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

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (491)

Search Parameters:
Keywords = bile salts

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
14 pages, 740 KiB  
Article
Growth Characteristics of Sheep-Derived Bacteroides fragilis and Preliminary Research on Effects in Mice and Lambs
by Cheng Cheng, Jinye Du, Jianping Tao and Darong Cheng
Microorganisms 2025, 13(1), 87; https://doi.org/10.3390/microorganisms13010087 (registering DOI) - 4 Jan 2025
Viewed by 338
Abstract
With the growing demand for sheep, the sheep farming industry has developed rapidly. However, lamb diarrhea, a disease with high mortality rates, significantly hampers the industry’s growth. Traditional antibiotic treatments often disrupt the Intestinal microbiota, induce antibiotic resistance, and cause adverse side effects, [...] Read more.
With the growing demand for sheep, the sheep farming industry has developed rapidly. However, lamb diarrhea, a disease with high mortality rates, significantly hampers the industry’s growth. Traditional antibiotic treatments often disrupt the Intestinal microbiota, induce antibiotic resistance, and cause adverse side effects, highlighting the urgent need to develop alternative therapies. Bacteroides fragilis, a candidate next-generation probiotic, has been closely associated with intestinal health. This study investigated the growth characteristics and probiotic effects of a sheep-derived Bacteroides fragilis isolate, focusing on its efficacy in alleviating lamb diarrhea and infectious intestinal diseases. The experiments demonstrated that the Bacteroides fragilis isolate grows well under mildly acidic conditions (pH 6–8), exhibits some tolerance to bile salts, and has survival rates of 38.89% and 92.22% in simulated gastric and intestinal fluids, respectively, indicating its potential as a probiotic. In a mouse model, Bacteroides fragilis intervention significantly alleviated colonic inflammation caused by Enterohemorrhagic Escherichia coli infection, enhanced tight junction protein expression, mitigated oxidative stress, and improved intestinal barrier function, with high-dose interventions showing superior effects. In lamb trials, Bacteroides fragilis intervention stopped diarrhea in four out of five lambs, partially restored intestinal microbiota diversity, and reduced the abundance of potential pathogens such as Aerococcus suis and Corynebacterium camporealensis. Therefore, Bacteroides fragilis exhibited remarkable effects in regulating intestinal homeostasis, alleviating inflammation, and promoting recovery from diarrhea. Full article
(This article belongs to the Special Issue Probiotics: The Current State of Scientific Knowledge)
21 pages, 4832 KiB  
Article
Development and Application of Lactobacillus plantarum PSCPL13 Probiotics in Olive Flounder (Paralichthys olivaceus) Farming
by Muhammad Aleem Abbas, Hae-Jung Kim, Ga-Yeong Lee, Hae-Yeon Cho, Syed Al Jawad Sayem, Eon-Bee Lee, Seung-Jin Lee and Seung-Chun Park
Microorganisms 2025, 13(1), 61; https://doi.org/10.3390/microorganisms13010061 - 2 Jan 2025
Viewed by 310
Abstract
Aquaculture has grown significantly, contributing to global food security and sustainability; however, intensified fish farming has increased disease susceptibility and antibiotic resistance. This study assessed the probiotic potential of Lactobacillus plantarum PSCPL13 (hereafter, PSCPL13), isolated from the intestines of Japanese eels, for enhancing [...] Read more.
Aquaculture has grown significantly, contributing to global food security and sustainability; however, intensified fish farming has increased disease susceptibility and antibiotic resistance. This study assessed the probiotic potential of Lactobacillus plantarum PSCPL13 (hereafter, PSCPL13), isolated from the intestines of Japanese eels, for enhancing the health of olive flounder. After screening 16 Lactobacillus isolates, PSCPL13 was selected because of its potential broad-spectrum antibacterial activity against many pathogens, such as Vibrio and Edwardsiella. This strain exhibited high acid and bile salt tolerance, which is crucial for intestinal survival. Molecular characterisation confirmed PSCPL13 to be L. plantarum. It was resistant to all tested antibiotics and exhibited significant enzyme activity. In vivo assays in olive flounder revealed that the body weight and length of the fish were significantly higher, while the prevalence of pathogens was lower in their gut microbiota. Regarding haematological parameters, the treated groups exhibited lower eosinophil counts and higher IgM levels, suggesting enhancement of the immune system. These findings indicate that PSCPL13 is a potential probiotic that can be used in aquaculture for naturally improving fish health, growth and immunity, in addition to combating antibiotic resistance and reducing environmental impacts. These findings not only highlight the potential of PSCPL13 in sustainable aquaculture but also provide a foundation for the development of future probiotics. Full article
(This article belongs to the Special Issue Aquatic Microorganisms and Their Application in Aquaculture)
Show Figures

Figure 1

Figure 1
<p>In vivo assessment of the toxicity of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13 in fish. The figure provides a flow diagram of the single oral toxicity test in fish.</p>
Full article ">Figure 2
<p>Isolation and characterisation of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13. (<b>A</b>) Scanning electron microscopy of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13. (<b>B</b>) PCR bands of isolated <span class="html-italic">Lactobacillus plantarum</span> PSCPL13.</p>
Full article ">Figure 3
<p>Characterisation of <span class="html-italic">Lactobacillus</span> isolates by 16S rRNA gene sequencing.</p>
Full article ">Figure 4
<p>Antibacterial activity of PSCPL13 against fish pathogens. Symbol descriptions: No growth (−−−), &lt;10<sup>2</sup> CFU/mL (−−), &lt;10<sup>3</sup> CFU/mL (−), &lt;10<sup>5</sup> CFU/mL (+), &lt;10<sup>7</sup> CFU/mL (++) and &lt;10<sup>8</sup> CFU/mL (+++).</p>
Full article ">Figure 5
<p>(<b>A</b>) Acid resistance and (<b>B</b>) bile salt tolerance of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13.</p>
Full article ">Figure 6
<p>Absolute organ weights (g) of male and female rats following oral administration of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13.</p>
Full article ">Figure 7
<p>Body weight changes in olive flounder after single-dose administration.</p>
Full article ">Figure 8
<p>(<b>A</b>) Body length and (<b>B</b>) weight changes in olive flounder after treatment with <span class="html-italic">Lactobacillus plantarum</span> PSCPL13. The asterisk (*) in the figure represents the statistically significant difference between the control and treatment groups at the appropriate time points. Significance was tested by <span class="html-italic">t</span>-test, * is for <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 9
<p>Counts (colony-forming units) of different bacteria on different agar plates under anaerobic and aerobic conditions. (<b>A</b>) Bacterial growth under anaerobic conditions. (<b>B</b>) Bacterial growth on blood agar under aerobic conditions. (<b>C</b>) Bacterial growth on MacConkey agar under aerobic conditions. (<b>D</b>) Bacterial growth on MRS agar under aerobic conditions. The asterisk (*) in the figure represents the statistically significant difference in bacterial count between the control and treatment groups at the appropriate time points. Significance was tested by <span class="html-italic">t</span>-test, * is for <span class="html-italic">p</span> &lt; 0.05.</p>
Full article ">Figure 10
<p>Effects of PSCPL13 on the gut microbiota of olive flounder. (<b>A</b>) Comparison of variations in the gut microbiota of olive flounder. (<b>B</b>) Relative percentages of microbes. The bar indicates the relative abundance of each bacterial genus. The fish in the control group were not treated with PSCPL13, whereas those in the treated group were administered PSCPL13.</p>
Full article ">Figure 11
<p>Effect of PSCPL13 on IgM levels in olive flounder.</p>
Full article ">Figure 12
<p>Overview of the beneficial effects of <span class="html-italic">Lactobacillus plantarum</span> PSCPL13.</p>
Full article ">
14 pages, 1177 KiB  
Review
The Pharmacokinetic Changes in Cystic Fibrosis Patients Population: Narrative Review
by Ayda Awaness, Rania Elkeeb, Sepehr Afshari and Eman Atef
Medicines 2025, 12(1), 1; https://doi.org/10.3390/medicines12010001 - 31 Dec 2024
Viewed by 351
Abstract
Cystic fibrosis (CF) is a rare genetic disorder commonly affecting multiple organs such as the lungs, pancreas, liver, kidney, and intestine. Our search focuses on the pathophysiological changes that affect the drugs’ absorption, distribution, metabolism, and excretion (ADME). This review aims to identify [...] Read more.
Cystic fibrosis (CF) is a rare genetic disorder commonly affecting multiple organs such as the lungs, pancreas, liver, kidney, and intestine. Our search focuses on the pathophysiological changes that affect the drugs’ absorption, distribution, metabolism, and excretion (ADME). This review aims to identify the ADME data that compares the pharmacokinetics (PK) of different drugs in CF and healthy subjects. The published data highlight multiple factors that affect absorption, such as the bile salt precipitation and the gastrointestinal pH. Changes in CF patients’ protein binding and body composition affected the drug distribution. The paper also discusses the factors affecting metabolism and renal elimination, such as drug–protein binding and metabolizing enzyme capacity. The majority of CF patients are on multidrug therapy, which increases the risk of drug–drug interactions (DDI). This is particularly true for those receiving the newly developed transmembrane conductance regulator (CFTR), as they are at a higher risk for CYP-related DDI. Our research highlights the importance of meticulously evaluating PK variations and DDIs in drug development and the therapeutic management of CF patients. Full article
Show Figures

Figure 1

Figure 1
<p>Literature search with excluded and included study numbers of PK in CF.</p>
Full article ">Figure 2
<p>(<b>A</b>) Effects of CYP3A4/5 enzyme substrates and inhibitors on ivacaftor, a CYP3A4/5 enzyme metabolizer. (<b>B</b>) Effects of CYP3A4/5 enzyme inducers on ivacaftor, a CYP3A4/5 enzyme metabolizer.</p>
Full article ">
23 pages, 11006 KiB  
Article
Lactobacillus agilis SNF7 Presents Excellent Antibacteria and Anti-Inflammation Properties in Mouse Diarrhea Induced by Escherichia coli
by Mingque Feng, Jia Cheng, Yalan Su, Jingdi Tong, Xiangfu Wen, Tianxiong Jin, Meiyi Ren, Deyuan Song, Jinshang Song, Xiaohan Li, Qinna Xie and Mingchao Liu
Int. J. Mol. Sci. 2024, 25(24), 13660; https://doi.org/10.3390/ijms252413660 - 20 Dec 2024
Viewed by 381
Abstract
Escherichia coli (E. coli) is a common pathogen that causes diarrhea in newborns and animals. Antibiotics are typically used to treat bacterial diarrhea, a global intestinal health issue. Probiotics have gained interest as a potential substitute for antibiotics in the management [...] Read more.
Escherichia coli (E. coli) is a common pathogen that causes diarrhea in newborns and animals. Antibiotics are typically used to treat bacterial diarrhea, a global intestinal health issue. Probiotics have gained interest as a potential substitute for antibiotics in the management of E. coli-induced diarrhea and present novel therapeutic options. In this study, the probiotic properties of Lactobacillus agilis SNF7 (L. agilis SNF7) isolated from feces were investigated, and whole genome sequencing was performed to evaluate the properties of the strain. Furthermore, we investigated the protective effects of L. agilis SNF7 in a mouse model of E. coli K99 infection. L. agilis SNF7 exhibits a high survival rate in artificial gastroenteric fluid and bile salt environments, along with an antagonistic effect against E. coli O111:K58 (B4), Staphylococcus aureus (S. aureus), and E. coli K99. Multiple genes with probiotic properties, including bacteriostasis, anti-inflammation, antioxidant, CAZyme, and the utilization of carbohydrate compounds, were identified in genome. L. agilis SNF7 prevented the gut barrier from being damaged by E. coli K99, reducing the clinical manifestations of the infection. Furthermore, L. agilis SNF7 reduced the expression of inflammatory cytokines (IL-6, IL-1β, and TNF-α) by inhibiting the phosphorylation of proteins linked to the NF-κB and MAPK signaling pathways. L. agilis SNF7 improved the intestinal microbial barrier, controlled the balance of the intestinal microecology, and reduced the entry of harmful microbes into the intestine. By controlling gut flora and reducing the inflammatory response, L. agilis SNF7 may be able to prevent and treat E. coli K99 infections. The application of L. agilis SNF7 in the creation of probiotic formulations to stop intestinal illnesses brought on by E. coli infections is clarified by this work. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Experimental design and the characteristics of <span class="html-italic">L. agilis</span> SNF7 in vitro. (<b>a</b>) Experimental design; (<b>b</b>) colony morphology; (<b>c</b>) survival of <span class="html-italic">L. agilis</span> SNF7 at different pH levels; (<b>d</b>) survival of <span class="html-italic">L. agilis</span> SNF7 at different bile salt concentrations; (<b>e</b>) survival of <span class="html-italic">L. agilis</span> SNF7 in artificial gastrointestinal fluid; (<b>f</b>) inhibitory effect of <span class="html-italic">L. agilis</span> SNF7 on three strains of pathogenic bacteria; (<b>g</b>) inhibitory effect of <span class="html-italic">L. agilis</span> SNF7 on three strains of pathogenic bacteria after different treatments; (<b>h</b>) hemolysis of <span class="html-italic">L. agilis</span> SNF7 (the picture above is <span class="html-italic">S. aureus</span> and the one below is <span class="html-italic">L. agilis</span> SNF7); (<b>i</b>) growth curve of <span class="html-italic">L. agilis</span> SNF7; (<b>j</b>) acid production curve of <span class="html-italic">L. agilis</span> SNF7; (<b>k</b>) sensitivity of <span class="html-italic">L. agile</span> SNF7 to antibiotics. Compared with the CK group, ns means has no significant difference (<span class="html-italic">p</span> &gt; 0.05), * means significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Genomic predictive function of <span class="html-italic">L. agilis</span> SNF7. (<b>a</b>) Annotation of the genome of <span class="html-italic">L. agilis</span> SNF7 in the GO database; (<b>b</b>) annotation of the genome of <span class="html-italic">L. agilis</span> SNF7 in the KEGG database; (<b>c</b>) annotation of the genome of <span class="html-italic">L. agilis</span> SNF7 in the eggNOG database; (<b>d</b>) annotation of the genome of <span class="html-italic">L. agilis</span> SNF7 in the CAZy database.</p>
Full article ">Figure 3
<p>Effects of <span class="html-italic">L. agilis</span> SNF7 on the physiological indexes of <span class="html-italic">E. coli</span> K99-induced diarrhea mice. (<b>a</b>) Diarrhea rate of mice during the test period; (<b>b</b>) fecal scores of mice during the test period; (<b>c</b>) body weight of mice in each group on day 8 of the experiment; (<b>d</b>) body weight of mice in each group on day 15 of the experiment; (<b>e</b>) changes in body weight of mice during the experiment; (<b>f</b>) spleen index; (<b>g</b>) liver index; (<b>h</b>) intestinal index. Same letters on columns means the difference is not significant (<span class="html-italic">p</span> &gt; 0.05), ns means has no significant difference (<span class="html-italic">p</span> &gt; 0.05). The difference is significant (<span class="html-italic">p</span> &lt; 0.05) when there are no identical letters between the columns.</p>
Full article ">Figure 4
<p>Effects of <span class="html-italic">L. agilis</span> SNF7 on the intestinal physical barrier of <span class="html-italic">E. coli</span> K99-induced diarrhea mice. (<b>a</b>) H&amp;E staining of mouse jejunal tissue (bar = 100 µm); (<b>b</b>) expression of Claudin-1 in mouse jejunal tissue (bar = 100 µm); (<b>c</b>) expression of occludin in mouse jejunal tissue (bar = 100 µm); (<b>d</b>) expression of ZO-1 in mouse jejunal tissue (bar = 100 µm); (<b>e</b>) expression of MUC2 in mouse jejunal tissue (bar = 100 µm); (<b>f</b>) area fraction of Claudin-1; (<b>g</b>) area fraction of occludin; (<b>h</b>) area fraction of ZO-1; (<b>i</b>) area fraction of MUC2. The same letters on the column show the difference is not significant (<span class="html-italic">p</span> &gt; 0.05). The difference is significant (<span class="html-italic">p</span> &lt; 0.05) when there are no identical letters between the columns.</p>
Full article ">Figure 5
<p>Effects of <span class="html-italic">L. agilis</span> SNF7 on <span class="html-italic">E. coli</span>-induced inflammatory factor secretion in mice jejunum tissues. (<b>a</b>) Relative expression of IL-6 mRNA; (<b>b</b>) relative expression of TNF-α mRNA; (<b>c</b>) relative expression of IL-1β mRNA; (<b>d</b>) IL-6 content; (<b>e</b>) TNF-α content; (<b>f</b>) IL-1β content. The same letters on the column show the difference is not significant (<span class="html-italic">p</span> &gt; 0.05). The difference is significant (<span class="html-italic">p</span> &lt; 0.05) when there are no identical letters between the columns.</p>
Full article ">Figure 6
<p>Effect of <span class="html-italic">L. agilis</span> SNF7 on NF-κB and MAPK signaling pathways in <span class="html-italic">E. coli</span> K99-induced diarrhea mice. (<b>a</b>) Protein bands of NF-κB signaling pathways; (<b>b</b>) p-IκBα/IκBα; (<b>c</b>) p-p65/p-65; (<b>d</b>) protein bands of MAPK signaling pathways; (<b>e</b>) p-p38/p38; (<b>f</b>) p-ERK/ERK; (<b>g</b>) p-JNK/JNK. The same letters on the column show the difference is not significant (<span class="html-italic">p</span> &gt; 0.05). The difference is significant (<span class="html-italic">p</span> &lt; 0.05) when there are no identical letters between the columns.</p>
Full article ">Figure 7
<p>Venn diagram, α diversity analysis, and β diversity analysis of cecum microorganisms in each group. (<b>a</b>) OTU Wayne diagram; (<b>b</b>) α diversity analysis; (<b>c</b>) beta-diversity analysis: PCoA analysis; (<b>d</b>) beta-diversity analysis: NMDS analysis. * Compared with the CK group, * means significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 8
<p>Modulation effect of <span class="html-italic">L. agilis</span> SNF7 on the gut microbiota in <span class="html-italic">E. coli</span> K99-induced diarrhea mice. (<b>a</b>) Phyla-level species distribution histogram; (<b>b</b>) phylum-level species composition heat map; (<b>c</b>) genus-level species distribution histogram; (<b>d</b>) generic-level species composition heat map; (<b>e</b>–<b>k</b>) relative abundance of genera with genus-level differences between groups. The same letters on the columns show the difference is not significant (<span class="html-italic">p</span> &gt; 0.05). The difference is significant (<span class="html-italic">p</span> &lt; 0.05) when there are no identical letters between the columns.</p>
Full article ">
8 pages, 1246 KiB  
Short Note
3-(4-Ferrocenyl-1H-1,2,3-triazol-1-yl)cholic Acid
by Valeria D’Annibale, Venanzio Raglione, Francesco Lisi, Elisa Verdirosi, Lorenza Romagnoli, Danilo Dini, Luciano Galantini and Andrea D’Annibale
Molbank 2024, 2024(4), M1940; https://doi.org/10.3390/M1940 - 19 Dec 2024
Viewed by 389
Abstract
Surfactants are very important compounds that are ubiquitous in biological systems and detergents. Among them, ferrocene surfactants are a very valuable class of stimuli-responsive materials since the presence of ferrocene moiety discloses the chance to control and even modify their amphiphilic properties via [...] Read more.
Surfactants are very important compounds that are ubiquitous in biological systems and detergents. Among them, ferrocene surfactants are a very valuable class of stimuli-responsive materials since the presence of ferrocene moiety discloses the chance to control and even modify their amphiphilic properties via a redox-induced change in the surfactant charge. In this paper, we report a new ferrocene-based surfactant: a ferrocene C-3 derivative of cholic acid, a non-classical surfactant. The title compound of this work was meant to show the significant self-assembly behaviour typical of bile salts, improved by the presence of the aromatic ferrocene subunit. We intended to demonstrate that the presence of the redox mediator should provide the derivative with sensitivity to an oxidative stimulus and control over the aggregation properties. The title compound was prepared in two steps from easily accessible precursors, and its optical properties were investigated through UV-Vis absorption spectroscopy. The determination of its critical micellar concentration and redox potential confirmed this derivative’s amphiphilic nature and its tendency to be reversibly oxidized. Full article
(This article belongs to the Section Organic Synthesis and Biosynthesis)
Show Figures

Figure 1

Figure 1
<p>Estimation of the c.m.c. by surface tension measurements, exploring a concentration range within 0–7 × 10<sup>−4</sup> M of derivative <b>4</b>. Linear fit of the data points (red dots) were reported in black.</p>
Full article ">Figure 2
<p>UV-Vis spectrum of compound <b>4</b> (conc. 4 mM) in water buffered with carbonate/bicarbonate.</p>
Full article ">Figure 3
<p>Cyclic voltammetry of <b>4</b> in DCM at different scan rates ([<a href="#B4-molbank-2024-M1940" class="html-bibr">4</a>] = 4 mg/mL).</p>
Full article ">Scheme 1
<p>Synthesis of compound <b>4</b>: <span class="html-italic">i</span>. CuSO<sub>4</sub>·5H<sub>2</sub>O, sodium ascorbate, 7:3 <span class="html-italic">t</span>-BuOH/H<sub>2</sub>O, 60 °C; 82%; <span class="html-italic">ii.</span> 2 N LiOH, MeOH, 50 °C, 63%.</p>
Full article ">
18 pages, 28989 KiB  
Article
In Vitro Evaluation of Probiotic Activities and Anti-Obesity Effects of Enterococcus faecalis EF-1 in Mice Fed a High-Fat Diet
by Hongying Cai, Qingya Wang, Xiling Han, Haiou Zhang, Na Wang, Yuyin Huang, Peilong Yang, Rui Zhang and Kun Meng
Foods 2024, 13(24), 4095; https://doi.org/10.3390/foods13244095 - 18 Dec 2024
Viewed by 448
Abstract
This research sought to assess the anti-obesity potential of Enterococcus faecalis EF-1. An extensive and robust in vitro methodology confirmed EF-1’s significant potential in combating obesity, probably due to its excellent gastrointestinal tract adaptability, cholesterol-lowering property, bile salt hydrolase activity, α-glucosidase inhibition, and [...] Read more.
This research sought to assess the anti-obesity potential of Enterococcus faecalis EF-1. An extensive and robust in vitro methodology confirmed EF-1’s significant potential in combating obesity, probably due to its excellent gastrointestinal tract adaptability, cholesterol-lowering property, bile salt hydrolase activity, α-glucosidase inhibition, and fatty acid absorption ability. Moreover, EF-1 exhibited antimicrobial activity against several pathogenic strains, lacked hemolytic activity, and was sensitive to all antibiotics tested. To further investigate EF-1’s anti-obesity properties in vivo, a high-fat diet (HFD) was used to induce obesity in C57BL/6J mice. Treatment with EF-1 (2 × 109 CFU/day) mitigated HFD-induced body weight gain, reduced adipose tissue weight, and preserved liver function. EF-1 also ameliorated obesity-associated microbiota imbalances, such as decreasing the Firmicutes/Bacteroidetes ratio and boosting the levels of bacteria (Faecalibacterium, Mucispirillum, Desulfovibrio, Bacteroides, and Lachnospiraceae_NK4A136_group), which are responsible for the generation of short-chain fatty acids (SCFAs). Concurrently, the levels of total SCFAs were elevated. Thus, following comprehensive safety and efficacy assessments in vitro and in vivo, our results demonstrate that E. faecalis EF-1 inhibits HFD-induced obesity through the regulation of gut microbiota and enhancing SCFA production. This strain appears to be a highly promising candidate for anti-obesity therapeutics or functional foods. Full article
(This article belongs to the Special Issue Functional Foods and Their Benefits for Health Regulation)
Show Figures

Figure 1

Figure 1
<p>Visual representation of BSH acidity of <span class="html-italic">E. faecalis</span> EF-1.</p>
Full article ">Figure 2
<p>Detection result of hemolysis test.</p>
Full article ">Figure 3
<p>The effect of <span class="html-italic">E. faecalis</span> EF-1 intervention on HFD-induced obese mice. (<b>A</b>) The average food intake across the weeks. (<b>B</b>) Body weight variations throughout the weeks. (<b>C</b>) The organ index (liver, white adipose tissue, kidney, and spleen). CT refers to the control group, HFD to the high-fat diet group, PC to the positive group, and EF-1 to the <span class="html-italic">E. faecalis</span> EF-1 intervention group. <span class="html-italic">n</span> = 8 mice per group. The symbols *, **, and *** indicate significant differences at the <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 levels, respectively, when comparing the HFD and CT groups. Similarly, the symbol # denotes significant differences at <span class="html-italic">p</span> &lt; 0.05 for the comparison between the EF-1 and HFD groups or between the PC and HFD groups.</p>
Full article ">Figure 4
<p><span class="html-italic">E. faecalis</span> EF-1 alleviated serum and liver biochemical indices in obese mice fed a HFD. (<b>A</b>) The role of <span class="html-italic">E. faecalis</span> EF-1 in altering serum biochemical indices. (<b>B</b>) The role of <span class="html-italic">E. faecalis</span> EF-1 in altering liver biochemical indices. (<b>C</b>) The observation of liver tissues stained with Oil Red O, with a scale reference of 100 μm. <span class="html-italic">n</span> = 8 mice per group. The symbols *, **, and *** indicate significant differences at the <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 levels, respectively, when comparing the HFD and CT groups. Similarly, the symbols #, ##, and ### denote significant differences at the same significance levels for the comparison between the EF-1 and HFD groups or between the PC and HFD groups.</p>
Full article ">Figure 5
<p><span class="html-italic">E. faecalis</span> EF-1 intervention alleviated the pathological state of the liver and WATs in mice fed a HFD. H&amp;E staining of liver tissue (<b>A</b>) and WATs (<b>B</b>), with a scale reference of 100 μm. <span class="html-italic">n</span> = 4 mice per group.</p>
Full article ">Figure 6
<p>The effect of <span class="html-italic">E. faecalis</span> EF-1 on the structure of gut microbiota. (<b>A</b>) The Chao 1 index. (<b>B</b>) The ACE index. (<b>C</b>) The Simpson index. (<b>D</b>) The Shannon index. (<b>E</b>) PCoA analysis. (<b>F</b>) The relative abundance of gut microbiota at the phylum level. (<b>G</b>) A heatmap displaying the hierarchical clustering of bacterial genera profiles at the genus level. <span class="html-italic">n</span> = 5 mice per group. Differences that are significant at the <span class="html-italic">p</span> &lt; 0.05 level between EF-1 and HFD groups are marked with #.</p>
Full article ">Figure 7
<p>Effects of <span class="html-italic">E. faecalis</span> EF-1 intervention on SCFA production. <span class="html-italic">n</span> = 5 mice per group. The symbols * and ** indicate significant differences at <span class="html-italic">p</span> &lt; 0.05 and <span class="html-italic">p</span> &lt; 0.01, respectively, when comparing HFD and CT groups. Similarly, the symbols # and ## denote significant differences at the same significance levels for the comparison between the EF-1 and HFD groups.</p>
Full article ">
18 pages, 4040 KiB  
Article
Taurodeoxycholate Aggregation Explored by Molecular Dynamics: Primary-To-Secondary Micelle Transition and Formation of Mixed Micelles with Fatty Acids
by Fatmegyul Mustan, Anela Ivanova and Slavka Tcholakova
Molecules 2024, 29(24), 5897; https://doi.org/10.3390/molecules29245897 - 13 Dec 2024
Viewed by 382
Abstract
Micelles formed by bile salts in aqueous solution are important for the solubilization of hydrophobic molecules in the gastrointestinal tract. The molecular level information about the mechanism and driving forces for primary-to-secondary micelle transition is still missing. In the current study, the micelle [...] Read more.
Micelles formed by bile salts in aqueous solution are important for the solubilization of hydrophobic molecules in the gastrointestinal tract. The molecular level information about the mechanism and driving forces for primary-to-secondary micelle transition is still missing. In the current study, the micelle formation of 50 mM solutions of taurodeoxycholate (TDC) is studied by atomistic molecular dynamics simulations. It is shown that primary micelles with an aggregation number of 8–10 emerge and persist within the first 50 ns. Then, they coalesce to form secondary micelles with an aggregation number of 19 molecules. This transition is governed by hydrophobic interactions, which significantly decrease the solvent-accessible surface area per molecule in the secondary micelles. The addition of monomers of the sodium salt of fatty acids (FAs), as agents aiding hydrophobic drug delivery, to secondary TDC micelles results in the co-existence of mixed FA-TDC and pure FA micelles. The studied saturated FAs, with chain lengths of C14:0 and C18:0, are incorporated into the micelle core, whereas TDC molecules position themselves around the FAs, forming a shell on the micelle surface. In contrast, the tails of the C18:1 unsaturated fatty acid mix homogeneously with TDC molecules throughout the entire micelle volume. The latter creates a very suitable medium for hosting hydrophobic molecules in the micelles containing unsaturated fatty acids. Full article
(This article belongs to the Section Computational and Theoretical Chemistry)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The average cluster size of TDC as a function of simulation time. Average values and standard deviations of the results obtained from two independent simulations are calculated.</p>
Full article ">Figure 2
<p>A schematic illustration of the micellization of the TDC formation of primary and secondary micelles. Cyan is the hydrophilic face; brown is the hydrophobic face of the skeleton; and yellow and pink molecules are TDC monomers released from dimers and associated with two different micelles.</p>
Full article ">Figure 3
<p>(<b>A</b>) The cumulative number RDF in % and (<b>B</b>) distributive RDF normalized to the area under the curve with respect to the geometric center of each micelle for the COG of SO<sub>3</sub> (blue circles) and COG of <span class="html-italic">H</span>1 (red circles) in TDC. The calculations are performed for each micelle separately and averaged over the last 10 ns of the simulation and over the different micelles.</p>
Full article ">Figure 4
<p>(<b>A</b>) The number of intermolecular hydrogen bonds per TDC molecule and (<b>B</b>) the SASA of the hydrophobic parts (steroid skeleton and CH<sub>3</sub> groups) per molecule in the micelles with different aggregation numbers averaged in the period between 37 and 47 ns. The SASA is calculated at a default radius of the solvent probe of 0.14 nm for water molecules.</p>
Full article ">Figure 5
<p>The average size of clusters as a function of the simulation time in the TDC systems with sodium myristate (orange circles), stearate (yellow squares), and oleate (pink rhombi). Average values and standard deviations of the results obtained from two independent simulations are calculated.</p>
Full article ">Figure 6
<p>The RDF normalized to the area under the curve with respect to the geometric center of each micelle for SO<sub>3</sub>, blue circles; for the first 6-membered ring (<span class="html-italic">H</span>1), red circles in TDC; for COO, green crosses; and for C9–C10, pink crosses in FAs, in the systems of (<b>A</b>) TDC + C14:0, (<b>B</b>) TDC + C18:0, and (<b>C</b>) TDC + C18:1. Empty points are in the RDF for TDC micelles without fatty acids. The analyzed atomic groups are colored in the molecular structures shown on the plots. The calculations are carried out for each micelle separately and averaged over the last 10 ns of the simulation and over all different micelles.</p>
Full article ">Figure 7
<p>The illustration of the placement of the various molecules in the mixed micelles of TDC (skeleton, cyan and brown, and taurine chain, green) with (<b>A</b>) myristate in orange, (<b>B</b>) stearate in yellow, and (<b>C</b>) oleate in purple.</p>
Full article ">Figure 8
<p>(<b>A</b>) The RDF between the center of geometry of pairs of the FAs in the systems of TDC with C14:0 (orange circles), C18:0 (yellow squares), and C18:1 (pink rhombi). (<b>B</b>) The full width at half maximum (FWHM) of the RDF profiles between FA-FA, TDC-FA, and TDC-TDC calculated for each micelle separately and averaged.</p>
Full article ">Figure 9
<p>The chemical structures of the studied molecules: (<b>A</b>) taurodeoxycholate, TDC, with notation of the cyclic fragments; (<b>B</b>) myristate, C14:0; (<b>C</b>) stearate, C18:0; and (<b>D</b>) oleate, C18:1.</p>
Full article ">
13 pages, 1897 KiB  
Article
Practical Considerations for Odevixibat Treatment in Patients with Progressive Familial Intrahepatic Cholestasis: A Single-Center Case Series
by Milena Marx, Steffen Hartleif, Johannes Hilberath, Christoph P. Berg, Ilias Tsiflikas, Stephan Singer and Ekkehard Sturm
J. Clin. Med. 2024, 13(24), 7508; https://doi.org/10.3390/jcm13247508 - 10 Dec 2024
Viewed by 534
Abstract
Background: Patients with progressive familial intrahepatic cholestasis (PFIC) experience cholestasis-associated symptoms, including severe pruritus. Odevixibat is an ileal bile acid transporter inhibitor indicated for treatment of PFIC in the European Union and for the treatment of pruritus in PFIC in the United [...] Read more.
Background: Patients with progressive familial intrahepatic cholestasis (PFIC) experience cholestasis-associated symptoms, including severe pruritus. Odevixibat is an ileal bile acid transporter inhibitor indicated for treatment of PFIC in the European Union and for the treatment of pruritus in PFIC in the United States. The aim of the current study was to characterize the real-world effectiveness and safety of odevixibat in patients with PFIC. Methods: This retrospective study included 9 patients with PFIC treated with odevixibat in a single center in Tübingen, Germany. Data were recorded using case report forms. Results: Of the 9 patients (PFIC1, n = 2; PFIC2, n = 7), 5 had improved serum bile acid levels, pruritus, liver function tests, and sleep with odevixibat treatment. Two siblings with periodic relapses of PFIC symptoms also had improved pruritus and sleep within 4 months of treatment. Two siblings with complete loss of bile salt export pump (BSEP) protein did not respond to treatment; both underwent liver transplantation (indications: hepatocellular carcinoma [HCC] manifestation [n = 1] and severe failure to thrive and refractory pruritus [n = 1]). Four patients reported abdominal complaints that were transient or responded to dose reduction; no other safety issues were reported. Conclusions: In this case series, clinical benefits were observed in most patients with PFIC1 and PFIC2 treated with odevixibat. In patients with periodic relapse of PFIC symptoms, ≥3 months of treatment with odevixibat may be required for symptom control. Patients with complete loss of BSEP did not have consistent symptom relief and require careful monitoring. Effectiveness and feasibility results from our cohort demonstrate potential for long-term benefits with odevixibat in real-world treatment of patients with PFIC. Full article
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)
Show Figures

Figure 1

Figure 1
<p>Serum bile acid levels and dosing over time with real-world odevixibat treatment. For patients who participated in the PEDFIC clinical trials, dotted lines indicate time spent in the PEDFIC trials as well as time during real-world odevixibat treatment. Solid lines indicate real-world odevixibat treatment. <sup>a</sup> No BSEP protein expression. <sup>b</sup> Nonfasting sBA was measured as 312 μmol/L at 14.6 months of treatment for patient 7. <sup>c</sup> Outside of PEDFIC trials. BSEP, bile salt export pump; PFIC, progressive familial intrahepatic cholestasis; sBA, serum bile acid.</p>
Full article ">Figure 2
<p>ALT (<b>a</b>) and total bilirubin (<b>b</b>) levels over time with real-world odevixibat treatment. For patients who participated in the PEDFIC clinical trials, dotted lines indicate time spent in the PEDFIC trials as well as time during real-world odevixibat treatment. Solid lines indicate real-world odevixibat treatment. <sup>a</sup> No BSEP protein expression. ALT, alanine aminotransferase; BSEP, bile salt export pump; PFIC, progressive familial intrahepatic cholestasis; TB, total bilirubin.</p>
Full article ">Figure 3
<p>Screening for HCC in a patient with BSEP3 (patient 1). Panel (<b>a</b>): sonography showed focal liver lesions; largest legion in segment 7 with maximum diameter 1.5 cm. Panel (<b>b</b>): contrast-enhanced ultrasound showed enhancement of lesion without evidence of wash-out. Panel (<b>c</b>): liver MRI with gadobutrol showed hypointense signal on T1w (<b>i</b>) and hyperintense signal on T2w (<b>ii</b>). DWI showed increased signal at high <span class="html-italic">b</span>-values (<b>iii</b>) and slightly low signal intensity on ADC map (<b>iv</b>). Lesion showed enhancement on dynamic contrast-enhanced imaging without wash-out in the venous phase (<b>v</b>). Repeat MRI with a hepatospecific paramagnetic gadolinium-based contrast agent, gadoxetate disodium, also showed enhancement in the dynamic contrast-enhanced imaging without wash-out (<b>vi</b>). Panel (<b>c</b>(<b>vi</b>)): subtraction image of the late hepatobiliary phase taken at 25 min. Panel (<b>d</b>): visualization of the specific echogenic ultrasound needle and position of the needle tip in the lesion (<b>i</b>) and gas artifacts along the puncture canal (<b>ii</b>). Panel (<b>e</b>): H&amp;E staining showed severe cholestatic hepatitis with giant cell transformation, hepatocellular and canalicular bilirubinostasis, ductular reaction, and inflammation. Panel (<b>f</b>): five-month follow-up MRI after biopsy showed a new lesion of 10 mm contiguous to the scar zone of the biopsy; slightly hyperintense on T2w (<b>i</b>) and hypointense on T1w (<b>ii</b>) with hyperarterialization on dynamic contrast-enhanced imaging (<b>iii</b>) and wash-out in the hepatobiliary phase ((<b>iv</b>)—transverse plane; (<b>v</b>)—coronal plane). The subtracted transverse plane of the hepatobiliary phase enhanced the wash-out phenomenon (<b>vi</b>). DWI showed increased signal at high <span class="html-italic">b</span>-values (<b>vii</b>) and low signal intensity on ADC map (<b>viii</b>). Panel (<b>g</b>): AFP levels. Panel (<b>h</b>): H&amp;E staining of explanted liver showed hepatoid tumor cell infiltrates compatible with diagnosis of HCC. Scale bar = 200 µm. Arrows indicate lesion location. ADC, apparent diffusion coefficient; AFP, alpha fetoprotein; BSEP, bile salt export pump; DWI, diffusion-weighted imaging; H&amp;E, hematoxylin and eosin; HCC, hepatocellular carcinoma; MRI, magnetic resonance imaging; T1w, T1 weighted; T2w, T2 weighted.</p>
Full article ">
21 pages, 8325 KiB  
Article
Thermodynamics of Micelle Formation of Selected Homologous 7-Alkyl Derivatives of Na-Cholate in Aqueous Solution: Steroid Skeleton and the Alkyl Chain Conformation
by Dileep Kumar and Mihalj Poša
Int. J. Mol. Sci. 2024, 25(23), 13055; https://doi.org/10.3390/ijms252313055 - 4 Dec 2024
Viewed by 533
Abstract
Bile acid salts are steroid biosurfactants that build relatively small micelles compared to surfactants with an alkyl chain due to the rigid conformation of the steroid skeleton. In order to increase the capacity of micellar solubilization of the hydrophobic molecular guest, certain C7 [...] Read more.
Bile acid salts are steroid biosurfactants that build relatively small micelles compared to surfactants with an alkyl chain due to the rigid conformation of the steroid skeleton. In order to increase the capacity of micellar solubilization of the hydrophobic molecular guest, certain C7 alkyl derivatives were synthesized. Namely, introducing an alkyl group in the C7 position of the steroid skeleton results in a more effective increase in the micelle’s hydrophobic domain (core) than the introduction in the C3 position. In comparison, fewer synthetic steps are required than if alkyl groups are introduced into the C12 position of cholic acid in the Grignard reaction. Here, the thermodynamic parameters of micellization (demicellization) of C7 alkyl (number of C atoms in the alkyl group: 2, 3, 4, and 8) derivatives of cholic acid anion in an aqueous solution without additives are examined (which have not yet been determined) in the temperature interval T (10–40) °C. The critical micellar concentration and the change in the standard molar enthalpy of demicellization (hdemic0) are determined by isothermal calorimetric titration (ICT). From the temperature dependence of hdemic0, the change in the standard molar heat capacity of demicellization is obtained (Cdemic0), the value of which is proportional to the hydrophobic surface of the monomer, which in the micellar state is protected from hydrophobic hydration. The values of Cdemic0 indicate that in the case of C7-alkyl derivatives of cholic acid anion with butyl and octyl chains, parts of the steroid skeleton and alkyl chain remain shielded from hydration after disintegration of the micelle. Conformational analysis can show that starting from the C7 butyl chain in the alkyl chain, sequences with gauche conformation are also possible without the formation of steric repulsive strain between the alkyl chain and the steroid skeleton so that the C7 alkyl chain takes an orientation above the convex surface of the steroid skeleton instead of an elongated conformation toward the aqueous solution. This is a significant observation, namely, if the micelle is used as a carrier of a hydrophobic drug and after the breakdown of the micelle in the biological system, the released drug has a lower tendency to associate with the monomer if its hydrophobic surface is smaller, i.e., the alkyl chain is oriented towards the angular methyl groups of the steroid skeleton (the ideal monomer increases the hydrophobic domain of the micelle, but in aqueous solution, it adopts a conformation with the as small hydrophobic surface as possible oriented towards the aqueous solution)—which then does not disturb the passage of the drug through the cell membrane. Full article
(This article belongs to the Section Physical Chemistry and Chemical Physics)
Show Figures

Figure 1

Figure 1
<p>The steroidal skeleton of the cholic acid anion makes this biosurfactant a rigid conformation compared to classical surfactants with an alkyl chain.</p>
Full article ">Figure 2
<p>Tested C7-alkyl derivatives of 5β-cholic acid, when determining the thermodynamic parameters of micellization, their Na salts are applied.</p>
Full article ">Figure 3
<p>An example of the dependence of the change in the standard molar enthalpy of demicellization (<b>A</b>) and the dependence of the logarithm of the cmc value on temperature for 7-OctC (<b>B</b>); <span class="html-italic">T<sub>H</sub></span> = 27 °C.</p>
Full article ">Figure 4
<p><span class="html-italic">T<sub>S</sub></span> temperature for 7-ButC: micelle formation has an enthalpic driving force.</p>
Full article ">Figure 5
<p>Temperature dependence of thermodynamic potentials of demicellization and entropy of demicellization: X = thermodynamic potentials <span class="html-italic">g</span> (dashed curve), <span class="html-italic">h</span> (solid line with black circles) and product of temperature and entropy (solid line with empty circles); example for 7-OctC.</p>
Full article ">Figure 6
<p>In the micellar state, a hydration layer (I) forms around the polar groups of monomers (micellar building units), which remains unchanged after the disintegration of the micelle. In the micellar state, the hydrophobic surface of the bile acid anion’s steroid skeleton forms the micelle’s hydrophobic core (II) and is protected from hydration. During demicellization, a new hydration layer (III) is formed above the hydrophobic surface of the steroid skeleton. In the hydration layer above the hydrophobic surface at low temperatures, it is true that the water molecules immediately above the atoms of the steroid skeleton are more ordered than the water molecules from the bulk solution (they have lower entropy than the bulk water) and have a coiled orientation for building H-bonds with water molecules from the inside (2D HL = two-dimensional representation of the hydration layer). With increasing temperature, the mobility of water molecules from the hydration layer above the hydrophobic surface of the steroid skeleton increases. The exchange frequency of these water molecules with water molecules from the bulk increases (the entropy of water molecules and the entropy of water molecules from the hydration layer become equal), and these water molecules lose their favorable orientation for building the H-bonds.</p>
Full article ">Figure 7
<p>Dependence of the change in the heat capacity of demicellization on the number of carbons of the C7 alkyl chain in the investigated bile salt derivatives.</p>
Full article ">Figure 8
<p>Syn-axial orientation of the methyl group from the C7 ethyl group of the derivative 7-EthC (NP = Newman projection formula and A, B = molecular subgraph).</p>
Full article ">Figure 9
<p>Partial conformation of the steroid skeleton of 7-EthC (NP = Newman projection formula) in which the methyl group from the C7 ethyl group is not in syn-axial orientation (A) with the corresponding axial hydrogens of the steroid skeleton but is oriented towards the interior of the solution (B).</p>
Full article ">Figure 10
<p>Conformation of the C7 propyl group in 7-PropC derivatives when the propyl hydrocarbon chain is in the gauche conformation: hydrophobic hydration decreases, but steric strain increases (NP = Newman projection formula and A, B = molecular subgraph).</p>
Full article ">Figure 11
<p>The conformation of the propyl group in which there is no steric strain (the methyl group and the C7 carbon from the steroid skeleton are in an antiperiplanar (<span class="html-italic">ap</span>) relationship NP6) but the hydrophobic hydration of the C7 propyl group is maximal (A).</p>
Full article ">Figure 12
<p>Partial conformations of 7-ButC, with this cholic acid anion derivative, a gauche conformation of the C7 side chain is possible without inducing a steric strain with the steroid skeleton (NP = Newman projection formula and A, B = molecular subgraph).</p>
Full article ">Figure 13
<p>In the case of C7 alkyl derivatives of the anion of cholic acid, if the alkyl chain contains four or more carbons, then the alkyl chain in partial gauche (synclinal, <span class="html-italic">sc</span>) and antiperiplanar (<span class="html-italic">ap</span>) conformations occupies the space above the convex surface of the steroid skeleton, which reduces the hydrophobic hydration.</p>
Full article ">Figure 14
<p><sup>1</sup>H NMR (400 MHz, DMSO-<span class="html-italic">d</span><sub>6</sub>) of 7-OctC.</p>
Full article ">Figure 15
<p>Reaction enthalpy (<span class="html-italic">Q</span>) vs. the total detergent concentration in the reaction cell (<span class="html-italic">C</span><sub>T</sub>); titration of 135 mM 7-OctC in water into 2 mL water at 10 °C (37 injections of 10 μL aliquots).</p>
Full article ">
13 pages, 2341 KiB  
Article
Combinatorial Treatment with Praziquantel and Curcumin Reduces Clonorchis sinensis Parasite Burden and Clonorchiasis-Associated Pathologies in Rats
by Soon-Ok Lee, Ki Back Chu, Keon-Woong Yoon, Su In Heo, Jin-Ho Song, Jianhua Li, Sung-Jong Hong and Fu-Shi Quan
Pharmaceutics 2024, 16(12), 1550; https://doi.org/10.3390/pharmaceutics16121550 - 3 Dec 2024
Viewed by 672
Abstract
Background/Objectives: Clonorchiasis is a foodborne parasitic disease that can lead to severe biliary fibrosis and cholangiocarcinoma. While praziquantel (PZQ) is available for clonorchiasis treatment, it cannot revert the histopathological damage incurred through parasite-induced fibrosis. Curcumin (CUR) is an emerging experimental drug possessing [...] Read more.
Background/Objectives: Clonorchiasis is a foodborne parasitic disease that can lead to severe biliary fibrosis and cholangiocarcinoma. While praziquantel (PZQ) is available for clonorchiasis treatment, it cannot revert the histopathological damage incurred through parasite-induced fibrosis. Curcumin (CUR) is an emerging experimental drug possessing anti-inflammatory and fibrosis-alleviating effects, thus signifying its potential as an anthelmintic drug. Here, we evaluated the effect of CUR+PZQ combinatorial drug treatment on C. sinensis infection as well as its effect on ameliorating fibrotic tissue damage in rats. Methods: Worm viabilities following CUR and PZQ treatments were confirmed through microscopy and tetrazolium salt absorption. Anthelminthic effect and hepatobiliary damage mitigation in rats were determined by quantifying worm recovery, histopathological staining, and enzyme-linked immunosorbent assay. Results: CUR+PZQ at LD50 doses demonstrated a time- and dose-dependent antiparasitic effect in vitro, which was markedly greater than either drug alone. Rats were infected with C. sinensis, and drugs were administered at 1 and 4 weeks post-infection (wpi) to assess drug-induced changes in worm burden. Significant reductions in worm burden recoveries were observed following CUR+PZQ treatment at both time points, accompanied by markedly reduced serum and mucosal IgG responses. ALT and AST levels were also substantially lower in combinatorial drug treatment groups than controls. Histopathological examinations confirmed that parasite-induced bile duct lumen widening and liver fibrosis were suppressed at 1 wpi, implying that CUR+PZQ co-treatment can alleviate clonorchiasis-associated pathologies. Conclusions: Our findings indicate that CUR+PZQ co-treatment improved parasite clearance and promoted the resolution of hepatobiliary tissue damage resulting from chronic clonorchiasis. Full article
(This article belongs to the Special Issue Plant Extracts and Their Biomedical Applications)
Show Figures

Figure 1

Figure 1
<p>A schematic overview of the experimental scheduling of in vitro and in vivo studies.</p>
Full article ">Figure 2
<p>CsMC and CsNEJ survival upon CUR, PZQ, and CUR+PZQ treatment. The CsMC were treated with CUR, PZQ, and CUR+PZQ for 24 h (<b>A</b>), 48 h (<b>B</b>), and 72 h (<b>C</b>). CsNEJs were also treated with identical drugs for 24 h (<b>D</b>), 48 h (<b>E</b>), and 72 h (<b>F</b>). Experiments were performed in triplicate, and representative dose- and time-dependent data are provided.</p>
Full article ">Figure 3
<p>Fluorescent microscopic analysis of CsMC and CsNEJ viability. The CsMC were treated with LC<sub>50</sub> doses of CUR, PZQ, and CUR+PZQ for 24, 48, and 72 h. Representative images depicting viable (green) and dead (red) CsMC with respective brightfield photographs are provided (<b>A</b>). Viable and dead CsMC are enumerated using brightfield microscopy (<b>B</b>) and fluorescence microscopy (<b>C</b>). Identical approaches were used to enumerate the number of viable CsNEJs. Representative drug-treated CsNEJs are provided (<b>D</b>), along with viability counts using light microscopy (<b>E</b>) and fluorescence-based methods (<b>F</b>). Statistical significance is indicated using asterisks (* <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). Images for (<b>A</b>,<b>D</b>) were acquired under 100× magnification.</p>
Full article ">Figure 4
<p>XTT-based viability evaluation of <span class="html-italic">C. sinensis</span>. Adult <span class="html-italic">C. sinensis</span> worm viabilities were determined using XTT. Parasite viabilities and survival under two different drug doses were determined after 72 h of treatment (<b>A</b>,<b>B</b>). Statistical significances are indicated using asterisks (* <span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Worm burden, antibodies, and liver damage assessment. Samples were harvested following two different treatment time points. Adult <span class="html-italic">C. sinensis</span> worms were recovered from the bile ducts of rats and quantified to calculate worm recovery (<b>A</b>). ELISA was performed using the sera (<b>B</b>) and duodenal samples (<b>C</b>) of rats to determine <span class="html-italic">C. sinensis</span>-specific antibody responses. ALT (<b>D</b>) and AST (<b>E</b>) concentrations in serum samples were evaluated. Statistical significances are indicated using asterisks (* <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01).</p>
Full article ">Figure 6
<p>Liver histopathology assessment. Liver samples were harvested and used to perform histopathological analyses. Representative liver tissue images from each group (<b>A</b>), along with the bile duct dilations of rats, are provided (<b>B</b>). H&amp;E staining showing the presence of <span class="html-italic">C. sinensis</span> worms and inflammatory cellular influx and parasites (<b>C</b>). Bile duct cellular hyperplasia due to chronic clonorchiasis was measured (<b>D</b>). Representative PSR staining images and cholangiofibrosis under different drug treatment conditions (<b>E</b>,<b>F</b>). Statistical significances are indicated using asterisks (* <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). Asterisk, <span class="html-italic">C. sinensis</span> adult; blue arrow, inflammatory cellular influx; red arrow, fibrosis. All images were acquired under 200× magnification.</p>
Full article ">
14 pages, 1375 KiB  
Article
AcrAB Efflux Pump Plays a Crucial Role in Bile Salts Resistance and Pathogenesis of Klebsiella pneumoniae
by Rundong Shu, Ge Liu, Yunyu Xu, Bojun Liu, Zhi Huang and Hui Wang
Antibiotics 2024, 13(12), 1146; https://doi.org/10.3390/antibiotics13121146 - 29 Nov 2024
Viewed by 702
Abstract
Bile salts possess innate antibacterial properties and can cause significant damage to bacteria. To survive in the mammalian gut, Klebsiella pneumoniae has developed mechanisms to tolerate bile salts; however, the specific mechanisms remain unclear. Transposon library screening revealed that the efflux pump AcrAB [...] Read more.
Bile salts possess innate antibacterial properties and can cause significant damage to bacteria. To survive in the mammalian gut, Klebsiella pneumoniae has developed mechanisms to tolerate bile salts; however, the specific mechanisms remain unclear. Transposon library screening revealed that the efflux pump AcrAB is involved in bile salt resistance. acrA and acrB mutants exhibited high sensitivity not only to bile salts but also to SDS and various antibiotics, with a switch-loop, comprising residues G615, F616, A617, and G618, proving to be crucial in this process. A colonization defect of acrA and acrB mutants was demonstrated to be located in the mouse small intestine, where the bile salt concentration is higher compared to the large intestine. Additionally, both acrA and acrB mutants displayed reduced virulence in the Galleria mellonella model. In conclusion, our results suggest that the Resistance-Nodulation-Cell Division efflux pump serves as a critical determinant in the pathogenesis of K. pneumoniae through various aspects. Full article
(This article belongs to the Section Mechanism and Evolution of Antibiotic Resistance)
Show Figures

Figure 1

Figure 1
<p>Bile salt tolerance distribution among <span class="html-italic">K. pneumoniae</span> clinical strains. Overnight culture of each <span class="html-italic">K. pneumoniae</span> clinical strain was transferred to fresh LB broth containing different concentrations of crude bile (2, 4, 6, 8 and 10%). Growth conditions were assessed based on optical density after 16 h of incubation aerobically.</p>
Full article ">Figure 2
<p>The growth of Δ<span class="html-italic">acrA</span> and Δ<span class="html-italic">acrB</span> mutant with crude bile. (<b>A</b>) Growth curve of strains in the presence of crude bile. cultures of the wild-type and candidate mutants were initiated by inoculating them at a 1:100 ratio into fresh LB medium in the presence of 0.5% crude bile. These cultures were then incubated with agitation at 37 °C, and the OD<sub>600</sub> was recorded at the specified time points. (<b>B</b>) Bile salt killing assay. Stationary cultures of wild-type, mutants, and complemented strains were diluted into either saline or saline containing 0.5% crude bile and were subsequently incubated for 1 h. The viable cell counts were determined through serial dilution and plating. Survival rates were calculated by normalizing CFU to the bile salt-treated group. The data represent the means ± SDs of results from three independent experiments. Significance levels are indicated as follows: *, <span class="html-italic">p</span> &lt; 0.05; ****, <span class="html-italic">p</span> &lt; 0.0001; ns, no statistical significance (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 3
<p>Effects of antimicrobial agents on multidrug efflux pump AcrAB mutants. (<b>A</b>) Growth of wild-type, ∆<span class="html-italic">acrA</span>, and ∆<span class="html-italic">acrB</span> mutants in the presence of different bile salts. Overnight cultures of wild-type and the mutants were diluted 1:100 into fresh LB medium with or without 0.5% sodium cholate, deoxidized cholate, or sodium taurocholate, respectively, and incubated with shaking at 37 °C. OD<sub>600</sub> was measured after 10 h. (<b>B</b>) SDS killing assay. Stationary cultures of wild-type and the mutants were diluted into either saline or saline containing 0.5% SDS. After a 1 h incubation, viable cells were enumerated. The survival rate was calculated by normalizing the CFU to the group treated with SDS. (<b>C</b>) The effects of AcrAB on <span class="html-italic">emrB</span> or <span class="html-italic">mdtB</span> gene transcription. RNA was extracted from mid-logarithmic growth phase cells of both the wild-type and mutant strains. The extracted RNA was then subjected to RT-qPCR analysis to assess the expression levels of the <span class="html-italic">emrB</span> and <span class="html-italic">mdtB</span> genes. qRT-PCR was performed and the results were normalized against 16S rRNA as the internal reference. The data are presented as means ± SDs based on the results of three independent experiments. Statistical significance is indicated as follows: **, <span class="html-italic">p</span> &lt; 0.01; ****, <span class="html-italic">p</span> &lt; 0.0001 (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 4
<p>Influence of the switch-loop on bile salt tolerance in <span class="html-italic">K. pneumoniae</span>. Cultures in stationary phase of wild-type, Δ<span class="html-italic">acrB</span> mutant, Δ<span class="html-italic">acrB</span><sup>C</sup>, and partial complemented strain Δ<span class="html-italic">acrB</span>(pLG-1) were diluted into both saline and saline containing 0.5% crude bile. After one-hour incubation, viable cells were counted. The survival rate was calculated by normalizing the CFU to the crude bile-treated group. The data represent the means ± SDs of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05; ns, no statistical significance (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 5
<p>The effect of multidrug efflux pump AcrAB in intestinal colonization. (<b>A</b>) Adult mouse competition assays. A total of 10<sup>8</sup> cells of the wild-type and Δ<span class="html-italic">acrA</span> or Δ<span class="html-italic">acrB</span> mutants were mixed in a 1:1 ratio and intragastrical administered to mice. Fecal pellets were collected from each mouse at the indicated time points and plated onto selective agar plates. The competitive index [<a href="#B28-antibiotics-13-01146" class="html-bibr">28</a>] was calculated as the ratio of mutant to wild-type colonies normalized to the input ratio. The horizontal line represents the mean CI of 3 mice. (<b>B</b>,<b>C</b>) Colonization of <span class="html-italic">K. pneumoniae</span> using an M-E-A-T model. Intestinal tissues were collected from three mice, each measuring approximately 2.0 cm in length. Subsequently, 200 µL of <span class="html-italic">K. pneumoniae</span> cells at a concentration of approximately 2 × 10<sup>7</sup> CFU were added to the inside-out tissues in a 30 mm Petri dish. After a 4 h incubation, the intestinal tissues were homogenized in 3 mL of PBS. Bacteria were enumerated by serial dilution and plated onto selective agar plates. Bacterial CFU in the large intestine (<b>B</b>) and bacterial CFU in the small intestine (<b>C</b>) are shown. ***, <span class="html-italic">p</span> &lt; 0.001; ns, no statistical significance (Student <span class="html-italic">t</span>-test).</p>
Full article ">Figure 6
<p>The effects of AcrAB on the virulence of <span class="html-italic">K. pneumoniae</span>. (<b>A</b>) Survival of <span class="html-italic">G. mellonella</span> larvae after injection with suspensions of wild-type or mutants. Impact of AcrAB on the survival rate of <span class="html-italic">K. pneumoniae</span> in <span class="html-italic">G. mellonella</span> larvae. Larvae were injected with 10 µL of PBS containing a lethal dose of approximately 10<sup>7</sup> CFU of <span class="html-italic">K. pneumoniae</span>. Following the injection, the larvae were incubated at 37 °C in the dark, and the survival rate was recorded at specified time points (<span class="html-italic">n</span> = 20 larvae per group). *, <span class="html-italic">p</span> &lt; 0.05 (Gehan–Breslow–Wilcoxon test). (<b>B</b>) The effect of AcrAB on the expression of the virulence genes of <span class="html-italic">K. pneumoniae</span>. Overnight cultures of the wild-type, Δ<span class="html-italic">acrA</span>, and Δ<span class="html-italic">acrB</span> mutants, each carrying P<span class="html-italic"><sub>ompA</sub></span>-<span class="html-italic">luxCDABE</span>, P<span class="html-italic"><sub>mrkA</sub></span>-<span class="html-italic">luxCDABE</span>, or P<span class="html-italic"><sub>iutA</sub></span>-<span class="html-italic">luxCDABE</span> transcriptional fusion plasmids, were diluted 1:100 into LB broth. The cultures were incubated with shaking at 37 °C, and luminescence was measured at the stationary phase and subsequently normalized to the corresponding OD<sub>600</sub>. Data represents the means ± SDs of three independent experiments. *, <span class="html-italic">p</span> &lt; 0.05, **, <span class="html-italic">p</span> &lt; 0.01, ***, <span class="html-italic">p</span> &lt; 0.001, ****, <span class="html-italic">p</span> &lt; 0.0001, ns, no significance (Student <span class="html-italic">t</span>-test).</p>
Full article ">
28 pages, 2842 KiB  
Article
Evaluation of Probiotic Potential and Functional Properties of Lactobacillus Strains Isolated from Dhan, Traditional Algerian Goat Milk Butter
by Mohamed Cherif Bentahar, Djilali Benabdelmoumene, Véronique Robert, Said Dahmouni, Wasim S. M. Qadi, Zineb Bengharbi, Philippe Langella, Bouasria Benbouziane, Ebtesam Al-Olayan, Esraa Adnan Dawoud Dawoud and Ahmed Mediani
Foods 2024, 13(23), 3781; https://doi.org/10.3390/foods13233781 - 25 Nov 2024
Cited by 1 | Viewed by 716
Abstract
Goat milk butter, locally known as “Dhan”, from the Sfisfa region of Algeria, holds significant cultural and economic value. This study investigates the probiotic properties of lactic acid bacteria (LAB) present in Dhan, focusing particularly on Lactobacillus strains. Molecular identification using 16S rRNA [...] Read more.
Goat milk butter, locally known as “Dhan”, from the Sfisfa region of Algeria, holds significant cultural and economic value. This study investigates the probiotic properties of lactic acid bacteria (LAB) present in Dhan, focusing particularly on Lactobacillus strains. Molecular identification using 16S rRNA revealed a dominance of Levilactobacillus brevis and Lactiplantibacillus plantarum, forming a substantial part of the bacterial profile. Three LAB isolates (DC01-A, DC04, and DC06) were selected from fresh samples, and rigorous analyses were performed to evaluate their probiotic properties. Safety assessments confirmed the absence of gelatinase, DNase, and haemolytic activities in all isolates. The isolates demonstrated high tolerance to bile salts and acidic conditions, along with the ability to survive simulated gastrointestinal digestion. Notably, strain DC06 exhibited exceptional survival at low pH (1.5) and high bile salt concentrations (0.15–0.3%). All isolates showed substantial growth in MRS medium with 2% phenol, although growth was significantly decreased at 5% phenol. Furthermore, our strains exhibited high adhesion rates to various solvents, demonstrating their potential for strong interaction with cell membranes. Specifically, adhesion to chloroform was observed at 98.26% for DC01-A, 99.30% for DC04, and 99.20% for DC06. With xylene, the adhesion rates were 75.94% for DC01-A, 61.13% for DC04, and 76.52% for DC06. The LAB strains demonstrated impressive growth in ethanol concentrations up to 12%, but their tolerance did not exceed this concentration. They also exhibited robust growth across temperatures from 10 °C to 37 °C, with strains DC04 and DC06 able to proliferate at 45 °C, though none survived at 50 °C. Additionally, the isolates showed significant resistance to oxidative stress induced by hydrogen peroxide (H2O2) and displayed medium to high autolytic activity, with rates of 50.86%, 37.53%, and 33.42% for DC01-A, DC04, and DC06, respectively. The cell-free supernatant derived from strain DC04 exhibited significant antimicrobial activity against the tested pathogens, while strain DC06 demonstrated moderate antioxidant activity with the highest DPPH scavenging rate at 68.56%, compared to the probiotic reference strain LGG at 61.28%. These collective findings not only suggest the probiotic viability of LAB strains found in Dhan but also highlight the importance of traditional food practises in contributing to health and nutrition. Consequently, this study supports the potential of traditional Dhan butter as a functional food and encourages further exploration of its health benefits. Full article
(This article belongs to the Section Food Microbiology)
Show Figures

Figure 1

Figure 1
<p>Biosafety evaluation of <span class="html-italic">Lactobacillus</span> strains. (<b>a</b>) DNase activity and (<b>b</b>) haemolytic activity.</p>
Full article ">Figure 2
<p>Antibiotic resistance of <span class="html-italic">Lactobacillus</span> strains to different antibiotics. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>. IMP; Imipenem, K; Kanamycin, LEV; Levromycin, VA; Vancomycin, OX; Oxacillin, CTX; Cefotaxim.</p>
Full article ">Figure 3
<p>Survival rate (%) of <span class="html-italic">Lactobacillus</span> strains at 0.15, 0.30, and 0.50% bile salt. <sup>a–c</sup>: different lowercase letters in superscript indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among bile salt concentration for same strains. <sup>A–D</sup>: different uppercase letters in superscript within indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among strains. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">Figure 4
<p>Auto-aggregation percentages of <span class="html-italic">Lactobacillus</span> strains. <span class="html-italic">Lactobacillus</span> LGG was used as a reference probiotic strain. Data are expressed as mean ± SD (n = 3). <sup>a–c</sup>: different superscript in lowercase letters represent significant differences (<span class="html-italic">p</span> &lt; 0.05) among time for the same strain. <sup>A–D</sup>: different uppercase letters in superscript indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among strains. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">Figure 5
<p>Hydrophobicity percentages of <span class="html-italic">Lactobacillus</span> strains. <span class="html-italic">L. rhamnosus</span> GG was used as a reference probiotic strain. Data are expressed as mean ± SD (n = 3). <sup>A–D</sup>: different superscript in uppercase letters represent significant differences (<span class="html-italic">p</span> &lt; 0.05) among strains for same solvent. <sup>a–c</sup>: different small letters in superscript indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among solvent for same strain. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">Figure 6
<p>Autolytic activity (%) of <span class="html-italic">Lactobacillus</span> strains. Data are expressed as mean ± SD (n = 3). <sup>a–d</sup>: different small letters in superscript indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among strains. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">Figure 7
<p>Antibacterial activity of lactic acid bacteria. (<b>A</b>) <span class="html-italic">Escherichia coli</span> ATCC10536, (<b>B</b>) <span class="html-italic">Pseudomonas aeruginosa</span> ATCC27853, and (<b>C</b>) <span class="html-italic">Staphylococcus aureus</span> ATCC25923. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">Figure 8
<p>Antioxidant activity of <span class="html-italic">Lactobacillus</span> strain cell-free supernatant evaluated using DPPH (%). <sup>a–d</sup>: different small letters in superscript indicate significant differences (<span class="html-italic">p</span> &lt; 0.05) among strains. DC01-A: <span class="html-italic">Levilactobacillus brevis</span>; DC04: <span class="html-italic">Lactiplantibacillus plantarum</span>; DC06: <span class="html-italic">Levilactobacillus brevis</span>.</p>
Full article ">
22 pages, 7027 KiB  
Article
Bilosomes and Niosomes for Enhanced Intestinal Absorption and In Vivo Efficacy of Cytarabine in Treatment of Acute Myeloid Leukemia
by Abdelrahman R. Said, Mona F. Arafa, Walaa A. El-Dakroury, Sultan Alshehri and Gamal M. El Maghraby
Pharmaceuticals 2024, 17(12), 1572; https://doi.org/10.3390/ph17121572 - 23 Nov 2024
Viewed by 492
Abstract
Cytarabine (CTR) is a hydrophilic anticancer drug used to treat leukemia. It suffers from poor permeability and intestinal metabolism, diminishing its oral bioavailability. Background/Objectives: The objective was to develop and evaluate niosomes and bilosomes for enhanced intestinal absorption; hence, oral bioavailability. Results: CTR-loaded [...] Read more.
Cytarabine (CTR) is a hydrophilic anticancer drug used to treat leukemia. It suffers from poor permeability and intestinal metabolism, diminishing its oral bioavailability. Background/Objectives: The objective was to develop and evaluate niosomes and bilosomes for enhanced intestinal absorption; hence, oral bioavailability. Results: CTR-loaded niosomes and bilosomes with vesicle sizes of 152 and 204.3 nm were successfully prepared with acceptable properties. The presence of bile salts increased the zeta potential of bilosomes. The recorded entrapment efficiency of cytarabine was acceptable for such a hydrophilic drug. CTR-bilosomes showed a pH-dependent drug release pattern with preferred release in pH 6.8. Intestinal absorption behavior indicated a site-dependent CTR absorption pattern with unfavorable absorption in the distal intestine. Niosomal and bilosomal formulations enhanced intestinal absorption parameters with evidence for a predominant paracellular absorption mechanism that bypasses intestinal barriers. The investigation of the anti-leukemic effect of niosomal and bilosomal formulations indicated that both formulations ameliorated the blood parameters, reflecting significant improvement in leukemia treatment compared with the drug solution. Pathological examination of blood films revealed decreased blast cells in peripheral blood in groups treated with tested formulations. Methods: Tested formulations were prepared according to the pro-concentrate method and characterized for particle size, zeta potential, entrapment efficiency, and in vitro release. CTR-loaded niosomes and bilosomes were evaluated for enhanced intestinal absorption utilizing the single-pass in situ intestinal perfusion method in rabbits, and the anti-leukemic effect was assessed using the benzene-induced leukemia model in rats. Conclusions: This study introduced surfactant vesicles for enhanced oral bioavailability of CTR. Full article
(This article belongs to the Section Pharmaceutical Technology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>(<b>A</b>) Chromatograms of calibration standards of CTR in different concentrations (2, 4, 6, 8 and 10 μg/mL); and (<b>B</b>) the constructed calibration curve between area under the curve (AUC) of the peak (mAU) and CTR concentration (μg/mL).</p>
Full article ">Figure 2
<p>(<b>A</b>,<b>B</b>) Transmission electron micrographs of CTR-niosomes; (<b>C</b>) histogram representing size distribution of CTR-niosome in diameter frequency (%); (<b>D</b>,<b>E</b>) CTR-bilosomes; and (<b>F</b>) histogram representing size distribution of CTR-bilosome in diameter frequency (%).</p>
Full article ">Figure 3
<p>Release profiles of cytarabine from (<b>a</b>) CTR-niosomes and CTR-bilosomes at pH 6.8 for 10 h; (<b>b</b>) Fitting of release profiles of the tested niosomes and bilosomes at pH 6.8 with Higuchi release kinetic model; (<b>c</b>) the CTR-niosomes at different intestinal pH, including 6.6, 6.8, and 7.4 for 2 h; (<b>d</b>) CTR-bilosomes at different intestinal pH, including 6.6, 6.8, and 7.4 for 2 h; and (<b>e</b>) CTR-niosomes and CTR-bilosomes at continuous pH variation medium for 10 h. * Amounts of CTR release from niosomes are significantly higher than that recorded from bilosomes at corresponding time points.</p>
Full article ">Figure 4
<p>Bar charts of (<b>a</b>) absorptive clearance; (<b>b</b>) L95%; and (<b>c</b>) percent of paracellular pathway in intestinal absorption of CTR passing different segments of GIT in the form of CTR solution (blue), niosomes (CTR-niosomes) (orange), and bilosomes (CTR-bilosomes) (green). Values are expressed in mean ± SD (n = 4). * Significantly different compared to the corresponding CTR solution. <span class="html-italic">p</span> &lt; 0.05 is considered significant.</p>
Full article ">Figure 5
<p>Comparative effect of CTR solution (group C), CTR-niosomes (group D), and CTR-bilosomes (group E) on (<b>a</b>) hemoglobin (Hgb); (<b>b</b>) red blood cell counts (RBCs); (<b>c</b>) hematocrit (Hct); and (<b>d</b>) white blood cell counts (WBCs). All graphs are compared to the healthy group (group A) and the diseased untreated group (group B). Values are expressed in mean ± SD (n = 5). <span class="html-italic">p</span> &lt; 0.05 is considered significant. a: significant compared to group A. b: significant compared to group B. c: significant compared to group C.</p>
Full article ">Figure 6
<p>Blood film pictures represent animals in (<b>A</b>) the normal group; (<b>B</b>) diseased group; (<b>C</b>) the group treated with CTR solution; (<b>D</b>) CTR-niosomes; and (<b>E</b>) CTR-bilosomes.</p>
Full article ">Figure 7
<p>Schematic illustration of experimental design of in vivo assessment of the anti-leukemic effect of CTR solution, CTR-niosomes, and CTR-bilosomes.</p>
Full article ">
19 pages, 2426 KiB  
Article
Assessment of Digestion and Absorption Properties of 1,3-Dipalmitoyl-2-Oleoyl Glycerol-Rich Lipids Using an In Vitro Gastrointestinal Digestion and Caco-2 Cell-Mediated Coupled Model
by Hyeon-Jun Chang, A-Young Lee and Jeung-Hee Lee
Molecules 2024, 29(22), 5442; https://doi.org/10.3390/molecules29225442 - 18 Nov 2024
Viewed by 590
Abstract
The digestion and absorption properties of 1,3-dipalmitoyl-2-oleoyl glycerol (POP)-rich lipids was evaluated using in vitro gastrointestinal digestion and a Caco-2 cell-mediated coupled model. Caco-2 cell viability and monolayer integrity were assessed by an MTT assay and transepithelial electrical resistance. The IC50 for bile [...] Read more.
The digestion and absorption properties of 1,3-dipalmitoyl-2-oleoyl glycerol (POP)-rich lipids was evaluated using in vitro gastrointestinal digestion and a Caco-2 cell-mediated coupled model. Caco-2 cell viability and monolayer integrity were assessed by an MTT assay and transepithelial electrical resistance. The IC50 for bile salts, pancreatin, and free fatty acid (FFA) were 0.22 mM, 0.22 mg/mL, and 1.47 mM, respectively, and no cytotoxicity was observed for bovine serum albumin (0.01–0.20 mM) or triacylglycerol (1.00–10.00 mM). The in vitro-digested POP-rich lipid containing FFA > 2.95 mM caused the disruption of monolayer tight junctions in Caco-2 cells. The major triacylglycerols (TAG) of POP-rich lipids were POP (50.8%), POO (17.8%), POL/OPL/PLO (7.6%), PPO (7.1%), and PLP (6.8%). Following digestion and uptake into Caco-2 cells, the resynthesized TAGs included PPO (20.6%), PPP (15.9%), POO (14.0%), POL/OPL/PLO (12.2%), POP (10.9%), OOO (7.5%), OPO (7.0%), OOL/OLO (6.7%), PLP (3.1%), and PPL (2.2%). The secreted major TAGs were POL/OPL/PLO (50.8%), PPP (11.1%), and OOL/OLO (8.4%), indicating a diverse TAG profile in newly synthesized lipids. This study provides a coupled model for lowering cytotoxicity and maintaining the monolayer in Caco-2 cells, and for evaluating the digestion and absorption properties of functional lipids containing specific fatty acids incorporated into TAG. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Caco-2 cell viabilities after treatment with sodium taurocholate (<b>A</b>), bile salts (<b>B</b>), pancreatin (<b>C</b>), bovine serum albumin (<b>D</b>), triacylglycerol (<b>E</b>), acylglycerol mixture (<b>F</b>), and oleic acid (<b>G</b>). Asterisks indicate a significant difference between the control (untreated group) and each treatment group according to the Dunnett test (*** <span class="html-italic">p</span> &lt; 0.001, <span class="html-italic">n</span> = 3).</p>
Full article ">Figure 2
<p>Recovery of transepithelial electrical resistance (TEER) after treatment with sodium taurocholate (<b>A</b>), bovine serum albumin (<b>B</b>), bile salts (<b>C</b>), and pancreatin (<b>D</b>) for 2 h. <sup>a–d</sup> Means with different letters above the bars at the same time are significantly different at <span class="html-italic">p</span> &lt; 0.05 by Duncan’s multiple range test (<span class="html-italic">n</span> = 2).</p>
Full article ">Figure 3
<p>Recovery of transepithelial electrical resistance (TEER) after treatment with triacylglycerol (<b>A</b>), the acylglycerol mixture (<b>B</b>), and oleic acid (<b>C</b>) for 2 h. <sup>a–e</sup> Means with different letters above the bars at the same time are significantly different at <span class="html-italic">p</span> &lt; 0.05 by Duncan’s multiple range test (<span class="html-italic">n</span> = 2).</p>
Full article ">
24 pages, 3871 KiB  
Article
Screening, Identification and Application of Lactic Acid Bacteria for Degrading Mycotoxin Isolated from the Rumen of Yaks
by Youli Yao, Jinting Luo, Peng Zhang, Yongben Wang, Boyu Lu, Guofang Wu, Jianbo Zhang, Xuan Luo and Lei Wang
Microorganisms 2024, 12(11), 2260; https://doi.org/10.3390/microorganisms12112260 - 7 Nov 2024
Viewed by 889
Abstract
Mycotoxin contamination is a major food safety issue worldwide, posing a serious threat to animal production performance and human health. Lactic acid bacteria are generally regarded as safe fermentation potential probiotics. They have the advantages of low toxicity, small pollution, strong specificity and [...] Read more.
Mycotoxin contamination is a major food safety issue worldwide, posing a serious threat to animal production performance and human health. Lactic acid bacteria are generally regarded as safe fermentation potential probiotics. They have the advantages of low toxicity, small pollution, strong specificity and high safety, and can reduce the contamination of microorganisms and mycotoxins. In this study, we compared the mycotoxin degradation capacity of 15 lactic acid bacteria strains from the rumen of Qinghai yak, through comprehensive analysis, we finally identified the strains as potential probiotics because they have a fast growth speed, strong acid production capacity (pH < 4.5), and they can grow normally in an environment with a pH of 3.5, bile salt concentration of 0.1%, and good self-agglutination and hydrophobicity. It was found that the fermentation group (Pediococcus acidilactici C2, Pediococcus acidilactici E28, Pediococcus pentosaceus A16 and Enterococcus lactis C16) could significantly reduce mycotoxin content, and both the nutritional and fermentation quality of the feed improved after 7 days of fermentation, meaning that the fermentation group can be used as a functional feed additive. Full article
(This article belongs to the Section Food Microbiology)
Show Figures

Figure 1

Figure 1
<p>Growth (<b>A</b>) and acid production (<b>B</b>) curves of LAB strains isolated from the rumen of yaks.</p>
Full article ">Figure 2
<p>Detoxification curves of LAB strains isolated from the rumen of yaks. (<b>A</b>) represents the detoxification curve of the LAB strain(s) for AFB1 toxin; (<b>B</b>) represents the detoxification curve of the LAB strain(s) for DON toxin; (<b>C</b>) represents the detoxification curve of the LAB strain(s) for ZEA toxin; (<b>D</b>) represents the detoxification curve of the LAB strain(s) for GLUS.</p>
Full article ">Figure 3
<p>Growth of LAB strains isolated from rumen of yaks under different pHs (<b>A</b>) and bile salts (<b>B</b>).</p>
Full article ">Figure 4
<p>Self-agglutination (<b>A</b>) and cell surface hydrophobicity (<b>B</b>) of LAB strains isolated from the rumen of yaks. The same letter indicates no significant difference (<span class="html-italic">p</span> &gt; 0.05), and different letters indicate significant difference (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>Phylogenetic tree of LAB strains isolated from rumen of yaks.</p>
Full article ">Figure 6
<p>Changes in mycotoxin content during feed fermentation inoculated with a culture of four strains isolated from rumen of yaks. (<b>A</b>) indicates the change in AFB1 content; (<b>B</b>) indicates the change in DON content; (<b>C</b>) indicates the change in ZEA content; (<b>D</b>) indicates the change in GLUS content. Note: * <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; no * indicates that the difference was not significant. CK represents unfermented feed and serves as the control group without inoculation of yak rumen strains. Z14 represents fermented feed and is the experimental group inoculated with a culture of four strains isolated from rumen of yaks.</p>
Full article ">Figure 7
<p>Changes in chemical parameters during feed fermentation inoculated with a culture of four strains isolated from rumen of yaks. (<b>A</b>) means Lactic Acid (LA); (<b>B</b>) means Propionic Acid (PA); (<b>C</b>) means Acetic Acid (AA); (<b>D</b>) means Butyric Acid (BA). Note: * <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; no * indicates that the difference was not significant.CK represents unfermented feed and serves as the control group without inoculation of yak rumen strains. Z14 represents fermented feed and is the experimental group inoculated with a culture of four strains isolated from rumen of yaks.</p>
Full article ">Figure 8
<p>Non-multidimensional-dimensional scaling analysis during feed fermentation inoculated with a culture of four strains isolated from rumen of yaks. The blue ones represent the experimental group (fermentation group), while the red ones represent the control group (unfermented group). The same pattern in different colors indicates different treatments in the same period. CK represents unfermented feed and serves as the control group without inoculation of yak rumen strains. Z14 represents fermented feed and is the experimental group inoculated with a culture of four strains isolated from rumen of yaks.</p>
Full article ">Figure 9
<p>Bacterial community of feed during fermentation at phylum (<b>A</b>) and genus (<b>B</b>) levels. Note: CK represents unfermented feed and serves as the control group without inoculation of yak rumen strains. Z14 represents fermented feed and is the experimental group inoculated with a culture of 4 strains isolated from rumen of yaks.</p>
Full article ">Figure 10
<p>Lefse (Linear discriminant analysis Effect Size) Analysis.</p>
Full article ">Figure 11
<p>Microorganisms and feed quality correlation analysis.</p>
Full article ">
Back to TopTop