[go: up one dir, main page]

 
 
Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,401)

Search Parameters:
Keywords = prebiotic

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 3031 KiB  
Article
Functional Muffins Exert Bifidogenic Effects along with Highly Product-Specific Effects on the Human Gut Microbiota Ex Vivo
by Stef Deyaert, Jonas Poppe, Lam Dai Vu, Aurélien Baudot, Sarah Bubeck, Thomas Bayne, Kiran Krishnan, Morgan Giusto, Samuel Moltz and Pieter Van den Abbeele
Metabolites 2024, 14(9), 497; https://doi.org/10.3390/metabo14090497 (registering DOI) - 14 Sep 2024
Viewed by 191
Abstract
GoodBiome™ Foods are functional foods containing a probiotic (Bacillus subtilis HU58™) and prebiotics (mainly inulin). Their effects on the human gut microbiota were assessed using ex vivo SIFR® technology, which has been validated to provide clinically predictive insights. GoodBiome™ Foods (BBM/LCM/OSM) [...] Read more.
GoodBiome™ Foods are functional foods containing a probiotic (Bacillus subtilis HU58™) and prebiotics (mainly inulin). Their effects on the human gut microbiota were assessed using ex vivo SIFR® technology, which has been validated to provide clinically predictive insights. GoodBiome™ Foods (BBM/LCM/OSM) were subjected to oral, gastric, and small intestinal digestion/absorption, after which their impact on the gut microbiome of four adults was assessed (n = 3). All GoodBiome™ Foods boosted health-related SCFA acetate (+13.1/14.1/13.8 mM for BBM/LCM/OSM), propionate (particularly OSM; +7.4/7.5/8.9 mM for BBM/LCM/OSM) and butyrate (particularly BBM; +2.6/2.1/1.4 mM for BBM/LCM/OSM). This is related to the increase in Bifidobacterium species (B. catenulatum, B. adolescentis, B. pseudocatenulatum), Coprococcus catus and Bacteroidetes members (Bacteroides caccae, Phocaeicola dorei, P. massiliensis), likely mediated via inulin. Further, the potent propionogenic potential of OSM related to increased Bacteroidetes members known to ferment oats (s key ingredient of OSM), while the butyrogenic potential of BBM related to a specific increase in Anaerobutyricum hallii, a butyrate producer specialized in the fermentation of erythritol (key ingredient of BBM). In addition, OSM/BBM suppressed the pathogen Clostridioides difficile, potentially due to inclusion of HU58™ in GoodBiome™ Foods. Finally, all products enhanced a spectrum of metabolites well beyond SCFA, including vitamins (B3/B6), essential amino acids, and health-related metabolites such as indole-3-propionic acid. Overall, the addition of specific ingredients to complex foods was shown to specifically modulate the gut microbiome, potentially contributing to health benefits. Noticeably, our findings contradict a recent in vitro study, underscoring the critical role of employing a physiologically relevant digestion/absorption procedure for a more accurate evaluation of the microbiome-modulating potential of complex foods. Full article
(This article belongs to the Special Issue Natural Metabolites on Gut Microbiome Modulation)
Show Figures

Figure 1

Figure 1
<p><b>Schematic overview of the study design using ex vivo SIFR<sup>®</sup> technology.</b> (<b>a</b>) Reactor design using the ex vivo SIFR<sup>®</sup> technology to evaluate the impact of GoodBiome<sup>TM</sup> Foods against an unsupplemented parallel control (NSC = no substrate control). (<b>b</b>) Timeline and analysis at different timepoints.</p>
Full article ">Figure 2
<p><b>The fecal microbiota covered clinically relevant interpersonal differences.</b> Abundances (%) of the key families (top 15), as quantified via shallow shotgun sequencing, in the fecal microbiota of each of the four human adults that provided a fecal donation for the current SIFR<sup>®</sup> study.</p>
Full article ">Figure 3
<p><b>GoodBiome™ Foods exerted marked effects on microbial metabolic activity over time.</b> The effects on (<b>A</b>) pH, (<b>B</b>) gas production, (<b>C</b>) total SCFA, (<b>D</b>) acetate, (<b>E</b>) propionate, (<b>F</b>) butyrate, (<b>G</b>) valerate, and (<b>H</b>) bCFA were compared for GoodBiome™ Foods versus an unsupplemented control (NSC) at 6 h, 24 h, 30 h, and 48 h after the initiation of colonic incubation. Data were presented as means across simulations for four individual donors (n = 3 per donor). The statistical significance of the treatment effects for the test products vs. NSC within each timepoint can be found in <a href="#app1-metabolites-14-00497" class="html-app">Figures S2 and S3</a>.</p>
Full article ">Figure 4
<p><b>GoodBiome™ Foods exerted significant impact on microbial composition at phylum level.</b> Samples were collected 30 h after the colonic incubations were initiated. Data were expressed as average absolute levels (cells/mL) of each phylum across simulations for four individual donors (n = 3 per donor). The statistical significance of the potential treatment effects within each comparison was determined via Benjamani–Hochberg post hoc testing. Significant changes (<span class="html-italic">p</span><sub>adjusted</sub> &lt; 0.05) were indicated with asterisks.</p>
Full article ">Figure 5
<p><b>GoodBiome™ Foods exerted significant impact on microbial composition at species level.</b> The bar charts were generated for species that were significantly (FDR = 0.05) affected by any of the treatments at 30 h, expressed as log2fold change (treatment/NSC), averaged across four human adults (n = 3 per donor). Purple and red bars indicated significant/consistent decreases and increases, respectively. Notable health- or disease-related taxa are highlighted in a gray box.</p>
Full article ">Figure 6
<p><b>The GoodBiome™ Foods exerted significant impact on taxa that are potentially relevant for human health.</b> Violin plots, expressed as log2fold change (treatment/NSC), were presented for four individual human adults (n = 3). The data were presented for (<b>A</b>) <span class="html-italic">Clostridiodes difficile</span> (<b>B</b>) <span class="html-italic">Bifidobacteriaceae</span>, (<b>C</b>) <span class="html-italic">Anaerobutyricum hallii</span>, (<b>D</b>) <span class="html-italic">Bacteroidaceae</span>, <span class="html-italic">Bacteroidales_u_f</span>, and <span class="html-italic">Tannerellaceae</span>. For (<b>B</b>–<b>D</b>), Pearson correlation analysis demonstrated significant positive correlations (<span class="html-italic">p</span> &lt; 0.05) between the absolute levels of these taxa (cells/mL) and the concentration (mM) of the most relevant SCFA related to these taxa, i.e., (<b>A</b>) acetate, (<b>B</b>) butyrate, and (<b>C</b>) propionate.</p>
Full article ">Figure 7
<p><b>The GoodBiome™ Foods exerted significant impact on the production of microbial metabolites, well beyond SCFA.</b> The bars were generated for metabolites that were significantly (FDR = 0.05) affected by any of the treatments, expressed as log2fold change (treatment/NSC), averaged across four human adults (n = 3 per test subject). Purple and red bars indicated significant decreases and increases, respectively.</p>
Full article ">
13 pages, 2078 KiB  
Article
Assessment of Enzymatically Derived Blackcurrant Extract as Cosmetic Ingredient—Antioxidant Properties Determination and In Vitro Diffusion Study
by Anja Petrov Ivanković, Marija Ćorović, Ana Milivojević, Stevan Blagojević, Aleksandra Radulović, Rada Pjanović and Dejan Bezbradica
Pharmaceutics 2024, 16(9), 1209; https://doi.org/10.3390/pharmaceutics16091209 (registering DOI) - 14 Sep 2024
Viewed by 287
Abstract
Blackcurrant is an anthocyanin-rich berry with proven antioxidant and photoprotective activity and emerging prebiotic potential, widely applied in cosmetic products. Hereby, highly efficient enzyme-assisted extraction of blackcurrant polyphenols was performed, giving extract with very high antioxidant activity. Obtained extract was characterized in terms [...] Read more.
Blackcurrant is an anthocyanin-rich berry with proven antioxidant and photoprotective activity and emerging prebiotic potential, widely applied in cosmetic products. Hereby, highly efficient enzyme-assisted extraction of blackcurrant polyphenols was performed, giving extract with very high antioxidant activity. Obtained extract was characterized in terms of anthocyanin composition, incorporated into three different cosmetic formulations and subjected to Franz cell diffusion study. Experimental values obtained using cellulose acetate membrane for all four dominant anthocyanins (delphinidin 3-glucoside, delphinidin 3-rutinoside, cyanidin 3-glucoside and cyanidin 3-rutinoside) were successfully fitted with the Korsmeyer–Peppas diffusion model. Calculated effective diffusion coefficients were higher for hydrogel compared to oil-in-water cream gel and oil-in-water emulsion, whereas the highest value was determined for cyanidin 3-rutinoside. On the other hand, after a 72 h long experiment with transdermal skin diffusion model (Strat-M® membrane), no anthocyanins were detected in the receptor fluid, and only 0.5% of the initial quantity from the donor compartment was extracted from the membrane itself after experiment with hydrogel. Present study revealed that hydrogel is a suitable carrier system for the topical delivery of blackcurrant anthocyanins, while dermal and transdermal delivery of these molecules is very limited, which implies its applicability for treatments targeting skin surface (i.e., prebiotic, photoprotective). Full article
(This article belongs to the Special Issue Advances in Natural Products for Cutaneous Application)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Characteristic chromatogram of the anthocyanins from blackcurrant extract: (1)—delphinidin-3-glucoside, (2)—delphinidin-3-rutinoside, (3)—cyanidin-3-glucoside and (4)—cyanidin-3-rutinoside (cya-3-glu), recorded at 520 nm.</p>
Full article ">Figure 2
<p>Concentration profile (<b>a</b>) and cumulative release (<b>b</b>) of anthocyanins from the hydrogel. Data represent the mean values of three independent experiments, and error bars indicate the standard deviations.</p>
Full article ">Figure 3
<p>Concentration profile (<b>a</b>) and cumulative release (<b>b</b>) of anthocyanins from the oil-in-water gel cream. Data represent the mean values of three independent experiments, and error bars indicate the standard deviations.</p>
Full article ">Figure 4
<p>Concentration profile (<b>a</b>) and cumulative release (<b>b</b>) of anthocyanins from the oil-in-water emulsion. Data represent the mean values of three independent experiments, and error bars indicate the standard deviations.</p>
Full article ">Figure A1
<p>Representative overlapped chromatograms of anthocyanins diffusion from hydrogel over time.</p>
Full article ">
31 pages, 3407 KiB  
Review
Glucose Metabolism-Modifying Natural Materials for Potential Feed Additive Development
by Wei-Chih Lin, Boon-Chin Hoe, Xianming Li, Daizheng Lian and Xiaowei Zeng
Pharmaceutics 2024, 16(9), 1208; https://doi.org/10.3390/pharmaceutics16091208 - 13 Sep 2024
Viewed by 269
Abstract
Glucose, a primary energy source derived from animals’ feed ration, is crucial for their growth, production performance, and health. However, challenges such as metabolic stress, oxidative stress, inflammation, and gut microbiota disruption during animal production practices can potentially impair animal glucose metabolism pathways. [...] Read more.
Glucose, a primary energy source derived from animals’ feed ration, is crucial for their growth, production performance, and health. However, challenges such as metabolic stress, oxidative stress, inflammation, and gut microbiota disruption during animal production practices can potentially impair animal glucose metabolism pathways. Phytochemicals, probiotics, prebiotics, and trace minerals are known to change the molecular pathway of insulin-dependent glucose metabolism and improve glucose uptake in rodent and cell models. These compounds, commonly used as animal feed additives, have been well studied for their ability to promote various aspects of growth and health. However, their specific effects on glucose uptake modulation have not been thoroughly explored. This article focuses on glucose metabolism is on discovering alternative non-pharmacological treatments for diabetes in humans, which could have significant implications for developing feed additives that enhance animal performance by promoting insulin-dependent glucose metabolism. This article also aims to provide information about natural materials that impact glucose uptake and to explore their potential use as non-antibiotic feed additives to promote animal health and production. Further exploration of this topic and the materials involved could provide a basis for new product development and innovation in animal nutrition. Full article
Show Figures

Figure 1

Figure 1
<p>Pathway of insulin-dependent glucose uptake and AMPK activation-induced translocation of GLUT4 vesicles. Pointed arrows represent activation or translocation in the signaling pathways. The “P” symbol with a circle represents a phosphorylation event. After insulin binds to the IR and triggers tyrosine autophosphorylation, the IRSs are activated to promote the PI3K/AKT pathway, eventually inducing the translocation of GLUT4 vesicles to the cell membranes and facilitating glucose uptake.</p>
Full article ">Figure 2
<p>Putative mechanisms by which ROS interferes with insulin-dependent glucose uptake. Pointed arrows represent activation or translocation in the signaling pathways. Line-headed arrows indicate inhibition of the signaling process. The “P” symbol with a circle represents a phosphorylation event. ROS directly inhibits the expression of PI3K/AKB and the translocation of GLUT4. ROS also causes protein misfolding, which can directly inflict IR. The cellular damage can induce the production of TNF-α, which inhibits the phosphorylation of IR. ER stress caused by ROS facilitates the ubiquitination of IRS via the JNK pathway, inhibiting the downstream signaling.</p>
Full article ">Figure 3
<p>Potential crosstalk between oxidative stress and inflammation through phytochemicals in poultry [<a href="#B75-pharmaceutics-16-01208" class="html-bibr">75</a>]. Copyright@2019, Animal Bioscience, Seoul, Republic of Korea.</p>
Full article ">Figure 4
<p>Mechanisms of phytochemicals to modulate glucose metabolism. Upward arrows (↑) and downward arrows (↓) represent the upregulation and downregulation of specific molecules, respectively. The antioxidant and anti-inflammatory effects of phytochemicals prevent the potential inhibition of insulin-dependent metabolism by oxidative stresses. Also, phytochemicals can directly stimulate the expressions of IRSs, PI3K/AKT, and AMPK, eventually facilitating GLUT4 translocation.</p>
Full article ">Figure 5
<p>Mechanisms of probiotics and prebiotics on the modulation of insulin-dependent glucose metabolism. Upward arrows (↑) and downward arrows (↓) represent the upregulation and downregulation of specific molecules, respectively. Probiotics and prebiotics can work independently or synergistically to modulate the intestinal microbiome and facilitate the production of SCFAs. The SCFAs then stimulate the GPR 41 and 43, promote pro-insulin GLP-1, and enhance host insulin secretion. Individually, prebiotics can directly promote the IRS/PI3K/AKT pathway and stimulate GLUT4 translocation. On the other hand, probiotics directly stimulate PI3K/AKT/GLUT4 with their metabolites, such as surfactin. Furthermore, some probiotics, after successful colonization in the intestinal environment, can exert their benefits through SCFA production.</p>
Full article ">Figure 6
<p>Mechanisms of Se and Cr<sup>3+</sup> on the modulation of insulin-dependent glucose metabolism. These two trace elements can reduce cellular ER stress via their antioxidant effects. Upward arrows (↑) and downward arrows (↓) represent the upregulation and downregulation of specific molecules, respectively. The reduced ER stress prevents IRSs from ubiquitination, which preserves the downstream signaling cascade of insulin-dependent glucose uptake. Se and Cr<sup>3+</sup> have been reported to promote the activation of serine and threonine kinases with subsequent IRS phosphorylation. Se and Cr<sup>3+</sup> have the ability to respectively stimulate GLP-1 for increased insulin production or activate AMPK-facilitated GLUT4 translocation.</p>
Full article ">Figure 7
<p>The importance of gut microbial metabolism in regulating insulin sensitivity in humans and mice [<a href="#B210-pharmaceutics-16-01208" class="html-bibr">210</a>]. Upward arrows (↑) and downward arrows (↓) represent the upregulation and downregulation of specific molecules, respectively. Question mark (?) indicates the hypothesized modulation effects. Copyright@2024, Nature, London, UK.</p>
Full article ">Figure 8
<p>Flow chart showing how functional non-antibiotic feed additives support animal health and preserve good product quality.</p>
Full article ">
12 pages, 2118 KiB  
Review
Akkermansia muciniphila as a Potential Guardian against Oral Health Diseases: A Narrative Review
by Molly H. Anderson, Karima Ait-Aissa, Amal M. Sahyoun, Ammaar H. Abidi and Modar Kassan
Nutrients 2024, 16(18), 3075; https://doi.org/10.3390/nu16183075 - 12 Sep 2024
Viewed by 405
Abstract
The oral microbiome is a diverse ecosystem containing a community of symbiotic, commensal, and pathogenic microorganisms. One key microorganism linked to periodontal disease (PD) is Porphyromonas gingivalis (P. gingivalis), a Gram-negative anaerobic bacterium known to have several virulence factors that trigger [...] Read more.
The oral microbiome is a diverse ecosystem containing a community of symbiotic, commensal, and pathogenic microorganisms. One key microorganism linked to periodontal disease (PD) is Porphyromonas gingivalis (P. gingivalis), a Gram-negative anaerobic bacterium known to have several virulence factors that trigger inflammation and immune evasion. On the other hand, Akkermansia muciniphila (A. muciniphila), a symbiotic bacterium, has been recently shown to play an important role in mitigating inflammation and reducing periodontal damage. In vivo and in vitro studies have shown that A. muciniphila decreases inflammatory mediators and improves immune responses, suggesting its role in mitigating PD and related inflammatory systemic conditions such as diabetes, hypertension, and obesity. This review discusses the anti-inflammatory effects of A. muciniphila, its impact on periodontal health, and its potential role in managing systemic diseases. The overall aim is to elucidate how this bacterium might help reduce inflammation, improve oral health, and influence broader health outcomes. Full article
(This article belongs to the Section Nutrition and Public Health)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Oral Bacterial Imbalance, Inflammation, and the Protective Role of <span class="html-italic">Akkermansia muciniphila</span>. Imbalance in oral bacteria leads to inflammation, which contributes to periodontal diseases and can impact systemic health. <span class="html-italic">Akkermansia muciniphila</span>, a gut bacterium with anti-inflammatory properties, can help alleviate these conditions by reducing inflammation and promoting oral and overall health. Red arrows indicate the potentiation or worsening of diseases due to inflammation, while green arrows signify the inhibition or alleviation of diseases by decreasing inflammation.</p>
Full article ">Figure 2
<p><span class="html-italic">Akkermansia. muciniphila</span> and inflammation. <span class="html-italic">A. muciniphila</span> by increasing acetate and propionate levels reduces pro-inflammatory cytokines (TNF-α, IL-6, IL-1α, IL-12) while simultaneously increasing the anti-inflammatory cytokine IL-10. These findings highlight the crucial role of <span class="html-italic">A. muciniphila</span> in mitigating inflammation and highlight its potential as a therapeutic agent. Symbol ↑ indicates upregulation/increase and symbol ↓ indicates downregulation/decrease.</p>
Full article ">Figure 3
<p><span class="html-italic">A. muciniphila</span> and Oral Health-induced inflammation and bone loss. <span class="html-italic">Porphyromonas gingivalis</span> (<span class="html-italic">P. gingivalis</span>) and <span class="html-italic">Fusobacterium nucleatum</span> (<span class="html-italic">F. nucleatum</span>) have detrimental effects on periodontal diseases (PD) by triggering inflammatory responses and bone destruction. <span class="html-italic">Akkermansia muciniphila</span> has been shown to reduce inflammation, mitigate bone destruction, and enhance anti-inflammatory responses in models of PD. These findings highlight the potential of <span class="html-italic">A. muciniphila</span> as a therapeutic agent in the management of PD-associated inflammation and bone loss. Red arrows indicate the potentiation or worsening of diseases due to inflammation and bone loss, while green arrows signify the inhibition or alleviation of diseases by decreasing inflammation and bone destruction. Symbol ↑ indicates increase.</p>
Full article ">Figure 4
<p><span class="html-italic">Akkermansia muciniphila</span> and Other Systemic Diseases Related to Oral Health. <span class="html-italic">Akkermansia muciniphila</span> (<span class="html-italic">A. muciniphila</span>) has emerged as a notable player in managing systemic conditions linked to inflammation, including periodontal disease (PD), obesity, type 2 diabetes (T2D), and hypertension (HTN). <span class="html-italic">A. muciniphila</span>’s influence on inflammatory pathways and metabolic parameters highlights its potential as a therapeutic agent for chronic diseases and emphasizes the importance of the gut microbiome in overall health. Green arrows signify the inhibition or alleviation of diseases by decreasing inflammation.</p>
Full article ">Figure 5
<p>Impact of Specific Foods on Metabolic Diseases-induced periodontal diseases and <span class="html-italic">A. muciniphila</span> Abundance. Certain foods that help manage metabolic diseases like hyperlipidemia, diabetes, and hypertension also improve oral health. These foods increase the abundance of <span class="html-italic">Akkermansia muciniphila</span> (<span class="html-italic">A. muciniphila</span>), which may enhance their positive effects on systemic diseases and oral health. This suggests that dietary strategies could effectively address chronic conditions by leveraging <span class="html-italic">A. muciniphila</span>’s role in the gut microbiome to improve overall health.</p>
Full article ">
20 pages, 2164 KiB  
Review
Deciphering the Gut–Liver Axis: A Comprehensive Scientific Review of Non-Alcoholic Fatty Liver Disease
by Samradhi Singh, Mona Kriti, Roberto Catanzaro, Francesco Marotta, Mustafa Malvi, Ajay Jain, Vinod Verma, Ravinder Nagpal, Rajnarayan Tiwari and Manoj Kumar
Livers 2024, 4(3), 435-454; https://doi.org/10.3390/livers4030032 - 12 Sep 2024
Viewed by 279
Abstract
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant global health issue. The condition is closely linked to metabolic dysfunctions such as obesity and type 2 diabetes. The gut–liver axis, a bidirectional communication pathway between the liver and the gut, plays a [...] Read more.
Non-alcoholic fatty liver disease (NAFLD) has emerged as a significant global health issue. The condition is closely linked to metabolic dysfunctions such as obesity and type 2 diabetes. The gut–liver axis, a bidirectional communication pathway between the liver and the gut, plays a crucial role in the pathogenesis of NAFLD. This review delves into the mechanisms underlying the gut–liver axis, exploring the influence of gut microbiota, intestinal permeability, and inflammatory pathways. This review also explores the potential therapeutic strategies centered on modulating gut microbiota such as fecal microbiota transplantation; phage therapy; and the use of specific probiotics, prebiotics, and postbiotics in managing NAFLD. By understanding these interactions, we can better comprehend the development and advancement of NAFLD and identify potential therapeutic targets. Full article
Show Figures

Figure 1

Figure 1
<p>This schematic illustrates the complex bidirectional communication between the liver and gut, known as the gut–liver axis. The diagram also highlights the flow of various substances and complex signals through this axis, which plays a crucial role in maintaining homeostasis and influencing disease states. SCFA: Short Chain Fatty Acids; BA: Bile Acids; ROS: Reactive Oxygen Species; PAMPs: Pathogen-Associated Molecular Patterns; DAMPs: Damage-Associated Molecular Patterns; CD4: Cluster of Differentiation.</p>
Full article ">Figure 2
<p>This diagram illustrates the progression of Non-Alcoholic Fatty Liver Disease (NAFLD) from a normal liver to hepatocellular carcinoma (HCC), highlighting the critical role of gut dysbiosis which advances through stages like fatty liver, hepatic steatosis, liver fibrosis, and liver cirrhosis. Gut dysbiosis, marked by decreased beneficial bacteria, namely <span class="html-italic">Firmicutes</span> and <span class="html-italic">Bifidobacterium,</span> and increased harmful bacteria such as <span class="html-italic">E. coli</span> and <span class="html-italic">Proteobacteria</span>, exacerbates these conditions by promoting inflammation and liver damage.</p>
Full article ">Figure 3
<p>The figure illustrates the dynamic interplay and communication mechanisms between the gut microbiota and the liver, highlighting various physiological processes and pathways involved. It also emphasizes how alterations in microbial metabolism and signaling pathways can influence the advancement of NAFLD and its associated complications. FRX: Farnesoid X Receptor; TGR5: Takeda G protein-coupled receptor 5; GPR: G protein Receptors; TNFα: Tumor Necrosis Factor-alpha; Th Cells: T helper cells; AMPK: Adenosine monophosphate-activated protein kinase; CD: Cluster of Differentiation; NH3: Ammonia; OH: Ethanol; PFKB: Phosphofructokinase B; IL1β: Interleukin 1β; NFκB: Nuclear Factor kappa-light-chain-enhancer of activated B cells.</p>
Full article ">
15 pages, 1129 KiB  
Article
In Vitro Utilization of Prebiotics by Listeria monocytogenes
by Tereza Kodešová, Anna Mašlejová, Eva Vlková, Šárka Musilová, Kristýna Horváthová and Hana Šubrtová Salmonová
Microorganisms 2024, 12(9), 1876; https://doi.org/10.3390/microorganisms12091876 - 11 Sep 2024
Viewed by 331
Abstract
Listeria monocytognes is an emerging pathogen responsible for the serious foodborne disease, listeriosis. The commensal gut microbiota is the first line of defense against pathogen internalization. The gut microbiome can be modified by prebiotic substrates, which are frequently added to food products and [...] Read more.
Listeria monocytognes is an emerging pathogen responsible for the serious foodborne disease, listeriosis. The commensal gut microbiota is the first line of defense against pathogen internalization. The gut microbiome can be modified by prebiotic substrates, which are frequently added to food products and dietary supplements. Prebiotics should selectively support the growth of beneficial microbes and thus improve host health. Nevertheless, little is known about their effect on the growth of L. monocytogenes. The aim of this study was to evaluate the growth ability of four L. monocytogenes strains, representing the most common serotypes, on prebiotic oligosaccharides (beta-(1,3)-D-glucan, inulin, fructooligosaccharides, galactooligosaccharides, lactulose, raffinose, stachyose and 2′-fucosyllactose and a mixture of human milk oligosaccharides) as a sole carbon source. The results showed that only beta-(1,3)-D-glucan was metabolized by L. monocytogenes. These cell culture data suggest that beta-(1,3)-D-glucan may not be selectively utilized by healthy commensal bacteria, and its role in intestinal pathogen growth warrants further exploration in vivo. Full article
(This article belongs to the Section Food Microbiology)
Show Figures

Figure 1

Figure 1
<p>The growth curves of <span class="html-italic">Listeria monocytogenes</span> strains cultivated on water-soluble prebiotics (2 g/L). All data for individual strains are averages from triplicates. Positive control = 2 g/L glucose. Residual control = 0.1 g/L glucose. OD = optical density. (<b>A</b>) Data show average of 4 strains: LM1 (serotype 4b), LM11 (serotype 1/2b), LM56 (serotype 1/2a) and LM79 (serotype 1/2c). (<b>B</b>) The growth of individual strains in the positive control (2 g/L glucose). (<b>C</b>) The growth of individual strains in the residual control (0.1 g/L glucose). (<b>D</b>) The growth of individual strains on inulin (2 g/L). (<b>E</b>) The growth of individual strains on fructooligosaccharides (2 g/L). (<b>F</b>) The growth of individual strains on galactooligosaccharides (2 g/L).</p>
Full article ">Figure 2
<p>The growth curves of <span class="html-italic">Listeria monocytogenes</span> strains on beta-(1,3)-D-glucan (partially water-soluble prebiotic) and the positive control. All data for individual strains are averages from triplicates. CFU—colony-forming units. (<b>A</b>) Data show the mean of 4 strains: LM1 (serotype 4b), LM11 (serotype 1/2b), LM 56 (serotype 1/2a) and LM 79 (serotype 1/2c). (<b>B</b>) The growth of individual strains in the positive control (2 g/L glucose). (<b>C</b>) The growth of individual strains on beta-(1,3)-D-glucan (2 g/L).</p>
Full article ">
26 pages, 633 KiB  
Article
Antibacterial Activity and Prebiotic Properties of Six Types of Lamiaceae Honey
by Filomena Nazzaro, Maria Neve Ombra, Francesca Coppola, Beatrice De Giulio, Antonio d’Acierno, Raffaele Coppola and Florinda Fratianni
Antibiotics 2024, 13(9), 868; https://doi.org/10.3390/antibiotics13090868 - 10 Sep 2024
Viewed by 669
Abstract
Our work investigated the antimicrobial and prebiotic properties of basil, mint, oregano, rosemary, savory, and thyme honey. The potential antimicrobial action, assessed against the pathogens Acinetobacter baumannii, Escherichia coli, Listeria monocytogenes, Pseudomonas aeruginosa, and Staphylococcus aureus, evidenced the capacity [...] Read more.
Our work investigated the antimicrobial and prebiotic properties of basil, mint, oregano, rosemary, savory, and thyme honey. The potential antimicrobial action, assessed against the pathogens Acinetobacter baumannii, Escherichia coli, Listeria monocytogenes, Pseudomonas aeruginosa, and Staphylococcus aureus, evidenced the capacity of the honey to influence the pathogenic hydrophobicity and hemolytic activities. Honey inhibited pathogen biofilms, acting especially on the mature biofilms, with inhibition rates of up to 81.62% (caused by the presence of mint honey on L. monocytogenes). S. aureus biofilms were the most susceptible to the presence of honey, with inhibition rates up of to 67.38% in the immature form (caused by basil honey) and up to 80.32% in the mature form (caused by mint honey). In some cases, the amount of nuclear and proteic material, evaluated by spectrophotometric readings, if also related to the honey’s biofilm inhibitory activity, let us hypothesize a defective capacity of building the biofilm scaffold or bacterial membrane damage or an incapability of producing them for the biofilm scaffold. The prebiotic potentiality of the honey was assessed on Lacticaseibacillus casei Shirota, Lactobacillus gasseri, Lacticaseibacillus paracasei subsp. paracasei, and Lacticaseibacillus rhamnosus and indicated their capacity to affect the whole probiotic growth and in vitro adhesive capacity, as well as the antioxidant and cytotoxic abilities, and to inhibit, mainly in the test performed with the L. casei Shirota, L. gasseri, and L. paracasei supernatants, the immature biofilm of the pathogens mentioned above. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>left</b>). Dendrogram of the Lamiaceae kinds of honey obtained using a UPGMA clustering algorithm, taking into account the results shown in <a href="#antibiotics-13-00868-t002" class="html-table">Table 2</a> and <a href="#antibiotics-13-00868-t003" class="html-table">Table 3</a>. (<b>right</b>). Dendrogram of the Lamiaceae kinds of honey obtained using a UPGMA clustering algorithm, taking into account the results shown in <a href="#app1-antibiotics-13-00868" class="html-app">Tables S1 and S2</a>.</p>
Full article ">
19 pages, 3535 KiB  
Article
Nutraceutical Capsules LL1 and Silymarin Supplementation Act on Mood and Sleep Quality Perception by Microbiota–Gut–Brain Axis: A Pilot Clinical Study
by Aline Boveto Santamarina, Victor Nehmi Filho, Jéssica Alves de Freitas, Lucas Augusto Moysés Franco, Joyce Vanessa Fonseca, Roberta Cristina Martins, José Antônio Orellana Turri, Bruna Fernanda Rio Branco da Silva, Arianne Fagotti Gusmão, Eloísa Helena Ribeiro Olivieri, José Pinhata Otoch and Ana Flávia Marçal Pessoa
Nutrients 2024, 16(18), 3049; https://doi.org/10.3390/nu16183049 - 10 Sep 2024
Viewed by 519
Abstract
Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut–brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study [...] Read more.
Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut–brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study aimed to investigate a new nutraceutical formulation comprising prebiotics, minerals, and silymarin’s impact on microbiota, inflammation, mood, and sleep quality. The study evaluated the LL1 + silymarin capsule supplementation over 180 days in overweight adults. We analyzed the fecal gut microbiota using partial 16S rRNA sequences, measured cytokine expression via CBA, collected anthropometric data, quality of life, and sleep questionnaire responses, and obtained plasma samples for metabolic and hormonal analysis at baseline (T0) and 180 days (T180) post-supplementation. Our findings revealed significant reshaping in gut microbiota composition at the phylum, genus, and species levels, especially in the butyrate-producer bacteria post-supplementation. These changes in gut microbiota were linked to enhancements in sleep quality, mood perception, cytokine expression, and anthropometric measures which microbiota-derived short-chain fatty acids might enhance. The supplementation tested in this study seems to be able to improve microbiota composition, reflecting anthropometrics and inflammation, as well as sleep quality and mood improvement. Full article
(This article belongs to the Special Issue Natural Products and Health: 2nd Edition)
Show Figures

Figure 1

Figure 1
<p>Consolidated Standards of Reporting Trials (CONSORT) flowchart of the experimental design.</p>
Full article ">Figure 2
<p>(<b>A</b>) Gut microbiota profile abundance in phyla. (<b>B</b>) Firmicutes/Bacteroidetes ratio. (<b>C</b>) Cladogram from LEfSe pre- and post-supplementation. (<b>D</b>) The logarithmic linear discriminant analysis (LDA) effect size (LEfSe) scores pre- and post-supplementation. (<b>E</b>) Heatmap of the modulated phyla. (<b>F</b>) Alpha (α) diversity indices of Chao1, Observed features, and Simpson index. Values are expressed as the percent of relative abundance (mean ± standard deviation). * <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 3
<p>(<b>A</b>) Heatmap depicting taxonomic readings of volunteers’ microbiomes pre- and post-supplementation. (<b>B</b>) Heatmap of the microbial genera with statistical significance. (<b>C</b>) Differential expression analysis of the genera from gut microbiota. (<b>D</b>) Gut microbiota profile abundance in genera. (<b>E</b>) Heatmap of the microbial species with statistical significance. Values are expressed as the percentage of relative abundance (mean ± standard deviation). * <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 ">
22 pages, 3077 KiB  
Article
The Effects of Single- or Mixed-Strain Fermentation of Red Bean Sourdough, with or without Wheat Bran, on Bread Making Performance and Its Potential Health Benefits in Mice Model
by Chengye Huang, Binle Zhang, Jing Huang, Youyi Liu, Cheng Chen, Jacob Ojobi Omedi, Li Liang, Zhongkai Zhou, Weining Huang and Ning Li
Foods 2024, 13(17), 2856; https://doi.org/10.3390/foods13172856 - 9 Sep 2024
Viewed by 495
Abstract
The effects of single- (Lactobacillus fermentum) or mixed-strain (Lactobacillus fermentum, Kluyveromyces marxianus) fermentation of red bean with or without wheat bran on sourdough bread quality and nutritional aspects were investigated. The results showed that, compared to unfermented controls, [...] Read more.
The effects of single- (Lactobacillus fermentum) or mixed-strain (Lactobacillus fermentum, Kluyveromyces marxianus) fermentation of red bean with or without wheat bran on sourdough bread quality and nutritional aspects were investigated. The results showed that, compared to unfermented controls, the tannins, phytic acid, and trypsin inhibitor levels were significantly reduced, whereas the phytochemical (TPC, TFC, and gallic acid) and soluble dietary fiber were increased in sourdough. Meanwhile, more outstanding changes were obtained in sourdough following a mixed-strain than single-strain fermentation, which might be associated with its corresponding β-glucosidase, feruloyl esterase, and phytase activities. An increased specific volume, reduced crumb firmness, and greater sensory evaluation of bread was achieved after mixed-strain fermentation. Moreover, diets containing sourdough, especially those prepared with mixed-strain-fermented red bean with wheat bran, significantly decreased serum pro-inflammatory cytokines levels, and improved the lipid profile, HDL/LDL ratio, glucose tolerance, and insulin sensitivity of mice. Moreover, gut microbiota diversity increased towards beneficial genera (e.g., Bifidobacterium), accompanied with a greater increase in short-chain fatty acid production in mice fed on sourdough-based bread diets compared to their controls and white bread. In conclusion, mixed-strain fermentation’s synergistic effect on high fiber-legume substrate improved the baking, sensory quality, and prebiotic effect of bread, leading to potential health benefits in mice. Full article
(This article belongs to the Section Food Biotechnology)
Show Figures

Figure 1

Figure 1
<p>The changes in β-glucosidase (<b>a</b>), feruloyl esterase (<b>b</b>), and phytase (<b>c</b>) enzyme activity during single- and mixed-strain sourdough fermentation of red bean flour with or without wheat bran. RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span>; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span>. RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span>. RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span>. Different lower-case and upper-case letters indicated significant differences at <span class="html-italic">p</span> &lt; 0.05 for the same sample at different time of fermentation and different samples at the same time of fermentation, respectively.</p>
Full article ">Figure 2
<p>Effect of single- and mixed-strain-fermented red bean flour with or without wheat bran in bread diets on (<b>a</b>) oral glucose tolerance test, (<b>b</b>) average area under the curve (AUC), (<b>c</b>) fasting plasma insulin, and (<b>d</b>) homeostasis model assessment-insulin resistance (HOMA-IR) test of mice. The bars represented the mean ± SEM (<span class="html-italic">n</span> = 6), with the different letters signifying difference at <span class="html-italic">p</span> &lt; 0.05. WB: wheat bread; RB: red bean flour bread; RWB: red bean–wheat bran bread; RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread. RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> bread. RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread.</p>
Full article ">Figure 3
<p>Effect of bread diets on the relative abundance of gut microbiota at, (<b>a</b>) the phylum level, (<b>b</b>) family level, (<b>c</b>) genus level in healthy mice. Basal: mice fed on AIN-93G diet; WB: wheat bread; RB: red bean flour bread; RWB: red bean–wheat bran bread; RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread; RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread.</p>
Full article ">Figure 3 Cont.
<p>Effect of bread diets on the relative abundance of gut microbiota at, (<b>a</b>) the phylum level, (<b>b</b>) family level, (<b>c</b>) genus level in healthy mice. Basal: mice fed on AIN-93G diet; WB: wheat bread; RB: red bean flour bread; RWB: red bean–wheat bran bread; RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread; RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread.</p>
Full article ">Figure 4
<p>Individual short-chain fatty acids (SCFAs) content (<b>a</b>) and total SCFA content (<b>b</b>) in the feces of the mice following different diet intervention. The bars represented the mean ± SEM (<span class="html-italic">n</span> = 3), with the different letters signifying difference at <span class="html-italic">p</span> &lt; 0.05. WB: wheat bread; RB: red bean flour bread; RWB: red bean–wheat bran bread; RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread; RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread.</p>
Full article ">Figure 5
<p>A schematic diagram showing a regulation model via different diet interventions in mice. Y: single (<span class="html-italic">L. fermentum</span>) strain fermentation; YK: mixed (<span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span>) strain fermentation. (↑) and (↓) represented an increase and decrease in the specific parameter, respectively. RB: red bean substrate; RWB: red bean with wheat bran substrate; RB: red bean flour bread; RWB: red bean–wheat bran bread; RBY: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RBYK: red bean sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread; RWBY: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> bread; RWBYK: red bean–wheat bran sourdough fermented by <span class="html-italic">L. fermentum</span> and <span class="html-italic">K. marxianus</span> bread.</p>
Full article ">
21 pages, 2441 KiB  
Article
Different Physiochemical Properties of Novel Fibre Sources in the Diet of Weaned Pigs Influence Animal Performance, Nutrient Digestibility, and Caecal Fermentation
by Agnieszka Rybicka, Pedro Medel, Emilio Gómez, María Dolores Carro and Javier García
Animals 2024, 14(17), 2612; https://doi.org/10.3390/ani14172612 - 8 Sep 2024
Viewed by 414
Abstract
The effect of including micronised fibre sources (FS) differing in fermentability and hydration capacity (HC) on growth performance, faecal digestibility, and caecal fermentation was investigated in piglets. There were four dietary treatments: a control diet (CON) and three treatments differing in the HC [...] Read more.
The effect of including micronised fibre sources (FS) differing in fermentability and hydration capacity (HC) on growth performance, faecal digestibility, and caecal fermentation was investigated in piglets. There were four dietary treatments: a control diet (CON) and three treatments differing in the HC and fermentability of FS added at 1.5% to prestarter (28–42 d) and starter (42–61 d) diets. These were: LHC (low-HC by-product-based insoluble fibre (IF) with a prebiotic fraction (PF) from chicory root); MHC (medium-HC by-product-based IF with a PF); and HHC (high-HC non-fermentable wood-based IF with no PF). There were eight replicates per treatment. Over the entire period, LHC and MHC piglets showed a 10% increase in daily growth and feed intake (p ≤ 0.019) and tended to have a reduced feed conversion ratio (p = 0.087) compared to HHC piglets. At 42 d, faecal protein digestibility increased by 5% in the LHC and MHC groups compared with the HHC group (p = 0.035) and did not differ from the CON group. Both LHC and MHC fibres were more fermented in vitro with caecal inocula from 61 d old piglets than HHC fibre (p ≤ 0.003). These results suggest that balanced soluble and insoluble fibre concentrates can improve piglet performance. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of dietary treatment on the moisture content of the intestinal digesta (ileum, caecum, colon, and faeces) of 61 d old piglets. CON: basal diet with no additional fibre inclusion, LHC: basal diet including 1.5% low-hydration capacity insoluble fibre with fermentable fraction, MHC: basal diet including 1.5% medium-hydration capacity insoluble fibre with fermentable fraction, and HHC: basal diet including 1.5% high-hydration capacity insoluble fibre; <span class="html-italic">n</span> = 8 for all treatments.</p>
Full article ">Figure 2
<p>Cumulative gas production curves (mL/g DM) of (<b>A</b>) LHC, MHC, HHC, and (<b>B</b>) different micronised fibre sources (chicory root, almond shell, olive kernel, wood, nutshell, and grape by-products) after their incubation with the caecal content from 61 d old piglets fed the control diet. Three different inocula were used, and each inoculum was pooled from the caecal content of 2 piglets fed the control diet. a, b, c: Within the same row, means with different letters differ (<span class="html-italic">p</span> &lt; 0.05; Tukey test). Values in the table indicate the SEM (<span class="html-italic">n</span> = 3) and <span class="html-italic">p</span>-value of the ANOVA analysing potential differences in gas production at each measurement time.</p>
Full article ">Figure 2 Cont.
<p>Cumulative gas production curves (mL/g DM) of (<b>A</b>) LHC, MHC, HHC, and (<b>B</b>) different micronised fibre sources (chicory root, almond shell, olive kernel, wood, nutshell, and grape by-products) after their incubation with the caecal content from 61 d old piglets fed the control diet. Three different inocula were used, and each inoculum was pooled from the caecal content of 2 piglets fed the control diet. a, b, c: Within the same row, means with different letters differ (<span class="html-italic">p</span> &lt; 0.05; Tukey test). Values in the table indicate the SEM (<span class="html-italic">n</span> = 3) and <span class="html-italic">p</span>-value of the ANOVA analysing potential differences in gas production at each measurement time.</p>
Full article ">Figure 3
<p>Cumulative gas production curves (mL/g DM) of (<b>A</b>) LHC: low-hydration capacity insoluble fibre with fermentable fraction, (<b>B</b>) MHC: medium-hydration capacity insoluble fibre with fermentable fraction, and (<b>C</b>) HHC: high-hydration capacity but non-fermentable insoluble fibre, when they were fermented either using caecal inoculum from 61 d old piglets fed a diet including the incubated fibre source (adapted microbiota) or a control diet without fibre sources (non-adapted microbiota). Three different inocula were used for each experimental dietary treatment, and each inoculum was the pooled caecal content from 2 piglets fed the same diet. Tables indicate the SEM (<span class="html-italic">n =</span> 3) and <span class="html-italic">p</span>-value of the ANOVA analysing potential differences in gas production values at each measurement time.</p>
Full article ">Figure 3 Cont.
<p>Cumulative gas production curves (mL/g DM) of (<b>A</b>) LHC: low-hydration capacity insoluble fibre with fermentable fraction, (<b>B</b>) MHC: medium-hydration capacity insoluble fibre with fermentable fraction, and (<b>C</b>) HHC: high-hydration capacity but non-fermentable insoluble fibre, when they were fermented either using caecal inoculum from 61 d old piglets fed a diet including the incubated fibre source (adapted microbiota) or a control diet without fibre sources (non-adapted microbiota). Three different inocula were used for each experimental dietary treatment, and each inoculum was the pooled caecal content from 2 piglets fed the same diet. Tables indicate the SEM (<span class="html-italic">n =</span> 3) and <span class="html-italic">p</span>-value of the ANOVA analysing potential differences in gas production values at each measurement time.</p>
Full article ">
23 pages, 1465 KiB  
Review
Insights into Gut Dysbiosis: Inflammatory Diseases, Obesity, and Restoration Approaches
by Andy Acevedo-Román, Natalia Pagán-Zayas, Liz I. Velázquez-Rivera, Aryanne C. Torres-Ventura and Filipa Godoy-Vitorino
Int. J. Mol. Sci. 2024, 25(17), 9715; https://doi.org/10.3390/ijms25179715 - 8 Sep 2024
Viewed by 949
Abstract
The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut’s barrier function, preventing the invasion of pathogens and maintaining the [...] Read more.
The gut microbiota is one of the most critical factors in human health. It involves numerous physiological processes impacting host health, mainly via immune system modulation. A balanced microbiome contributes to the gut’s barrier function, preventing the invasion of pathogens and maintaining the integrity of the gut lining. Dysbiosis, or an imbalance in the gut microbiome’s composition and function, disrupts essential processes and contributes to various diseases. This narrative review summarizes key findings related to the gut microbiota in modern multifactorial inflammatory conditions such as ulcerative colitis or Crohn’s disease. It addresses the challenges posed by antibiotic-driven dysbiosis, particularly in the context of C. difficile infections, and the development of novel therapies like fecal microbiota transplantation and biotherapeutic drugs to combat these infections. An emphasis is given to restoration of the healthy gut microbiome through dietary interventions, probiotics, prebiotics, and novel approaches for managing gut-related diseases. Full article
(This article belongs to the Special Issue New Molecular Insights into the Gut Microbiome)
Show Figures

Figure 1

Figure 1
<p>Overview of factors that affect the gut microbiota and current available therapies for IBD, namely ulcerative colitis and Crohn’s disease, as well as in <span class="html-italic">Clostridioides difficile</span> infection. Panel (<b>A</b>) summarizes the factors that induce dysbiosis in the microbial gut communities, such as microbiome/host interaction, environmental factors, high-fat diet, and genetic predisposition leading to a reduction in gut alpha diversity. Panel (<b>B</b>) shows available microbiota restoration therapies for UC and CD and treatments against <span class="html-italic">C. difficile</span> colonization (probiotic supplementation, Rebyota/SER-109 Partial Enteral Nutrition, high-fiber prebiotic diet, and fecal microbiota transplants). Created with Biorender.</p>
Full article ">Figure 2
<p>Overview of how diet and the consumption of fiber and fermented foods change the gut microbiota. Panel (<b>A</b>) shows how a high-fat diet induces a reduction in IgA and permeability as well as a reduction in probiotic Bifidobacteria. Panel (<b>B</b>) shows how fiber and fermented foods such as kimchi increase protective bacteria and lean phenotypes. Panel (<b>C</b>) shows how combining diet and probiotics helps decrease inflammation and obese phenotype markers. In the figure, upward green arrows indicate an increase in specific taxa, while red downward arrows indicate a decrease. Created with Biorender.</p>
Full article ">
18 pages, 1470 KiB  
Article
Evaluation of Prebiotic and Health-Promoting Functions of Honeybee Brood Biopeptides and Their Maillard Reaction Conjugates
by Sakaewan Ounjaijean, Supakit Chaipoot, Rewat Phongphisutthinant, Gochakorn Kanthakat, Sirinya Taya, Pattavara Pathomrungsiyounggul, Pairote Wiriyacharee and Kongsak Boonyapranai
Foods 2024, 13(17), 2847; https://doi.org/10.3390/foods13172847 - 7 Sep 2024
Viewed by 374
Abstract
This study addresses the growing interest in natural functional ingredients by evaluating the prebiotic and health-promoting functions of honeybee brood biopeptides (HBb-Bps) and their conjugates. The purpose was to investigate their antioxidant activities, enzyme inhibition properties, and effects on probiotic growth and short-chain [...] Read more.
This study addresses the growing interest in natural functional ingredients by evaluating the prebiotic and health-promoting functions of honeybee brood biopeptides (HBb-Bps) and their conjugates. The purpose was to investigate their antioxidant activities, enzyme inhibition properties, and effects on probiotic growth and short-chain fatty acid (SCFA) production. The HBb-Bps were conjugated with honey, glucose, and fructose via the Maillard reaction. Antioxidant activities were assessed using DPPH and ABTS assays. The inhibitory effects on amylase, pancreatic lipase, and the angiotensin-converting enzyme (ACE) were measured. Probiotic growth and SCFA production were evaluated using L. plantarum TISTR846, and L. lactis TISTR1464. The HBb-Bps and their conjugates exhibited enhanced antioxidant activities post-Maillard reaction. They showed moderate enzyme inhibition, which decreased after conjugation. However, ACE inhibition increased with conjugation. The HBb-Bps significantly promoted probiotic growth and SCFA production, with further enhancement by the Maillard reaction. Overall, the HBb-Bps and their conjugates demonstrate significant prebiotic and health-promoting functions, suggesting their potential as natural ingredients in functional foods and nutraceuticals. Further research should focus on the in vivo effects and, given their solubility and stability these biopeptides could be incorporated into functional food formulations, such as health beverages, protein bars, and other fortified foods designed to deliver specific health benefits. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Antioxidative activities by (<b>a</b>) DPPH and (<b>b</b>) ABTS scavenging assay of individual HBb-Bps, honey, glucose, and fructose solutions at 12.5, 25, 50, and 100 mg/mL. (<b>c</b>) DPPH, and (<b>d</b>) ABTS scavenging assay of the combination between HBb-Bps and honey, glucose, or fructose solutions at 12.5, 25, 50, and 100 mg/mL before and after interaction through spontaneous aging for 20 days under a moist-dry heating process. Different lowercase letters indicate significant differences among the different concentrations (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>In vitro anti-obesity properties by (<b>a</b>) inhibition of α-amylase activity of individual HBb-Bps, honey, glucose, fructose, and acarbose (positive control) solutions at 1, 10, and 100 mg/mL. (<b>b</b>) inhibition of α-amylase activity of the combination of HBb-Bps and honey, glucose, or fructose solutions at 10 mg/mL before and after interaction through spontaneous aging for 20 days under a moist-dry heating process, compared to 10 mg/mL of acarbose as positive control. Different lowercase letters indicate significant differences among the different concentrations (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>In vitro anti-hyperlipidemia properties by (<b>a</b>) inhibition of pancreatic lipase activity of individual HBb-Bps, honey, glucose, fructose, and orlistat (positive control) solutions at 1, 10, and 100 mg/mL. (<b>b</b>) inhibition of pancreatic lipase activity of the combination of HBb-Bps and honey, glucose, or fructose solutions at 10 mg/mL before and after interaction through spontaneous aging for 20 days under a moist-dry heating process, compared to 10 mg/mL of orlistat as positive control. Different lowercase letters indicate significant differences among the different concentrations (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>In vitro anti-hypertensive property by (<b>a</b>) inhibition of ACE activity of individual HBb-Bps, honey, glucose, fructose, and enalapril (positive control) solutions at 1, 10, and 100 mg/mL. (<b>b</b>) Inhibition of ACE activity of the combination of HBb-Bps and honey, glucose, or fructose solutions at 10 mg/mL before and after interaction through spontaneous aging for 20 days under a moist-dry heating process, compared to 10 mg/mL of enalapril as positive control. Different lowercase letters indicate significant differences among the different concentrations (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Prebiotic index (PI) values at 12, 24, and 48 h of <span class="html-italic">L. plantarum</span>, and <span class="html-italic">L. lactis</span> fermentation in the prebiotic-enriched media (HBb-Bps, HBb-Bp honey, FOS, and inulin). The result represents the average of two independent fermentations and triplicate analysis of each sample ± standard deviation. Data represented as mean ± SD (n = 3); lowercase letter (a–c) indicate values within each row with different superscript letters were significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>Production of SCFA (acetic acid, Propionic acid, and butyric acid) after 48 h of <span class="html-italic">L. plantarum</span> and <span class="html-italic">L. lactic</span> fermentation in the control carbon source (dextrose) or prebiotic-enriched media (HBb-Bps, HBb-Bp honey, FOS, and inulin). The result represents the average of two independent fermentations and triplicate analysis of each sample ± standard deviation.</p>
Full article ">
30 pages, 444 KiB  
Review
Nutritional Strategies to Alleviate Stress and Improve Welfare in Dairy Ruminants
by Basiliki Kotsampasi, Maria Anastasia Karatzia, Dimitrios Tsiokos and Stella Chadio
Animals 2024, 14(17), 2573; https://doi.org/10.3390/ani14172573 - 4 Sep 2024
Viewed by 518
Abstract
Dairy ruminants provide a major part of the livestock and agriculture sectors. Due to the increase in world population and the subsequent increase in dairy product demands, the dairy sector has been intensified. Dairy farming intensification and the subsequent increase in animal nutritional [...] Read more.
Dairy ruminants provide a major part of the livestock and agriculture sectors. Due to the increase in world population and the subsequent increase in dairy product demands, the dairy sector has been intensified. Dairy farming intensification and the subsequent increase in animal nutritional demands and the increase in the average global temperature as well have subjected animals to various stress conditions that impact their health and welfare. Various management practices and nutritional strategies have been proposed and studied to alleviate these impacts, especially under heat stress, as well as during critical periods, like the transition period. Some of the nutritional interventions to cope with stress factors and ensure optimal health and production are the inclusion of functional fatty acids and amino acids and feed additives (minerals, prebiotics, probiotics, essential oils and herbs, phytobiotics, enzymes, etc.) that have been proven to regulate animals’ metabolism and improve their antioxidant status and immune function. Thus, these nutritional strategies could be the key to ensuring optimum growth, milk production, and reproduction efficiency. This review summarizes and highlights key nutritional approaches to support the remarkable metabolic adaptations ruminants are facing during the transition period and to reduce heat stress effects and evaluate their beneficial effects on animal physiology, performance, health, as well as welfare. Full article
22 pages, 1476 KiB  
Review
Gut Microbiota-Mediated Biotransformation of Medicinal Herb-Derived Natural Products: A Narrative Review of New Frontiers in Drug Discovery
by Christine Tara Peterson
J 2024, 7(3), 351-372; https://doi.org/10.3390/j7030020 - 4 Sep 2024
Viewed by 664
Abstract
The discovery of natural products has been pivotal in drug development, providing a vast reservoir of bioactive compounds from various biological sources. This narrative review addresses a critical research gap: the largely underexplored role of gut microbiota in the mediation and biotransformation of [...] Read more.
The discovery of natural products has been pivotal in drug development, providing a vast reservoir of bioactive compounds from various biological sources. This narrative review addresses a critical research gap: the largely underexplored role of gut microbiota in the mediation and biotransformation of medicinal herb-derived natural products for therapeutic use. By examining the interplay between gut microbiota and natural products, this review highlights the potential of microbiota-mediated biotransformation to unveil novel therapeutic agents. It delves into the mechanisms by which gut microbes modify and enhance the efficacy of natural products, with a focus on herbal medicines from Ayurveda and traditional Chinese medicine, known for their applications in treating metabolic and inflammatory diseases. The review also discusses recent advances in microbiota-derived natural product research, including innovative methodologies such as culturomics, metagenomics, and metabolomics. By exploring the intricate interactions between gut microorganisms and their substrates, this review uncovers new strategies for leveraging gut microbiota-mediated processes in the development of groundbreaking therapeutics. Full article
(This article belongs to the Special Issue Herbal Medicines: Current Advances and Clinical Prospects)
Show Figures

Figure 1

Figure 1
<p>Medicinal herb-derived natural products and the gut microbiota. The gut microbiota mediates the biosynthesis and biotransformation of natural products derived from medicinal herbs to impact both health and disease. Figure created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>; URL accessed on 19 July 2024.</p>
Full article ">Figure 2
<p>Structure of medicinal herb-derived metabolites with gut microbiota-mediated action mechanisms. The compounds displayed are <b>1</b>. sinensetin, <b>2</b>. curcumin, <b>3</b>. urolithin A, <b>4</b>. quercetin, <b>5</b>. gallic acid, <b>6</b>. thymoquinone, <b>7</b>. chebulinic acid, <b>8</b>. isoquercetin, <b>9</b>. ginsenoside Rh4, <b>10</b>. ginsenoside compound K, <b>11</b>. ginsenoside Rb1, <b>12</b>. berberine, <b>13</b>. berbamine, and <b>14</b>. basic polysaccharide structure.</p>
Full article ">
16 pages, 6718 KiB  
Article
Development of Functional Muffins with Fruits of the Chilean Forest (Calafate and Maqui) and Supplemented with Prebiotic Fiber
by Paula García-Milla, Rocío Peñalver and Gema Nieto
Appl. Sci. 2024, 14(17), 7757; https://doi.org/10.3390/app14177757 - 2 Sep 2024
Viewed by 361
Abstract
Inadequate nutrient intake, coupled with increased oxidative stress, leads to an imbalance responsible for the current major diseases. Many foods have traditionally been used as traditional medicine, including native berries from southern Chile. Both Maqui and Calafate possess high antioxidant activity, which grants [...] Read more.
Inadequate nutrient intake, coupled with increased oxidative stress, leads to an imbalance responsible for the current major diseases. Many foods have traditionally been used as traditional medicine, including native berries from southern Chile. Both Maqui and Calafate possess high antioxidant activity, which grants them medicinal power and makes them an excellent alternative for improving health. The aim of this study is to create a functional food with therapeutic properties capable of counteracting oxidative stress and thereby contributing to improving people’s health. To achieve this, a muffin with inulin, Maqui, and Calafate has been developed. The results show that the incorporation of inulin alone increases the fiber content and antioxidant capacity of the muffins; however, Maqui and Calafate contribute significantly more. Furthermore, contents of phenolic compounds are elevated, and an increase in the folic acid content is observed in the samples compared to the control. We can conclude that producing products with inulin, Maqui, and Calafate can be used to enhance the nutritional value and increase the antioxidant activity of bakery products, providing nutrients while also delivering unique characteristics in color, aroma, and flavor, making them appealing to consumers. Full article
Show Figures

Figure 1

Figure 1
<p>Shades of the products analyzed.</p>
Full article ">Figure 2
<p>Descriptive analysis of overall muffin likeability, as reported by volunteers participating in the sensory analysis of the samples. N/I: not informed.</p>
Full article ">Figure 3
<p>Descriptive analysis of the muffin consumption frequency reported by volunteers participating in the sensory analysis of the samples. N/I: not informed.</p>
Full article ">
Back to TopTop