large boxes—median values ± standard error of median (SEM); whiskers—median values ± 1.96 SEM. <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 6
<p>Hair density (<span class="html-italic">n</span>/cm<sup>2</sup>) distribution before and after the trial: (<b>a</b>) Experimental Group 1; (<b>b</b>) Placebo Control Group 2; (<b>c</b>) Caffeine Control Group 3. Small boxes—median values; large boxes—median values ± standard error of median (SEM); whiskers—median values ± 1.96 SEM. <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>Thick hair presence (%) before and after the trial: (<b>a</b>) Experimental Group 1; (<b>b</b>) Placebo Control Group 2; (<b>c</b>) Caffeine Control Group 3. Small boxes—median values; large boxes—median values ± standard error of median (SEM); whiskers—median values ± 1.96 SEM. <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 8
<p>Medium hair presence (%) before and after the trial: (<b>a</b>) Experimental Group 1; (<b>b</b>) Placebo Control Group 2; (<b>c</b>) Caffeine Control Group 3. Small boxes—median values; large boxes—median values ± standard error of median (SEM); whiskers—median values ± 1.96 SEM. <span class="html-italic">p</span>&lt; 0.05.</p>
Full article ">Figure 9
<p>Thin hair presence (%) before and after the trial: (<b>a</b>) Experimental Group 1; (<b>b</b>) Placebo Control Group 2; (<b>c</b>) Caffeine Control Group 3. Small boxes—median values; large boxes—median values ± standard error of median (SEM); whiskers—median values ± 1.96 SEM. <span class="html-italic">p</span> &lt; 0.05, <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 10
<p>Relationship between ATP content in hair follicles and labile protein leakage from the hair shaft in patients with two types of alopecia (<span class="html-italic">n</span> = 154). Blue dots represent values for the individual person. Red line is a correlation curve.</p>
Full article ">Figure 11
<p>Sources of oxidative stress in different types of alopecia.</p>
Full article ">Figure A1
<p>Microphotographs of hair density before and after the trial in the patient K-va of the experimental Group 1. (<b>a</b>) Hair density on the frontal area before the trial; (<b>b</b>) hair density on the frontal area after the trial; (<b>c</b>) hair density on the occipital area before the trial; (<b>d</b>) hair density on the occipital area after the trial.</p>
Full article ">Figure A1 Cont.
<p>Microphotographs of hair density before and after the trial in the patient K-va of the experimental Group 1. (<b>a</b>) Hair density on the frontal area before the trial; (<b>b</b>) hair density on the frontal area after the trial; (<b>c</b>) hair density on the occipital area before the trial; (<b>d</b>) hair density on the occipital area after the trial.</p>
Full article ">Figure A2
<p>Microphotographs of hair density before and after the trial in the patient Ko-va of the placebo control Group 2. (<b>a</b>) Hair density on the frontal area before the trial; (<b>b</b>) hair density on the frontal area after the trial; (<b>c</b>) hair density on the occipital area before the trial; (<b>d</b>) hair density on the occipital area after the trial.</p>
Full article ">Figure A3
<p>Microphotographs of hair density before and after the trial in the patient I-va of the caffeine control Group 3. (<b>a</b>) Hair density on the frontal area before the trial; (<b>b</b>) hair density on the frontal area after the trial; (<b>c</b>) hair density on the occipital area before the trial; (<b>d</b>) hair density on the occipital area after the trial.</p>
Full article ">
11 pages, 1930 KiB  
Article
Shikonin Binds and Represses PPARγ Activity by Releasing Coactivators and Modulating Histone Methylation Codes
by Ui-Hyun Park, HyeSook Youn, Eun-Joo Kim and Soo-Jong Um
Nutrients 2023, 15(7), 1797; https://doi.org/10.3390/nu15071797 - 6 Apr 2023
Cited by 2 | Viewed by 1650
Abstract
Shikonin, a natural ingredient produced by Lithospermum erythrorhizon, has anti-inflammatory, anti-cancer, and anti-obesity effects. It also inhibits adipocyte differentiation; however, the underlying molecular and epigenetic mechanisms remain unclear. We performed RNA-sequencing of shikonin-treated 3T3-L1 cells. Gene ontology and gene set enrichment analysis [...] Read more.
Shikonin, a natural ingredient produced by Lithospermum erythrorhizon, has anti-inflammatory, anti-cancer, and anti-obesity effects. It also inhibits adipocyte differentiation; however, the underlying molecular and epigenetic mechanisms remain unclear. We performed RNA-sequencing of shikonin-treated 3T3-L1 cells. Gene ontology and gene set enrichment analysis showed that shikonin is significantly associated with genes related to adipogenesis, histone modification, and PPARγ. Shikonin treatment downregulated the mRNA expression of PPARγ-responsive genes and rosiglitazone-induced transcriptional activity of PPARγ. Microscale thermophoresis assays showed a KD value 1.4 ± 0.13 μM for binding between shikonin and PPARγ. Glutathione S-transferase pull-down assays exhibited that shikonin blocked the rosiglitazone-dependent association of PPARγ with its coactivator CBP. In addition, shikonin decreased the enrichment of the active histone code H3K4me3 and increased the repressive code H3K27me3 of PPARγ target promoters. Shikonin is a PPARγ antagonist that suppresses adipogenesis by regulating the enrichment of histone codes during adipogenesis. Therefore, it may be used to treat obesity-related disorders via epigenetic changes. Full article
Show Figures

Figure 1

Figure 1
<p>Genome-wide RNA-seq analysis. (<b>A</b>) Clustering analysis of 2406 genes with 1.5-fold changes following shikonin treatment. (<b>B</b>) Gene Ontology (GO) analysis of genes with 1.5-fold changes. (<b>C</b>–<b>E</b>) Bedgraph analysis of three adipogenic genes downregulated by shikonin treatment. Enrichment of tag fragments from RNA-seq results are shown in the gene bodies of <span class="html-italic">Fabp4</span> (<b>C</b>), <span class="html-italic">Adipoq</span> (<b>D</b>), and <span class="html-italic">Lpl</span> (<b>E</b>).</p>
Full article ">Figure 2
<p>Genome-wide RNA-seq analysis of shikonin-regulated genes. (<b>A</b>) Gene sets significantly associated with PPARγ were targeted through GSEA, and the normalized enrichment score (NES) was measured. Gene sets with a nominal <span class="html-italic">p</span>-value of &lt;0.05 and false discovery rate (FDR) <span class="html-italic">q</span>-value of &lt;0.25 were considered significantly enriched. (<b>B</b>) Scatter plot analysis of selected gene sets. In total, 13,965 significant genes (log2 value of mRNA expression &gt; 4.0 in RNA-seq of DMSO or shikonin) are displayed in gray. Among them, 2406 genes with a 1.5-fold change in expression level are shown in green. The distribution of genes associated with the indicated GO terms are displayed by brown, blue, and red spots. (<b>C</b>) Effects of shikonin on the mRNA expression of five PPARγ response genes. 3T3-L1 cells were differentiated and treated with DMSO or shikonin, then their transcript expressions were measured by RT-qPCR and normalized to GAPDH. Results are presented as the relative expression compared to DMSO controls. Values are represented as the means ± SDs from three independent experiments (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Antagonistic effects of shikonin on PPARγ regulation. (<b>A</b>) Results of the luciferase reporter gene assay. HEK293 cells were cotransfected as described in the Materials and Method section and treated with rosiglitazone (Rosi, 0.5 μM) and the indicated concentrations of shikonin. Luciferase values were normalized to the β-galactosidase activity. The error bars represent means ± SDs of three independent experiments (** <span class="html-italic">p</span> &lt; 0.01). (<b>B</b>) Effects of shikonin on Rosi-induced expression of the <span class="html-italic">Lpl</span> gene. 3T3-L1 cells were differentiated and treated with 1 μM of Rosi and the indicated concentrations of shikonin for 6 days. Expression of mRNA, measured by RT-qPCR, was normalized to the GAPDH expression level and indicated as fold change relative to that of the DMSO control. Bars represent means ± SDs of three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01). (<b>C</b>) Microscale thermophoresis (MST) assays were performed as described in the Materials and Methods section. (<b>D</b>) Effects of shikonin on the rosiglitazone-induced interaction of PPARγ with the coactivator CBP. GST pull-down assays were performed as described in the Materials and Methods section. The immobilized GST-CBP (amino acids 1–460 or 602–1095) fusion protein was incubated with the indicated concentration of shikonin in the presence of 1 μM of Rosi and a His-PPARγ ligand-binding domain (LBD). Bound proteins were visualized by western blotting using an anti-His antibody. The input represents 5% of His-PPARγ used for the binding assays.</p>
Full article ">Figure 4
<p>Epigenetic regulation of PPARγ target genes by shikonin. Eight days after adipogenesis, 3T3-L1 cells were fixed and harvested for ChIP assays using the indicated antibodies against H3H4me3 and H3K27me3. Promoter occupancy was determined using qPCR and primer sets on the targeted promoters of three genes (<span class="html-italic">Hs11b1</span>, <span class="html-italic">Acsl1</span>, and <span class="html-italic">Retn</span>). Data are represented as means ± SDs for three independent experiments (* <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">
25 pages, 2369 KiB  
Review
Biomarkers of Micronutrients and Phytonutrients and Their Application in Epidemiological Studies
by Jianheng Zheng, Feng Wu, Feijie Wang, Junrui Cheng, Hong Zou, Yuan Li, Jun Du and Juntao Kan
Nutrients 2023, 15(4), 970; https://doi.org/10.3390/nu15040970 - 15 Feb 2023
Cited by 4 | Viewed by 2631
Abstract
Nutritional biomarkers can be used as important indicators of nutritional status and play crucial roles in the prevention as well as prognosis optimization of various metabolism-related diseases. Measuring dietary with the deployment of biomarker assessments provides quantitative nutritional information that can better predict [...] Read more.
Nutritional biomarkers can be used as important indicators of nutritional status and play crucial roles in the prevention as well as prognosis optimization of various metabolism-related diseases. Measuring dietary with the deployment of biomarker assessments provides quantitative nutritional information that can better predict the health outcomes. With the increased availability of nutritional biomarkers and the development of assessment tools, the specificity and sensitivity of nutritional biomarkers have been greatly improved. This enables efficient disease surveillance in nutrition research. A wide range of biomarkers have been used in different types of studies, including clinical trials, observational studies, and qualitative studies, to reflect the relationship between diet and health. Through a comprehensive literature search, we reviewed the well-established nutritional biomarkers of vitamins, minerals, and phytonutrients, and their association with epidemiological studies, to better understand the role of nutrition in health and disease. Full article
Show Figures

Figure 1

Figure 1
<p>An overview of the impact of vitamin A, B complex, and D on cellular biochemistry. (Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>) CoA: Coenzyme A; DHF: Dihydrofolate; DMT: DNA methyltransferase; Hcy: Homocysteine; MAT: Methionine adenosyl transferase; MS: Methionine synthase; MTHFR: Methyltetrahydro-folate reductase; PCAF: p300/CBP-associated factor; SAH:S-adenosyl homocysteine; SAHH: S-adenosyl homocysteine hydrolase; THF: Tetrahydrofolate; TS: Thymidylate synthase; VDR: Vitamin D receptor.</p>
Full article ">Figure 2
<p>An overview of the iodine biomarkers and related metabolism pathway. (Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">Figure 3
<p>Brief depiction of the systemic iron metabolism and potential iron biomarkers. (Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>). BMP6: Bone morphogenetic protein 6; BMPR: Bone morphogenetic protein receptor; DMT1: Divalent metal transporter 1; Fe: Iron/Ferrum; FPN: Ferroportin; HJV: Hemojuvelin; RBC: Red blood cell; SIRPα: Signal regulatory protein alpha; SMAD: Small-body-size mothers against decapentaplegic homolog 1; TSAT: Percentage transferrin saturation.</p>
Full article ">Figure 4
<p>Epidemiological application of biomarkers associated with zinc homeostasis. (Created with <a href="http://BioRender.com" target="_blank">BioRender.com</a>).</p>
Full article ">
23 pages, 6050 KiB  
Article
Ameliorative Effect of Mannuronate Oligosaccharides on Hyperuricemic Mice via Promoting Uric Acid Excretion and Modulating Gut Microbiota
by Biqian Wei, Pengfei Ren, Ruzhen Yang, Yuan Gao, Qingjuan Tang, Changhu Xue and Yuming Wang
Nutrients 2023, 15(2), 417; https://doi.org/10.3390/nu15020417 - 13 Jan 2023
Cited by 7 | Viewed by 2807
Abstract
Mannuronate oligosaccharide (MOS) is α-D-mannuronic acid polymer with 1,4-glycosidic linkages that possesses beneficial biological properties. The aim of this study was to investigate the hypouricemic effect of MOS in hyperuricemic mice and demonstrate the possible protective mechanisms involved. In this research, 200 mg/kg/day [...] Read more.
Mannuronate oligosaccharide (MOS) is α-D-mannuronic acid polymer with 1,4-glycosidic linkages that possesses beneficial biological properties. The aim of this study was to investigate the hypouricemic effect of MOS in hyperuricemic mice and demonstrate the possible protective mechanisms involved. In this research, 200 mg/kg/day of MOS was orally administered to hyperuricemic mice for four weeks. The results showed that the MOS treatment significantly reduced the serum uric acid (SUA) level from 176.4 ± 7.9 μmol/L to 135.7 ± 10.9 μmol/L (p < 0.05). MOS alleviated the inflammatory response in the kidney. Moreover, MOS promoted uric acid excretion by regulating the protein levels of renal GLUT9, URAT1 and intestinal GLUT9, ABCG2. MOS modulated the gut microbiota in hyperuricemic mice and decreased the levels of Tyzzerella. In addition, research using antibiotic-induced pseudo-sterile mice demonstrated that the gut microbiota played a crucial role in reducing elevated serum uric acid of MOS in mice. In conclusion, MOS may be a potential candidate for alleviating HUA symptoms and regulating gut microbiota. Full article
Show Figures

Figure 1

Figure 1
<p>Experimental designs.</p>
Full article ">Figure 2
<p>Ameliorative effect of MOS on HUA in mice. Body weight (<b>A</b>); Weight gain in mice during modeling (<b>B</b>); Serum UA (<b>C</b>); Serum BUN (<b>D</b>). ## <span class="html-italic">p</span> &lt; 0.01, and #### <span class="html-italic">p</span> &lt; 0.0001 versus normal. ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001 versus model. The ‘ns’ indicates no statistical difference.</p>
Full article ">Figure 3
<p>The effect of MOS on uric acid synthesis in HUA mice. Serum XOD activity (<b>A</b>); Serum ADA activity (<b>B</b>); Liver XOD activity (<b>C</b>); Liver ADA activity (<b>D</b>); mRNA levels of liver XOD (<b>E</b>); mRNA levels of liver ADA (<b>F</b>). # <span class="html-italic">p</span> &lt; 0.05, and ## <span class="html-italic">p</span> &lt; 0.01 versus normal. * <span class="html-italic">p</span> &lt; 0.05, and ** <span class="html-italic">p</span> &lt; 0.01 versus model.</p>
Full article ">Figure 4
<p>The ameliorative benefits of MOS on renal injuries. Renal H&amp;E-stained sections with a magnification of 200× (<b>A</b>); IL1β (<b>B</b>); IL-12 (<b>C</b>); IL-18 (<b>D</b>). Green arrow: renal tubular dilatation, red arrow: inflammatory cell infiltration, black arrow: epithelial cell detachment, blue arrow: attenuated inflammatory cell infiltration. #### <span class="html-italic">p</span> &lt; 0.0001 versus normal. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 versus model.</p>
Full article ">Figure 5
<p>Improvement of uric acid excretion by MOS. Renal URAT1 and GLUT9 protein expression (<b>A</b>); Quantitative analysis of renal URAT1 and GLUT9 protein (<b>B</b>); Intestinal ABCG2, GLUT9 protein expression (<b>C</b>); Quantitative analysis of intestinal ABCG2, GLUT9 protein (<b>D</b>). # <span class="html-italic">p</span> &lt; 0.05 versus normal. * <span class="html-italic">p</span> &lt; 0.05 versus model.</p>
Full article ">Figure 6
<p>Alterations in intestinal flora structure and composition under MOS treatment. NMDS analysis (<b>A</b>); Venn diagram of ASVs (<b>B</b>); Changes in gut flora structure at the gate level (<b>C</b>); The Bacteroidetes/Firmicutes ratio (<b>D</b>); Relative abundance at the genus level shown as a heat map (<b>E</b>). N: Normal group, M: Model group. # <span class="html-italic">p</span> &lt; 0.05 versus normal. * <span class="html-italic">p</span> &lt; 0.05 versus model. N: Normal group, M: Model group.</p>
Full article ">Figure 7
<p>DEseq2 analysis of differential species. N vs. M (<b>A</b>); MOS vs. M (<b>B</b>). All data shown are consistent with <span class="html-italic">p</span> &lt; 0.05 and a Fold change &gt; 2. N: Normal group, M: Model group.</p>
Full article ">Figure 8
<p>Changes in metabolic parameters of HUA mice correlate with gut microbiota. Spearman’s correlation analysis between intestinal flora and metabolic parameters (<b>A</b>); The Random Forest (<b>B</b>); Abundance of <span class="html-italic">Tyzzerella</span> (<b>C</b>). In plot A, * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. In plot C, ### <span class="html-italic">p</span> &lt; 0.001 versus normal, *** <span class="html-italic">p</span> &lt; 0.001 versus model. N: Normal group, M: Model group.</p>
Full article ">Figure 9
<p>Effect of MOS treatment on SCFAs. Total SCFAs content (<b>A</b>); Acetic acid (<b>B</b>); Propionic acid (<b>C</b>); Isobutyric acid (<b>D</b>); Butyric acid (<b>E</b>); Isovaleric acid (<b>F</b>); Valeric acid (<b>G</b>). # <span class="html-italic">p</span> &lt; 0.05, and ## <span class="html-italic">p</span> &lt; 0.01 versus normal. * <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 versus model.</p>
Full article ">Figure 10
<p>Effect of MOS on antibiotic-treated mice. Serum uric acid (<b>A</b>); Serum BUN (<b>B</b>); Serum XOD activity (<b>C</b>); Serum XOD activity (<b>D</b>); Hepatic XOD activity (<b>E</b>); Hepatic ADA activity (<b>F</b>); Urinary uric acid (<b>G</b>); Fecal uric acid (<b>H</b>); Renal URAT1 and GLUT9 protein expression (<b>I</b>); Quantitative analysis of renal URAT1 and GLUT9 protein (<b>J</b>); Intestinal ABCG2, GLUT9 protein expression (<b>K</b>); Quantitative analysis of intestinal ABCG2, GLUT9 protein (<b>L</b>); Total SCFAs content (<b>M</b>); Acetic acid (<b>N</b>); Propionic acid (<b>O</b>); Isobutyric acid (<b>P</b>); Butyric acid (<b>Q</b>); Isovaleric acid (<b>R</b>); Valeric acid (<b>S</b>). # <span class="html-italic">p</span> &lt; 0.05, ## <span class="html-italic">p</span> &lt; 0.01, ### <span class="html-italic">p</span> &lt; 0.001, and #### <span class="html-italic">p</span> &lt; 0.0001 versus normal. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 versus model.</p>
Full article ">
18 pages, 3096 KiB  
Article
Identification of Immune-Active Peptides in Casein Hydrolysates and Its Transport Mechanism on a Caco-2 Monolayer
by Haiyan Xue, Jingjing Han, Jun Ma, Hongxin Song, Baoyuan He, Xiaofeng Liu, Meixia Yi and Lei Zhang
Foods 2023, 12(2), 373; https://doi.org/10.3390/foods12020373 - 13 Jan 2023
Cited by 4 | Viewed by 2097
Abstract
In this study, we investigated the transport mechanism of immune-active peptide fragments isolated from casein gastrointestinal hydrolysates via a Caco-2 monolayer. The casein gastrointestinal hydrolysates could stimulate B-lymphocyte proliferation and reduce the TNF-α level. Then, we identified the bioactive peptide fragments derived from [...] Read more.
In this study, we investigated the transport mechanism of immune-active peptide fragments isolated from casein gastrointestinal hydrolysates via a Caco-2 monolayer. The casein gastrointestinal hydrolysates could stimulate B-lymphocyte proliferation and reduce the TNF-α level. Then, we identified the bioactive peptide fragments derived from casein gastrointestinal hydrolysis using LC-MS/MS. Our results demonstrated that the transport mechanism of five immune-active peptides at the cell level was bypass transport. In addition, the majority of peptide RYPLGYL was transported through the monolayer cell membrane as an intact form for playing immune-active functions. The KHPIK and FFSDK were mainly degraded into small fragments, except for a small amount passing through Caco-2 cells in an entire form. Overall, these results suggested that casein or its immune-active peptides might play a role in regulation of the intestinal immune system. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of casein hydrolysate on B lymphocytes stimulation. (<b>A</b>) B lymphocyte proliferation activities of peptides after 24 h. (<b>B</b>) Effect of casein hydrolysis on secretions of TNF-α in B lymphocyte. In the figure: CH-Casein hydrolysate, PH-Permeate hydrolysate, EH-Entrapment hydrolysate. Data are presented as the mean ± SD (<span class="html-italic">n</span> = 6). ns, <span class="html-italic">p</span> &gt; 0.05; *, <span class="html-italic">p</span> &lt; 0.05; ***, <span class="html-italic">p</span> &lt; 0.001 versus control group.</p>
Full article ">Figure 2
<p>Immunoactivity test of the five synthetic peptides. (<b>A</b>) Cell proliferation rate at different concentrations of peptides. (<b>B</b>) Effect of different peptide concentrations on the secretion of TNF-α in B lymphocytes. Data are presented as the mean ± SD (<span class="html-italic">n</span> = 6). **, <span class="html-italic">p</span> &lt; 0.01 versus control group.1-RYPLGYL, 2-KHPIKH, 3-KHPIK, 4-FFSDK, 5-YGG, 6-control group, 7-blank group.</p>
Full article ">Figure 3
<p>Evaluation of the monolayer density. (<b>A</b>) Caco-2 cells growth (10×). (<b>B</b>) Effect of transportation time on transmittance. (<b>C</b>) Effect of culture time on cell membrane resistance. (<b>D</b>) Effect of culture time on Alkaline phosphatase (AKP) activity ratio.</p>
Full article ">Figure 4
<p>The optimal transportation of conditions of KHPIK. (<b>A</b>) effect of KHPIK concentration on transportation. (<b>B</b>) effect of incubation time on transportation. (<b>C</b>) effect of KHPIK pH on transportation. (<b>D</b>) effect of KHPIK transit direction on transportation. Data is presented as the mean ± SD (<span class="html-italic">n</span> = 6). ns, <span class="html-italic">p</span> &gt; 0.05; *, <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 versus the control group.</p>
Full article ">Figure 5
<p>The composition of the five peptides analyzed by RP−HPLC after passing through the Caco−2 cell monolayer. (<b>A</b>) RYPLGYL (<b>B</b>) KHPIKH (<b>C</b>) KHPIK (<b>D</b>) FFSDK (<b>E</b>)YGG.</p>
Full article ">Figure 6
<p>Effects of three absorption inhibitors or stimulator on peptide segment transport. (<b>A</b>) RYPLGYL. (<b>B</b>) KHPIKH. (<b>C</b>) KHPIK. (<b>D</b>) FFSDK. (<b>E</b>) YGG. Data are presented as the mean ± SD (<span class="html-italic">n</span> = 6). *, <span class="html-italic">p</span> &lt; 0.05; **, <span class="html-italic">p</span> &lt; 0.01; ***, <span class="html-italic">p</span> &lt; 0.001 versus control group.</p>
Full article ">Figure 7
<p>Effects of three efflux inhibitors on peptide segment transport. (<b>A</b>) RYPLGYL. (<b>B</b>) KHPIKH. (<b>C</b>) KHPIK. (<b>D</b>) FFSDK. (<b>E</b>) YGG. Data are presented as the mean ± SD (<span class="html-italic">n</span> = 6). ns, <span class="html-italic">p</span> &gt; 0.05; *, <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 versus control group.</p>
Full article ">Figure 8
<p>RYPLGYL, KHPIK, and FFSDK LC-MS result analysis. (<b>a</b>) LC/MS of RYPLGYL passing through membrane liquid. (<b>b</b>) LC/MS of KHPIK passing through membrane liquid. (<b>c</b>) LC/MS of FFSDK passing through membrane liquid.</p>
Full article ">Figure 8 Cont.
<p>RYPLGYL, KHPIK, and FFSDK LC-MS result analysis. (<b>a</b>) LC/MS of RYPLGYL passing through membrane liquid. (<b>b</b>) LC/MS of KHPIK passing through membrane liquid. (<b>c</b>) LC/MS of FFSDK passing through membrane liquid.</p>
Full article ">
36 pages, 14991 KiB  
Article
An Integrated Analysis of Mechanistic Insights into Biomolecular Interactions and Molecular Dynamics of Bio-Inspired Cu(II) and Zn(II) Complexes towards DNA/BSA/SARS-CoV-2 3CLpro by Molecular Docking-Based Virtual Screening and FRET Detection
by Karunganathan Sakthikumar, Bienfait Kabuyaya Isamura and Rui Werner Maçedo Krause
Biomolecules 2022, 12(12), 1883; https://doi.org/10.3390/biom12121883 - 15 Dec 2022
Viewed by 2203
Abstract
Novel constructed bioactive mixed-ligand complexes (1b) [CuII(L)2(phen)] and (2b) [ZnII(L)2(phen)] {where, L = 2-(4-morpholinobenzylideneamino)phenol), phen = 1,10-phenanthroline} have been structurally analysed by various analytical and spectroscopic techniques, including, magnetic moments, thermogravimetric [...] Read more.
Novel constructed bioactive mixed-ligand complexes (1b) [CuII(L)2(phen)] and (2b) [ZnII(L)2(phen)] {where, L = 2-(4-morpholinobenzylideneamino)phenol), phen = 1,10-phenanthroline} have been structurally analysed by various analytical and spectroscopic techniques, including, magnetic moments, thermogravimetric analysis, and X-ray crystallography. Various analytical and spectral measurements assigned showed that all complexes appear to have an octahedral geometry. Agar gel electrophoresis’s output demonstrated that the Cu(II) complex (1b) had efficient deoxyribonucleic cleavage and complex (2b) demonstrated the partial cleavage accomplished with an oxidation agent, which generates spreadable OH through the Fenton type mechanism. The DNA binding constants observed from viscosity, UV–Vis spectral, fluorometric, and electrochemical titrations were in the following sequence: (1b) > (2b) > (HL), which suggests that the complexes (1b2b) might intercalate DNA, a possibility that is supported by the biothermodynamic measurements. In addition, the observed binding constant results of BSA by electronic absorption and fluorometric titrations indicate that complex (1b) revealed the best binding efficacy as compared to complex (2b) and free ligand. Interestingly, all compounds are found to interact with BSA through a static approach, as further attested by FRET detection. The DFT and molecular docking calculations were also performed to realize the electronic structure, reactivity, and binding capability of all test samples with CT-DNA, BSA, and the SARS-CoV-2 3CLPro, which revealed the binding energies were in a range of −8.1 to −8.9, −7.5 to −10.5 and −6.7–−8.8 kcal/mol, respectively. The higher reactivity of the complexes than the free ligand is supported by the FMO theory. Among all the observed data for antioxidant properties against DPPH, OH, O2−• and NO free radicals, complex (1a) had the best biological efficacy. The antimicrobial and cytotoxic characteristics of all test compounds have been studied by screening against certain selected microorganisms as well as against A549, HepG2, MCF-7, and NHDF cell lines, respectively. The observed findings revealed that the activity enhances coordination as compared to free ligand via Overtone’s and Tweedy’s chelation mechanisms. This is especially encouraging given that in every case, the experimental findings and theoretical detections were in perfect accord. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The proposed structure of complexes (<b>1b</b>–<b>2b</b>) <b>[M<sup>II</sup>(L)<sub>2</sub> (phen)]</b>.</p>
Full article ">Figure 2
<p>The ethidium bromide displacement assay: gel electrophoresis showing the chemical nuclease activity of CT-DNA by the synthesized ligand (<b>HL</b>) and its complexes (<b>1b</b>–<b>2b</b>) in the presence of hydrogen peroxide. Lane: 1 DNA alone + H<sub>2</sub>O<sub>2</sub>; Lane: 2 ligand (<b>HL</b>) + DNA + H<sub>2</sub>O<sub>2</sub>; Lane: 3 complex (<b>1b</b>) + DNA + H<sub>2</sub>O<sub>2</sub>; Lane: 4 complex (<b>2b</b>) + DNA + H<sub>2</sub>O<sub>2</sub>.</p>
Full article ">Figure 3
<p>The absorption spectra of ligand (<b>HL</b>) and its complexes (<b>1b</b>–<b>2b</b>) in Tris-HCl buffer pH = 7.2 at 25 °C in the presence of increasing amount of CT-DNA. The arrow indicates the changes in absorbance upon increasing the CT-DNA concentration and another arrow shows isosbestic points indicates that equilibrium is established between DNA and complexes.</p>
Full article ">Figure 4
<p>(<b>a</b>) DNA thermal denaturation profile at 260 nm in the absence and presence of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>) in 5 mM Tris-HCl/50 mM NaCl buffer pH = 7.2, [DNA]/[Complex] = 1(R), the denaturation temperature (T<sub>m</sub>) was taken as the mid-point of the hyperchromic transition. (<b>b</b>) The derivative melting curve for DNA thermal denaturation at 260 nm in the absence and presence of compounds in 5 mM Tris-HCl/50 mM NaCl buffer pH = 7.2, [DNA]/[Complex] = 1(R).</p>
Full article ">Figure 4 Cont.
<p>(<b>a</b>) DNA thermal denaturation profile at 260 nm in the absence and presence of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>) in 5 mM Tris-HCl/50 mM NaCl buffer pH = 7.2, [DNA]/[Complex] = 1(R), the denaturation temperature (T<sub>m</sub>) was taken as the mid-point of the hyperchromic transition. (<b>b</b>) The derivative melting curve for DNA thermal denaturation at 260 nm in the absence and presence of compounds in 5 mM Tris-HCl/50 mM NaCl buffer pH = 7.2, [DNA]/[Complex] = 1(R).</p>
Full article ">Figure 5
<p>The fluorescence quenching curve of <b>EB</b> bound DNA in the presence of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>). Concentration [DNA] = 240 μM, and [compound] = 0–240 μM. The fluorescence emission spectra of the EB-DNA adduct absorbed at 610 nm.</p>
Full article ">Figure 6
<p>The overlap of UV–Vis spectra of ligand (<b>HL</b>) and its complexes (<b>1b</b>–<b>2b</b>) (<b>Acceptor</b>) at 334–336 nm with fluorescence emission spectrum of BSA (<b>Donor</b>) at 350 nm.</p>
Full article ">Figure 7
<p>The absorption spectra of BSA alone and in the presence of increasing amount of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>) in Tris-HCl buffer pH = 7.2 at 25 °C. The arrow indicates the changes in absorbance upon increasing the compound concentration.</p>
Full article ">Figure 7 Cont.
<p>The absorption spectra of BSA alone and in the presence of increasing amount of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>) in Tris-HCl buffer pH = 7.2 at 25 °C. The arrow indicates the changes in absorbance upon increasing the compound concentration.</p>
Full article ">Figure 8
<p>The optimized geometries of the free ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>).</p>
Full article ">Figure 9
<p>The frontier molecular orbitals of the free ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>). HOMO-LUMO gaps in the gas phase and methanol are provided.</p>
Full article ">Figure 10
<p>The schematic structures of the three biomacromolecules considered for docking studies of ligand (<b>HL</b>) and complexes (<b>1b</b>–<b>2b</b>). Only one protomer of the dimeric Mpro enzyme is provided.</p>
Full article ">Figure 11
<p>The best docking poses of the ligand (<b>HL</b>) and its complexes (<b>1b</b>–<b>2b</b>) against BSA, CT-DNA and Mpro.</p>
Full article ">Figure 12
<p>Histogram showing the comparative antimicrobial activities of ligand (<b>HL</b>) and its mixed ligand complexes (<b>1b</b>–<b>2b</b>) by Agar disc diffusion method.</p>
Full article ">Figure 13
<p>The comparison of cytotoxic effects of ligand (<b>HL</b>) and its complexes (<b>1b</b>–<b>2b</b>) with standard drug cisplatin (<b>CP</b>) against cancer and normal cell lines. Error limits ± 2.5–5.0% (<span class="html-italic">p</span> ≤ 0.05).</p>
Full article ">
16 pages, 1935 KiB  
Article
Activation of OR10A3 by Suberic Acid Promotes Collagen Synthesis in UVB-Irradiated Dermal Fibroblasts via the cAMP-Akt Pathway
by Wesuk Kang, Dabin Choi, Bomin Son, Soyoon Park and Taesun Park
Cells 2022, 11(24), 3961; https://doi.org/10.3390/cells11243961 - 7 Dec 2022
Cited by 5 | Viewed by 2051
Abstract
In recent years, there has been a great deal of interest in the ectopic roles of olfactory receptors (ORs) throughout the human body. Especially, the ectopic function of OR in the skin is one of the most actively researched areas. Suberic acid, a [...] Read more.
In recent years, there has been a great deal of interest in the ectopic roles of olfactory receptors (ORs) throughout the human body. Especially, the ectopic function of OR in the skin is one of the most actively researched areas. Suberic acid, a scent compound, was hypothesized to increase collagen synthesis in the ultraviolet B (UVB)-irradiated human dermal fibroblasts (Hs68) through a specific olfactory receptor. Suberic acid ameliorated UVB-induced decreases in collagen production in Hs68 cells. Using in silico docking to predict the binding conformation and affinity of suberic acid to 15 ectopic ORs detectable in Hs68, several ORs were identified as promising candidates. The effect of suberic acid on collagen synthesis in UVB-exposed dermal fibroblasts was nullified only by a reduction in OR10A3 expression via specific siRNA. In addition, using the cells transiently expressing OR10A3, we demonstrated that suberic acid can activate OR10A3 by assessing the downstream effector cAMP response element (CRE) luciferase activity. We examined that the activation of OR10A3 by suberic acid subsequently stimulates collagen synthesis via the downstream cAMP-Akt pathway. The findings support OR10A3 as a promising target for anti-aging treatments of the skin. Full article
Show Figures

Figure 1

Figure 1
<p>Ultraviolet B (UVB) inhibits procollagen synthesis in Hs68 cells. The cells were irradiated for 24 h with various doses of UVB (5, 10, 15, 20 and 25 µM). Procollagen concentration was determined by enzyme linked immunosorbent assay (ELISA) assay. Data are presented as mean ± standard error of the mean (SEM) of three independent experiments. Statistically significant differences are marked as * <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; ns, not significant.</p>
Full article ">Figure 2
<p>Suberic acid attenuated the reduction of collagen production in UVB-irradiated Hs68 cells. (<b>A</b>) The molecular structure of suberic acid. (<b>B</b>) Cell viability of Hs68 cells treated with various concentrations of suberic acid (SA; 25–400 μM) for 48 h. (<b>C</b>) Procollagen concentrations in the supernatant of Hs68 cells treated with various doses of suberic acid (6.25–400 μM) for 48 h after 10 mJ/cm<sup>2</sup> UVB exposure. (<b>D</b>) Hs68 cells were treated with vehicle or 100 μM suberic acid for 48 h in the absence of UVB irradiation and the concentrations of procollagen in the supernatant were evaluated. The results are shown as means SEM (<span class="html-italic">n</span> = 3). Significant differences between groups are indicated by * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant.</p>
Full article ">Figure 3
<p>Suberic acid increased the production of collagen through OR10A3 in UVB-exposed Hs68 cells. (<b>A</b>) The cells were treated with or without siRNA against 5 ORs (OR1L8, OR2H2, OR10A3, OR10A4 and OR10A6). After treatment for 24 h, the relative mRNA expression of 5 ORs was analyzed. Significant differences between groups are shown as ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 vs. NT siRNA. (<b>B</b>) Hs68 cells were treated with suberic acid (SA; 100 uM) or the vehicle after being exposed to UVB. The siRNA against 5 ORs or NT siRNA were pre-treated for 1 h before the suberic acid treatment. Procollagen contents in the supernatant were analyzed. Significant differences between groups are shown as * <span class="html-italic">p</span> &lt; 0.05 vs. Con; ## <span class="html-italic">p</span> &lt; 0.01 vs. UVB; ns, not significant. Results are shown as means ± SEM (<span class="html-italic">n</span> = 3).</p>
Full article ">Figure 4
<p>The treatment with suberic acid led to the activation of OR10A3. (<b>A</b>) Control plasmid vector or plasmid harboring OR10A3 were introduced into HEK293T cells. The cells were harvested after 48 h and the overexpression of OR10A3 was verified by a semi-quantitative PCR in OR10A3-transfected HEK293T cells. (<b>B</b>) HEK293T cells were co-transfected with the plasmids encoding OR10A3, RTP1S, Golf, and Ric8b, pCRE-luc, and pRL-TK for 48 h. Then, the cells were incubated with vehicle control or suberic acid (SA; 100 uM) for 6 h, and CRE promoter activity was analyzed using the dual-luciferase assay. The results are expressed as the mean SEM of three independent experiments. ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>The activation of OR10A3 by suberic acid increased the production of collagen via downstream cAMP in UVB-exposed Hs68 cells. (<b>A</b>) The cells were treated with suberic acid (SA; 100 uM) or the vehicle and then intracellular cAMP levels were analyzed. (<b>B</b>) Procollagens concentrations in the supernatant of UVB-exposed Hs68 cells treated with suberic acid (100 uM) and SQ22,536 (50 uM) for 24 h. Results are shown as means ± SEM (<span class="html-italic">n</span> = 3). Significant differences between groups are shown as * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant.</p>
Full article ">Figure 6
<p>Suberic acid increased collagen production in UVB-exposed Hs68 cells through the Akt-dependent signaling pathway. Hs68 cells were treated with suberic acid or the vehicle after being exposed to UVB. (<b>A</b>) The OR10A3 siRNA or SQ22,536 (50 uM) was pre-treated for 1 h before the suberic acid treatment (SA; 100 uM). Then, Akt phosphorylation was analyzed. (<b>B</b>) The LY294002 (50 uM) was pre-treated for 1 h before the suberic acid treatment. Procollagen contents in the supernatant were analyzed. (<b>C</b>) Suberic acid-mediated Akt phosphorylation was analyzed without UVB exposure. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as an internal control. Data are presented as mean ± SEM of three independent experiments. Statistically significant differences are marked as * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01; ns, not significant.</p>
Full article ">Figure 7
<p>The activation of OR10A3 by suberic acid enhanced gene expression of Akt downstream transcription factor and its target gene COL1A1 in UVB-exposed Hs68 cells. The cells were treated with suberic acid (SA; 100 uM) or the vehicle after being exposed to UVB. The OR10A3 siRNA or NT siRNA was pretreated for 1 h before the suberic acid treatment. (<b>A</b>) The mRNA expression of SP1 was analyzed. (<b>B</b>,<b>C</b>) The mRNA and the protein expression of COL1A1 were analyzed. GAPDH was used as an internal control. Data are presented as mean ± SEM of three independent experiments. Statistically significant differences are marked as * <span class="html-italic">p</span> &lt; 0.05; ns, not significant.</p>
Full article ">Figure 8
<p>A schematic diagram illustrating the proposed mechanism by which suberic acid increases collagen through OR10A3/cAMP/Akt-dependent signaling pathway in UV-exposed dermal fibroblasts.</p>
Full article ">
24 pages, 4901 KiB  
Article
Apple Derived Exosomes Improve Collagen Type I Production and Decrease MMPs during Aging of the Skin through Downregulation of the NF-κB Pathway as Mode of Action
by Martina Trentini, Ilaria Zanolla, Federica Zanotti, Elena Tiengo, Danilo Licastro, Simeone Dal Monego, Luca Lovatti and Barbara Zavan
Cells 2022, 11(24), 3950; https://doi.org/10.3390/cells11243950 - 7 Dec 2022
Cited by 19 | Viewed by 3222
Abstract
Skin ageing is strictly related to chronic inflammation of the derma and the decay of structural proteins of the extracellular matrix. Indeed, it has become common practice to refer to this phenomenon as inflammageing. Biotech innovation is always in search of new active [...] Read more.
Skin ageing is strictly related to chronic inflammation of the derma and the decay of structural proteins of the extracellular matrix. Indeed, it has become common practice to refer to this phenomenon as inflammageing. Biotech innovation is always in search of new active principles that induce a youthful appearance. In this paper, apple-derived nanovesicles (ADNVs) were investigated as novel anti-inflammatory compounds, which are able to alter the extracellular matrix production of dermal fibroblasts. Total RNA sequencing analysis revealed that ADNVs negatively influence the activity of Toll-like Receptor 4 (TLR4), and, thus, downregulate the NF-κB pro-inflammatory pathway. ADNVs also reduce extracellular matrix degradation by increasing collagen synthesis (COL3A1, COL1A2, COL8A1 and COL6A1) and downregulating metalloproteinase production (MMP1, MMP8 and MMP9). Topical applications for skin regeneration were evaluated by the association of ADNVs with hyaluronic-acid-based hydrogel and patches. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) SEM Imaging of ADNVs (MAG, 5.0 KX; WD, 15 mm; EHT, 15 kV; High Vacuum); (<b>B</b>) Protein quantification of the ADNV fraction after isolation (μg/mL); and (<b>C</b>) TRPS analysis output.</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Release curve of ADNVs from hyaluronic-acid-based materials, MeHA patch and HY 2% gel, at room temperature (RT = 25 °C) and physiological temperature (37 °C). ADNV release is expressed in form of protein concentration (μg/mL) in time (hours). (<b>C</b>) Values corresponding to the area underneath each curve in (<b>A</b>,<b>B</b>) (SE). (<b>D</b>) Illustrative image of MeHA patch and HY 2% gel. **** <span class="html-italic">p</span> &lt; 0.0001.</p>
Full article ">Figure 3
<p>(<b>A</b>) MTT results for NCTC L929 cells grown in control medium (CTRL) and in ADNV-spiked medium (ADNVs); cell viability is expressed as absorbance. (<b>B</b>,<b>C</b>) Migration assay results are displayed as images taken in time, from 0 to 12 h (hours); the red line indicates the scratch’s middle, while black arrows point towards cells that have significantly moved since the previous image was taken.</p>
Full article ">Figure 4
<p>(<b>A</b>,<b>B</b>) Volcano plot of microarray outputs for primary dermal fibroblasts grown w/ TNFα (<b>A</b>) or w/o TNFα. Both graphs represent control (w/o ADNVs) vs treated (w/ ADNVs) conditions. Ten most relevant genes in terms of fold change and significance are reported.</p>
Full article ">Figure 5
<p>(<b>A</b>–<b>C</b>) Expression logarithmic ratios of differentially regulated genes from the w/ TNFα microarray involved in ECM maintenance. Expression is displayed as a bar, while significance, expressed as −log10(<span class="html-italic">p</span>-value), is reported in orange.</p>
Full article ">Figure 6
<p>(<b>A</b>–<b>C</b>) Expression logarithmic ratios of differentially regulated genes from the w/ TNFα microarray involved in the inflammation response. Expression is displayed as a bar, while significance, expressed as −log10(<span class="html-italic">p</span>-value), is reported in orange. (<b>D</b>) KEGG pathway: IL-1b and NF-κB pathway, with color-coded protein slots corresponding to DEGs expression.</p>
Full article ">Figure 7
<p>Canonical pathway analysis by IPA for w/ TNFα. (<b>A</b>) Ten first entries. (<b>B</b>–<b>E</b>) Genes involved in the first four pathways with expression log ratio values.</p>
Full article ">Figure 8
<p>(<b>A</b>) Upstream miRNA regulators for w/ TNFα IPA analysis, z-Score and <span class="html-italic">p</span>-value are shown. (<b>B</b>) IPA regulator’s ranked by influenced functions, shown as percentage of the total regulator effects. (<b>C</b>) KEGG pathway: Toll-like receptors signaling pathway, with color-coded protein slots corresponding to DEGs expression.</p>
Full article ">Figure 9
<p>(<b>A</b>) Canonical pathway analysis by IPA for w/o TNFα. (<b>A</b>) Ten first entries. (<b>B</b>) Gene expression of genes involved in cholesterol biosynthesis (w/o TNFα dataset); expression is displayed as a bar, while significance, expressed as −log10(<span class="html-italic">p</span>-value), is reported in orange. (<b>C</b>) RT-qPCR results for gene expression in w/ TNFα fibroblasts after 1× and 0.5× ADNV treatment (** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001).</p>
Full article ">Figure 10
<p>Mitochondrial stress under all conditions is expressed as percentage of cells with above mitochondrial ROS production. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
9 pages, 762 KiB  
Commentary
NLRP3 and Gut Microbiota Homeostasis: Progress in Research
by Hongming Pan, Yuting Jian, Feijie Wang, Shaokun Yu, Jiannan Guo, Juntao Kan and Wei Guo
Cells 2022, 11(23), 3758; https://doi.org/10.3390/cells11233758 - 24 Nov 2022
Cited by 17 | Viewed by 2111
Abstract
The inflammasome is a platform for inflammatory signaling, and the NLRP3 inflammasome recognizes stimuli in vitro and in vivo, and releases inflammatory cytokines that trigger inflammation and pyroptosis. In the gut, the NLRP3 inflammasome is a key sensor for protecting the body from [...] Read more.
The inflammasome is a platform for inflammatory signaling, and the NLRP3 inflammasome recognizes stimuli in vitro and in vivo, and releases inflammatory cytokines that trigger inflammation and pyroptosis. In the gut, the NLRP3 inflammasome is a key sensor for protecting the body from damage and exogenous pathogens. It plays a fundamental role in maintaining the stability of the gut’s immune system. We focus on the role of NLRP3 as a key node in maintaining the homeostasis of gut microbiota which has not been fully highlighted in the past; gut microbiota and innate immunity, as well as the NLRP3 inflammasome, are discussed in this article. Full article
Show Figures

Figure 1

Figure 1
<p>Activated inflammasomes in the gut recognize stimuli and initiate inflammatory responses. Inflammasomes in the intestine recognize the pathogen-associated molecular patterns (PAMPS) of exogenous pathogens invading the host, or the damaged and dying cells release the dangerous-associated molecular patterns (DAMPs) in the body, which will stimulate ASC to assemble with pro-caspase-1 to form inflammasomes. The assembled inflammasomes promote pro-caspase-1 to self-cleavage and produce activated caspase-1, and the activated caspase-1 will convert pro-IL-1 β and pro-IL-18 to IL-1 β and IL-18, which in turn triggers an inflammatory response in the intestine (diagram by Figdraw).</p>
Full article ">Figure 2
<p>NLRP3 inflammasome in the intestine recognizes bacteria, viruses, or other pathogens and promotes the activation of pro-cassase-1 to caspase-1. On the one hand, caspase-1 will cleave GSDMD to produce an active segment N-terminal, which can bind to the cell membrane to form many 10–14 nm holes to cause pyroptosis and release IL-1 β and IL-18 to promote inflammatory reaction. On the other hand, activated caspase-1 activates pro-IL-1 β and pro-IL-18, activated IL-1 β, further inducing the expression of inflammatory cytokines and chemokines in the inflammatory part; it can promote the recruitment and activation of neutrophils and dendritic cells in the inflammatory part, and promote the differentiation of Th17 and Tregs. IL-18 can activate intestinal epithelial cells to produce AMPs, stimulate the production of Th1, NK, and NKT to secrete IFN- γ, or combine with IL-12 to induce more γδ T cells which secrete IL-17. NLRP3 inflammasome promotes the local inflammatory response of the intestinal and helps remove the pathogenic factors invading the body to maintain intestinal homeostasis through the above way (diagram by Figdraw).</p>
Full article ">
19 pages, 6883 KiB  
Article
Enhancement of Efficacy of Retinoids through Enhancing Retinoid-Induced RAR Activity and Inhibiting Hydroxylation of Retinoic Acid, and Its Clinical Efficacy on Photo-Aging
by Seongsu Kang, Hyejin Lee, Seung-Hyun Jun, Sun-Gyoo Park and Nae-Gyu Kang
Pharmaceutics 2022, 14(11), 2412; https://doi.org/10.3390/pharmaceutics14112412 - 8 Nov 2022
Cited by 2 | Viewed by 4557
Abstract
Retinoids, one of the most robust bioactive materials, have been widely used to improve various dermatological and pathological conditions. The body has an endogenous mechanism that modulates the exogenous retinoid above physiological concentrations, which limits the bioavailability or pharmacological efficacy of retinoids. Considering [...] Read more.
Retinoids, one of the most robust bioactive materials, have been widely used to improve various dermatological and pathological conditions. The body has an endogenous mechanism that modulates the exogenous retinoid above physiological concentrations, which limits the bioavailability or pharmacological efficacy of retinoids. Considering that most retinoids trigger extensive irritation in users, it is necessary to enhance the pharmacological efficacy of retinoids, thereby achieving a higher efficacy at a lower dosage. Here, we present approaches for enhancing the efficacy of retinol by enhancing retinoid-induced RAR gamma (RAR-γ) activity and inhibiting the hydroxylation of retinoic acid. Using both in vitro and ex vivo experiments, retinoid boosters were demonstrated to enhance pharmacological efficacy. A small pilot study was conducted to investigate the efficacy for improvement of facial wrinkles, whose results revealed that these boosters could enhance the pharmacological efficacy of topical applications of both retinol and retinoic acid for cosmetic use. These results promote not only a higher compliance among retinoids users, but also provide significant insights into the mechanisms underlying the action of retinoids. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>RAR-γ activation by retinoids and camphor. (<b>a</b>) RAR-γ activation by retinoids. Retinoic acid (RA), retinaldehyde (RAL), and retinol (ROL) were analyzed. RAR-γ activation in the reporter cell line was measured by luciferase activity. (<b>b</b>) RAR-γ activation by camphor. (<b>c</b>) Relative mRNA expression of retinoid metabolism-related genes. (<b>d</b>) Type-1 procollagen expression of fibroblast. * Significantly different results (Student’s <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt;  0.05).</p>
Full article ">Figure 2
<p>Screening RAMBA-mimicking compounds inhibiting RA hydroxylation. (<b>a</b>) HPLC spectrum for RA metabolites. HaCaT were treated with 1 µM RA for 48 h. Culture media and cell extracts were lyophilized and analyzed. For the inhibitor group (black line), 25 µM of apigenin was used. (<b>b</b>) An enlarged detail of the spectrum surrounded by the dotted box in (<b>a</b>). (<b>c</b>) RA hydroxylation efficiency, calculated as follows: 100 × peak area (oxidized RA metabolites, 5~15 min)/peak area (oxidized RA metabolites + RA). PI refers to prolinamidoethyl imidazole. (<b>d</b>) Relative mRNA expression of CYPs. HaCaT was cultured with candidates (25 µM) and RA for 48 h. Combination refers to co-treatment of apigenin, baicalin, and luteolin (each 25 µM). The bars denote the standard deviations. * Significantly different results (Student’s <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>Efficacy of RA hydroxylation inhibitors in vitro, ex vivo, and in vivo. (<b>a</b>) Expression of collagen by fibroblasts co-cultured with keratinocytes. The <span class="html-italic">COL1A1</span> mRNA levels were analyzed by RT-qPCR. Keratinocytes (HaCaT) were cultured with RA or hydroxylation inhibitors prior to co-culture. A detailed experimental procedure is shown. Booster included 12.5 µM apigenin and baicalin and 125 µM prolinamidoethyl imidazole. (<b>b</b>) Epidermis thickening effects of RA and hydroxylation inhibitors in ex vivo experiments. An artificially reconstituted 3D skin model was used. (<b>c</b>) Change in the density of the epidermis and dermis in vivo. In a small clinical pilot study, O/W-type cream supplemented with RA (0.01%) and hydroxylation inhibitors (0.01% of apigenin, baicalin, and prolinamidoethyl imidazole) were applied onto the skin on the forearm of subjects twice a day for eight weeks. (<b>d</b>) Sonography analysis of RA- and booster-treated areas. Standard deviation bars are shown. * Significantly different results (Student’s <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Efficacy of retinoids booster ex vivo and in vivo. (<b>a</b>) Epidermis thickening effects of retinol and retinol/booster. Scale bar, 50 µm. (<b>b</b>) Expression of IL-1α measured by ELISA. (<b>c</b>) Study design. Half &amp; half, and single-blinded clinical test was performed. The table on the right shows the booster formulas for retinol. (<b>d</b>,<b>e</b>) Improvement in the appearance of wrinkles by retinol and retinol/booster (after 11 weeks of application). Three types of wrinkles were measured by Antera 3D. * Significantly different results (Paired <span class="html-italic">t</span>-test, <span class="html-italic">p</span> &lt; 0.05). (<b>f</b>) Self-assessment for efficacy. Human subjects were asked which cream they thought was better at improving the appearance of their skin.</p>
Full article ">Figure 5
<p>Long-term use of retinol and booster (194 days). Data for the most responsive human subject for each clinical features are shown. All data were analyzed by Antera 3D. (<b>a</b>) Improvement in the appearance of crow’s feet. (<b>b</b>) Improvement in the appearance of frontal eye wrinkle. (<b>c</b>) Improvement in the appearance of pores (front cheek area). (<b>d</b>) Improvement in hyperpigmentation spots. (<b>e</b>) Improvement in skin texture. Skin surface (lower row) was evaluated by microscopy. Scale bar (white), 200 µm.</p>
Full article ">
Back to TopTop