[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
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
remove_circle_outline
remove_circle_outline

Search Results (1,817)

Search Parameters:
Keywords = glucose supplementation

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
21 pages, 6174 KiB  
Article
Probiotics as Renal Guardians: Modulating Gut Microbiota to Combat Diabetes-Induced Kidney Damage
by Saleh Bakheet Al-Ghamdi
Biology 2025, 14(2), 122; https://doi.org/10.3390/biology14020122 - 24 Jan 2025
Viewed by 382
Abstract
Gut microbiota plays a pivotal role in various health challenges, particularly in mitigating diabetes-induced renal damage. Numerous studies have highlighted that modifying gut microbiota is a promising therapeutic strategy for preserving kidney function and mitigating diabetes-related complications. This study aimed to evaluate the [...] Read more.
Gut microbiota plays a pivotal role in various health challenges, particularly in mitigating diabetes-induced renal damage. Numerous studies have highlighted that modifying gut microbiota is a promising therapeutic strategy for preserving kidney function and mitigating diabetes-related complications. This study aimed to evaluate the protective effects of Lactobacillus acidophilus ATCC 4356 supplementations on kidney health in a rat model of diabetes-induced renal damage. Four groups were studied: control, probiotic supplementation, diabetic, and diabetic with probiotic supplementation. Diabetes was induced using a single streptozotocin (STZ) injection after a 12 h fast, and probiotic supplementation (1 × 10⁹ CFU/kg daily) was administered two weeks prior to diabetes induction and continued throughout the experimental period. Weekly assessments included fasting blood glucose, insulin, glycation markers, and kidney function tests. Glucose metabolism and insulin sensitivity were analyzed through oral glucose tolerance test (OGTT) and insulin sensitivity test (IST). The microbiome was analyzed using 16S rRNA gene sequencing to evaluate changes in diversity and composition. Probiotic supplementation significantly enhanced microbial diversity and composition. Alpha diversity indices such as Shannon and Chao1 demonstrated higher values in the probiotic-treated diabetic group compared to untreated diabetic rats. The Firmicutes/Bacteroidetes ratio, a key indicator of gut health, was also restored in the probiotic-treated diabetic group. Results: Probiotic supplementation significantly improved glycemic control, reduced fasting blood glucose levels, and enhanced insulin sensitivity in diabetic rats. Antioxidant enzyme levels, depleted in untreated diabetic rats, were restored, reflecting reduced oxidative stress. Histological analysis showed better kidney structure, reduced inflammation, and decreased fibrosis. Furthermore, the Comet assay results confirmed a reduction in DNA damage in probiotic-treated diabetic rats. Conclusion: Lactobacillus acidophilus ATCC 4356 supplementation demonstrated significant protective effects against diabetes-induced renal damage by restoring gut microbiota diversity, improving glycemic control, and reducing oxidative stress. These findings highlight the potential of targeting the gut microbiota and its systemic effects on kidney health as a therapeutic approach for managing diabetes-related complications. Further research is needed to optimize probiotic treatments and assess their long-term benefits in diabetes management and kidney health. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>,<b>B</b>) The impact of <span class="html-italic">Lactobacillus acidophilus</span> on body weight and insulin levels in diabetic rats. <span class="html-italic">* p</span> &lt; 0.05.</p>
Full article ">Figure 2
<p>(<b>A</b>–<b>F</b>) The influence of <span class="html-italic">Lactobacillus acidophilus</span> on glucose tolerance and insulin sensitivity in T2D rats. <span class="html-italic">* p</span> &lt; 0.05.</p>
Full article ">Figure 3
<p>(<b>A</b>–<b>G</b>) Impact of <span class="html-italic">Lactobacillus acidophilus</span> on kidney function and antioxidant markers in T2D rats. <span class="html-italic">* p</span> &lt; 0.05.</p>
Full article ">Figure 4
<p>(Image (control—LA—T2D—T2D+LA) (<b>A</b>–<b>C</b>)) This figure shows how <span class="html-italic">Lactobacillus acidophilus</span> supplementation protects kidneys against tissue damage in type 2 diabetes mellitus (T2DM) rats. Control (C), <span class="html-italic">Lactobacillus acidophilus</span>, T2DM treated (T2D), and T2DM treated with <span class="html-italic">Lactobacillus acidophilus</span> (T2D+LA) are among the groups shown. Comparative to the normal control group, statistical significance is indicated as <span class="html-italic">p</span> &lt; 0.05. Furthermore, mentioned against the diabetic control group are * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01. Image 1 (Control) in Image (Control–T2D+LA) microscopic images derived from hematoxylin and eosin (H&amp;E)-stained kidney sections. The kidney tissue looks normal. There is no evidence of damage or inflammation among the well-defined and orderly glomeruli. This picture stands for the control or healthy condition. Image 2: (C+LA) This picture resembles the first one (control) rather exactly. The glomeruli or surrounding tissue are not clearly changed or damaged. One could regard it as either normal or almost normal. Third image (T2D): This picture clearly exhibits pathogenic changes. An accumulation of inflammatory cells in the circular area points to the fibrosis or inflammation there. Furthermore, changing in the surrounding tissue are indications of more severe kidney damage than in the first two pictures. Fourth image (T2D+LA): Less dramatic changes are shown in this picture than in Image 3. Though there are still some indications of enlarged intercellular spaces and mild degenerative changes, the glomeruli seem in better condition with better tissue organization. Though not totally normal, this condition is better than in Image 3. Having n = 10 for every group, the data are shown as the median interquartile range (IQR). These results highlight, by lowering structural damage in diabetic kidney tissues, the possible kidney-protective action of <span class="html-italic">Lactobacillus acidophilus</span>.</p>
Full article ">Figure 5
<p>Four separate groups of rats—the control group, the type 2 diabetes (T2D) group, and the type 2 diabetes with <span class="html-italic">Lactobacillus acidophilus</span> treatment (T2D+LA) group—have their tail lengths (µm) shown here. The experimental technique used the overnight alkaline comet assay, which is good in identifying DNA single-stranded breaks, double-stranded breaks, and alkali-labile sites.</p>
Full article ">Figure 6
<p>Bacterial phyla proportions.</p>
Full article ">Figure 7
<p>Shannon diversity index.</p>
Full article ">Figure 8
<p>Firmicutes/Bacteroidetes ratio.</p>
Full article ">Figure 9
<p>Bacterial abundance heatmap.</p>
Full article ">Figure 10
<p>Microbial characteristics.</p>
Full article ">Figure 11
<p><span class="html-italic">Firmicutes</span>/<span class="html-italic">Bacteroidetes</span> proportions.</p>
Full article ">Figure 12
<p>PCoA of microbial communities.</p>
Full article ">
18 pages, 2931 KiB  
Article
E. coli Biomolecules Increase Glycolysis and Invasive Potential in Lung Adenocarcinoma
by Alexis A. Vega, Parag P. Shah, Eric C. Rouchka, Brian F. Clem, Calista R. Dean, Natassja Woodrum, Preeti Tanwani, Leah J. Siskind and Levi J. Beverly
Cancers 2025, 17(3), 380; https://doi.org/10.3390/cancers17030380 - 24 Jan 2025
Viewed by 299
Abstract
Introduction: Recent studies have discovered that lung cancer subtypes possess distinct microbiome profiles within their tumor microenvironment. Additionally, the tumor-associated microbiome exhibits altered bacterial pathways, suggesting that certain bacterial families are more capable of facilitating tumor progression than others. We hypothesize that there [...] Read more.
Introduction: Recent studies have discovered that lung cancer subtypes possess distinct microbiome profiles within their tumor microenvironment. Additionally, the tumor-associated microbiome exhibits altered bacterial pathways, suggesting that certain bacterial families are more capable of facilitating tumor progression than others. We hypothesize that there exists a crosstalk between lung adenocarcinoma (LUAD) cells and bacterial cells. Methods and Materials: RNA sequencing (RNA-seq) was performed on LUAD cell lines to explore the paracrine signaling effects of bacterial biomolecules. Based on our RNA-seq data, we investigated glycolysis by measuring glucose uptake and lactate production, invasive potential through invasion assays, and epithelial-to-mesenchymal transition (EMT) markers. Since lipopolysaccharides (LPS), abundant on the cell walls of Gram-negative bacteria, can activate toll-like receptor 4 (TLR4), we inhibited TLR4 with C34 to assess its relationship with the observed phenotypic changes. To identify the bacterial biomolecules responsible for these changes, we treated the media with RNAse enzyme, charcoal or dialyzed away molecules larger than 3 kDa. Results and Discussion: RNA-seq revealed 948 genes upregulated in the presence of E. coli biomolecules. Among these, we observed increased expression of Hexokinase II (HKII), JUN proto-oncogene, and Snail Family Transcriptional Repressor 1. We verified the elevation of glycolytic enzymes through Western blot and saw elevation of 2-deoxyglucose uptake and lactate production in LUAD cell lines incubated in E. coli biomolecules. In addition to E. coli elevating glycolysis in LUAD cell lines, E. coli exposure enhanced invasive potential as demonstrated by Boyden chamber assays. Notably, inhibition of TLR4 did not reduce the impact of E. coli biomolecules on glycolysis or the invasive potential of LUAD. Modulating the E. coli-supplemented media with RNAse enzyme or dextran-coated charcoal or using a spin column to remove biomolecules smaller than 3 kDa resulted in changes in HKII and Claudin protein expression. These findings suggest a direct relationship between E. coli and LUAD, wherein several cancer hallmarks are upregulated. Future studies should further investigate these bacterial biomolecules and their role in the tumor microenvironment to fully understand the impact of microbial shifts on cancer progression. Full article
(This article belongs to the Special Issue Lung Cancer—Molecular Insights and Targeted Therapies (Volume II))
Show Figures

Figure 1

Figure 1
<p>Differentially expressed genes of LUAD incubated in bacterially supplemented media. (<b>A</b>) Pipeline for analysis of RNA-seq taken from lung adenocarcinoma cell lines after bacterial biomolecule treatment. (<b>B</b>) Venn diagram representing the unique and shared upregulated LUAD genes affected under bacterial supplementation whose adjusted <span class="html-italic">p</span>-value &lt; 0.05 and log2FC &gt; 1.5. Genes related to glycolysis and the Hippo Pathway have been placed in a table below the Venn diagram. (<b>C</b>) Upregulated KEGG pathways in both LUAD cell lines. Transcriptomic data were filtered based on <span class="html-italic">p</span>-value &lt; 0.05 and log2FC &gt; 1.5. Data were then analyzed using BioKEGG (<a href="https://biopython.org/docs/1.76/api/Bio.KEGG.html" target="_blank">https://biopython.org/docs/1.76/api/Bio.KEGG.html</a>, accessed on 20 January 2025) and pathways were ranked by number of transcriptomic data genes associated with a specific pathway. (<b>D</b>) Top 20 KEGG pathways enriched under the presence of <span class="html-italic">E. coli</span> supplementation for A549 and H2009. Adjusted <span class="html-italic">p</span>-values for each comparison included in the figure key. (<b>E</b>) PCA plot of RNAseq data.</p>
Full article ">Figure 2
<p><span class="html-italic">E. coli</span>-supplemented media enhance glycolysis in LUAD. (<b>A</b>) Gene set enrichment analysis (GSEA) plot of representative gene sets in A549 incubated with <span class="html-italic">E. coli</span>-supplemented media for genes involved in glycolysis. (<b>B</b>) Western blot representing multiple glycolytic enzymes. V = vehicle (1:1, RPMI: PBS), B = bacteria (1:1, RPMI: bacterially infused media), and L = LPS (1:1, PRMI: PBS + 5 μM LPS). The uncropped bolts are shown in <a href="#app1-cancers-17-00380" class="html-app">Supplementary Materials</a>. (<b>C</b>) Densitometry analysis of HKI, HKII, PKM2, and LDHA was performed using ImageJ and normalized by GAPDH. Normalized densitometry data were compared against respective LUAD cell line vehicle control. Data were analyzed using one-way ANOVA with Tukey post hoc test; n = 3 for each cell line, ** <span class="html-italic">p</span>-value &lt; 0.0021. (<b>D</b>) Radiolabeled deoxyglucose readings quantified by normalizing scintillation counts per minute (CPM) to protein concentration. Data were analyzed using unpaired <span class="html-italic">t</span>-test; n = 3 for each cell line, * <span class="html-italic">p</span>-value &lt;0.0332, ** <span class="html-italic">p</span>-value &lt; 0.0021. (<b>E</b>) Lactate production of LUAD cell lines incubated in vehicle or <span class="html-italic">E. coli</span> biomolecules measured by luminescence and normalized by cell count. Relative luminescence units (RLUs) were analyzed using unpaired <span class="html-italic">t</span>-test; n = 3 for each cell lines, ** <span class="html-italic">p</span>-value &lt; 0.0021, *** <span class="html-italic">p</span>-value &lt; 0.0002.</p>
Full article ">Figure 3
<p><span class="html-italic">E. coli</span> takes up LUAD produced lactate. (<b>A</b>) Relative luminescence units (RLUs) of lactate levels in media after 24 h of incubation without <span class="html-italic">E. coli</span> (media) and with <span class="html-italic">E. coli</span> (media + <span class="html-italic">E. coli</span>). Media were prepared by incubated RPMI-1640 with no LUAD (circle), A549 (square), or H2009 (triangle) for 24 h prior to addition of <span class="html-italic">E. coli</span>. Data were analyzed using two-way ANOVA with Sidak post hoc test (n = 3, **** <span class="html-italic">p</span>-value &lt; 0.0001) with comparison of respective media with and without <span class="html-italic">E. coli</span>. (<b>B</b>) mM glucose levels or RPMI media and RPMI media following 24 h incubation with LUAD, as determined through glucose-glo assay. (<b>C</b>) RLU of lactate levels in media after 8 h incubation with <span class="html-italic">E. coli</span> with LUAD media (LUAD + <span class="html-italic">E. coli</span>) or LUAD media supplemented with glucose to a final concentration of 10 mM glucose (LUAD media + glucose + <span class="html-italic">E. coli</span>). RLUs were normalized against lactate levels in LUAD media and analyzed using one-way ANOVA with Sidak post hoc test (n = 3, *** <span class="html-italic">p</span>-value &lt; 0.0002, **** <span class="html-italic">p</span>-value &lt; 0.0001). (<b>D</b>) RLU of glucose levels in media after 8 h incubation with <span class="html-italic">E. coli</span> with RPMI-1640 media (media + <span class="html-italic">E. coli</span>) or LUAD media (LUAD + <span class="html-italic">E. coli</span>). RLUs were normalized against glucose levels in LUAD media and analyzed using one-way ANOVA with Sidak post hoc test (n = 3, ** <span class="html-italic">p</span>-value &lt; 0.0021, **** <span class="html-italic">p</span>-value &lt; 0.0001). (<b>E</b>) RLUs of glutamine levels in media after 24 h incubation with A549 (cancer) and incubation of LUAD media with <span class="html-italic">E. coli</span> for 24 h (cancer -&gt; <span class="html-italic">E. coli</span>). RLUs were compared against glutamine levels found in RPMI-1640 (media) and analyzed using one-way ANOVA with Sidak post hoc test (n = 3, **** <span class="html-italic">p</span>-value &lt; 0.0001).</p>
Full article ">Figure 4
<p><span class="html-italic">E. coli</span>-supplemented media enhance invasive potential of LUAD cell lines. (<b>A</b>) Gene set enrichment analysis (GSEA) plot of representative gene sets in A549 incubated with <span class="html-italic">E. coli</span>-supplemented media for genes involved in epithelial-to-mesenchymal transition (EMT). (<b>B</b>) Western blot representing multiple genes involved in EMT for various LUAD cell lines incubated in vehicle (1:1, RPMI: PBS), bacteria (1:1, RPMI: bacterially infused media), and LPS (1:1, PRMI: PBS + 0.5 µg/mL LPS). Densitometry analysis was performed using ImageJ and data were normalized to GAPDH. Normalized values were compared against respective vehicle controls. The uncropped bolts are shown in <a href="#app1-cancers-17-00380" class="html-app">Supplementary Materials</a>. (<b>C</b>) Representative images of wound-healing assay to determine the chemo-attractant potential of <span class="html-italic">E. coli</span> biomolecules. After scratch was made, Matrigel infused with <span class="html-italic">E. coli</span>-supplemented media was layered on top and images were taken across multiple days. (<b>D</b>) Data analysis of wound-healing assay (n = 3) of LUAD cells in either vehicle (1:1, RPMI: PBS) or media supplemented with <span class="html-italic">E. coli</span> (1:1, RPMI: <span class="html-italic">E. coli</span> media). Data were analyzed using two-way ANOVA with Sidak post hoc test. (<b>E</b>) Data of single-celled LUAD motility in either vehicle or <span class="html-italic">E. coli</span> after 48 h (n = 20) using Keyence Live Cell Imager. Data were analyzed using unpaired <span class="html-italic">t</span>-test. (<b>F</b>) Invasion assay representation of LUAD cell lines incubated with serum-starved vehicle or <span class="html-italic">E. coli</span>. 10% FBS serum was used as the chemo-attractant. (<b>G</b>) Data analysis of invasion assay (n = 8) of LUAD cells in either vehicle or <span class="html-italic">E. coli</span>. Invasion assay data were normalized by average invasion in vehicle and analyzed using Student’s <span class="html-italic">t</span>-test, **** <span class="html-italic">p</span>-value &lt; 0.0001.</p>
Full article ">Figure 5
<p>TLR4 inhibition does not impact <span class="html-italic">E. coli</span>-influenced glycolysis. (<b>A</b>) AlamarBlue viability assay of LUAD cell lines following treatment of increasing levels of TLR4 inhibitor, C34. LUAD cell lines were incubated for 24 h prior to incubation with alamarBlue. RFUs attained were normalized for each LUAD cell line by RFUs at lowest concentration (0.01 µM C34). (<b>B</b>) Western blot representation of A549 cell lines incubated with increasing concertation of C34. Membrane was blotted for p-MAPK and MAPK with GAPDH as a loading control along with anti-rabbit secondary. Densitometry was measured using ImageJ, normalizing by GAPDH and further normalizing p-MAPK by respective MAPK. Normalized values were compared to p-MAPK densitometry values of vehicle without C34. The uncropped bolts are shown in <a href="#app1-cancers-17-00380" class="html-app">Supplementary Materials</a>. (<b>C</b>) Western blot representation of A549 cell lines incubated with <span class="html-italic">E. coli</span> biomolecules (<span class="html-italic">E. coli</span>)/without (vehicle) and with/without 5 µM C34. Membrane was blotted for HKII, with GAPDH used as a loading control. Densitometry was analyzed using ImageJ, with values normalized to GAPDH and normalized values relative to vehicle without 5 µM C34. (<b>D</b>) Relative luminescent units (RLUs) of lactate measurement in media of A549, H2009, and PC9 incubated with <span class="html-italic">E. coli</span> biomolecules with and without 5 µM C34. Data were analyzed using unpaired <span class="html-italic">t</span>-test, n = 3.</p>
Full article ">Figure 6
<p>Treatment of <span class="html-italic">E. coli</span>-supplemented media further impacts LUAD. (<b>A</b>) Western blot representation of A549 incubated in vehicle or <span class="html-italic">E. coli</span>-supplemented media untreated (V) or treated with RNAse (R) or dextran-coated charcoal (C) or dialyzed to remove molecules &lt;3 kDa (D). Membrane was blotted for HKII, CJUN, and Claudin, with GAPDH used as a loading control. Densitometry was analyzed using ImageJ, wherein densitometry values were normalized to respective GAPDH and normalized values were compared against A549 incubated in untreated vehicle media. The uncropped bolts are shown in <a href="#app1-cancers-17-00380" class="html-app">Supplementary Materials</a>. (<b>B</b>) RT-qPCR analysis of A549 cells incubated in vehicle or <span class="html-italic">E. coli</span>-supplemented media (bacteria) untreated (vehicle) or treated with dextran-coated charcoal or dialyzed to remove molecules &lt;3 kDa. Data were analyzed using a two-way ANOVA with a Tukey post hoc test (n = 3, ** <span class="html-italic">p</span>-value &lt; 0.0021, *** <span class="html-italic">p</span>-value &lt; 0.0002, and **** <span class="html-italic">p</span>-value &lt; 0.0001). (<b>C</b>) Graphical representation of <span class="html-italic">E. coli</span> secreting biomolecules which LUAD can take up, resulting in altered phenotypes.</p>
Full article ">
19 pages, 2810 KiB  
Article
Apple Polyphenol Mitigates Diabetic Nephropathy via Attenuating Renal Dysfunction with Antioxidation in Streptozotocin-Induced Diabetic Rats
by Chieh-Yu Wang, Dai-Lin Wu, Meng-Hsun Yu, Chih-Ying Wang, Hsin-Wen Liang and Huei-Jane Lee
Antioxidants 2025, 14(2), 130; https://doi.org/10.3390/antiox14020130 - 23 Jan 2025
Viewed by 353
Abstract
Diabetic nephropathy (DN) is a major cause of morbidity and mortality among patients with diabetes mellitus (DM). Studies have highlighted the critical role of reactive oxygen species (ROS) in the pathogenesis of DM and its complications. Apple polyphenol (AP) has demonstrated antioxidant properties [...] Read more.
Diabetic nephropathy (DN) is a major cause of morbidity and mortality among patients with diabetes mellitus (DM). Studies have highlighted the critical role of reactive oxygen species (ROS) in the pathogenesis of DM and its complications. Apple polyphenol (AP) has demonstrated antioxidant properties in various models. In this study, we investigated the effects of AP on DN in a rat model. Type 1 diabetes was induced in Sprague–Dawley rats via a single intraperitoneal injection of streptozotocin (65 mg/kg) (n = 8). Rats with blood glucose levels exceeding 250 mg/dL were treated with AP at dosages of 0.5%, 1%, or 2% (w/w) in drinking water for 10 weeks. AP administration significantly improved early-stage DN markers, including reductions in the blood urea nitrogen-to-creatinine ratio and the urinary albumin-to-creatinine ratio (ACR), in a dose-dependent manner. AP treatment also significantly lowered blood triglyceride levels and reduced lipid peroxidation in kidney tissues. Histological analysis revealed that AP attenuated renal hydropic change, reduced glomerular basement membrane thickening, and restored mitochondrial morphology in diabetic rats. Additionally, the upregulation of transforming growth factor-beta (TGF-β) observed in the diabetic kidney was attenuated by AP treatment. In H2O2-stimulated rat mesangial cells, AP reduced ROS levels, accompanied by a reduction in TGF-β expression. These findings suggest that AP exerts protective effects against DN by improving renal function and mitigating oxidative stress, indicating its potential as a nutraceutical supplement for slowing DN progression. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Effect of AP on kidney damage in diabetic rats. (<b>A</b>) experimental design. SD rats were assigned into the following groups: control; citrate buffer; 2% AP only; 65 mg/kg of STZ; STZ and administered with 0.5%, 1%, or 2% of AP. After being treated with AP for 10 weeks, the kidneys were removed, and the morphology is shown in (<b>B</b>), upper panels; H&amp;E stains were performed and the histological changes are shown in (<b>B</b>), lower panels. Blue arrowheads point the hydropic changes, shown as a pale and swollen change of the proximal convoluted tubules; black arrowheads represent the GBM, shown as a white edge surrounding the glomeruli. Scale bar: 100 mm. (<b>C</b>), quantification of the rate of hydropic change; results were represented as mean ± SD; values not sharing a common letter in the same row are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>The effect of AP on the glomerulus and mitochondria profile. Electron microscopy analysis was performed to examine the glomerulus and mitochondria in kidney sections from the animals. (<b>A</b>), upper panels display the histological analysis of glomeruli. CL, capillary lumen; MC, mesangial cell; MM, mesangial matrix; red arrows indicate the podocyte foot processes; black arrows indicate partial foot process effacement; the red asterisk shows the glomerular basement membrane; scale bar, 2 mm; lower panels display the mitochondrial morphology in the kidney. The black asterisk shows the damaged mitochondria in the kidneys; scale bar, 0.5 mm. (<b>B</b>), quantification of the thickness of GBM; results are represented as mean ± SD; values not sharing a common letter in the same row are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>AP reduced TGF-β expression in diabetic kidneys. Immunohistological analysis was performed to evaluate TGF-β expression in rat kidneys. In addition to analyzing kidney samples from animals treated for 10 weeks, the TGF levels were also compared in kidney samples from animals treated for 4 weeks. (<b>A</b>), upper panels, 4 weeks; lower panels, 10 weeks. Black arrows point to positive-stained TGF-β in the proximal convoluted tubules. Blue arrows point TGF-β in the glomeruli. Scale bar, 200 mm. (<b>B</b>), quantification results showing the expressed intensity of TGF-β. Results are represented as mean ± SD; values not sharing a common letter in the same row are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>(<b>A</b>), cell viability: AP reduced oxidative status/TGF-β and restored mitochondrial function in H<sub>2</sub>O<sub>2</sub>-induced RMCs. RMCs at a concentration of 1 × 10<sup>5</sup> cells/mL were treated with 500 mM of H<sub>2</sub>O<sub>2</sub> or co-treated with AP for 24 h. Results from three independent experiments are represented as mean ± SD; values not sharing a common letter in the same row are significantly different (<span class="html-italic">p</span> &lt; 0.05); (<b>B</b>), the DCF-DA intensity detected by FACScan cytometry: the area to the right of the red dashed line was defined as exhibiting ROS signals; (<b>C</b>), JC-1-stained cells were analyzed using fluorescence microscopy with the quantified result. Magnification: ×200; (<b>D</b>), the protein level of TGF-b with the quantified results. The quantified results of JC-1 and TGF-b examinations from three independent experiments are represented as mean ± SD; values within the same row that do not share a common letter are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>In the models of STZ-induced SD rats and H<sub>2</sub>O<sub>2</sub>-treated rat mesangial cells, AP mitigates DN by improving renal function and mitigating oxidative stress. This illustration was created in BioRender. Lee, H. (2024) <a href="http://BioRender.com/y22r012" target="_blank">http://BioRender.com/y22r012</a>, accessed on 13 November 2024.</p>
Full article ">
15 pages, 1621 KiB  
Article
Effects of Supplementing Rumen-Protected Glutathione on Lactation Performance, Nutrients, Oxidative Stress, Inflammation, and Health in Dairy Cows During the Transition Period
by Yu Hao, Xuejie Jiang, Rui Sun, Yunlong Bai, Chuang Xu, Yuxi Song and Cheng Xia
Vet. Sci. 2025, 12(2), 84; https://doi.org/10.3390/vetsci12020084 - 23 Jan 2025
Viewed by 326
Abstract
Glutathione (GSH), widely present in plant and animal cells and crucial for combating oxidative stress and inflammation, has not been evaluated in dairy cows. This study aims to evaluate the effects of rumen-protected glutathione (RPGSH) supplementation on lactation, nutrient metabolism, oxidative stress, inflammation, [...] Read more.
Glutathione (GSH), widely present in plant and animal cells and crucial for combating oxidative stress and inflammation, has not been evaluated in dairy cows. This study aims to evaluate the effects of rumen-protected glutathione (RPGSH) supplementation on lactation, nutrient metabolism, oxidative stress, inflammation, and health in transition dairy cows. Forty Holstein dairy cows (2.65 ± 0.78 of parity, 2.81 ± 0.24 of body condition score, 9207.56 ± 1139.18 kg of previous 305-day milk yield, 657.53 ± 55.52 kg of body weight, mean ± SD) were selected from a large cohort of 3215 cows on day 21 before expected calving (day −21 ± 3 d). Cows were randomly stratified into four dietary treatment groups (n = 10 per group): control (basal diet + 0 g/d RPGSH); T1 (basal diet + 1.5 g/d RPGSH); T2 (basal diet + 2 g/d RPGSH); and T3 (basal diet + 3 g/d RPGSH). Supplementation commenced approximately 21 days (±3) prepartum and continued through 21 days postpartum. Blood samples were collected at −21 ± 3, −14 ± 3, −7 ± 3, 0, 7, 14, and 21 d for analysis of serum metabolic parameters related to oxidative stress and inflammation. Milk composition was analyzed from samples collected on days 3, 7, 14, and 21 postpartum. Compared with the control group, supplementation with 2 g/d of RPGSH reduced somatic cell count (p < 0.05) and the incidence of postpartum diseases in dairy cows. No differences were observed among the groups in milk yield, milk fat, protein, lactose, total solids, dry matter intake, or energy-corrected milk. However, fat-corrected milk and feed efficiency were higher in the T2 group compared to the control (p < 0.05). Calcium and phosphorus levels did not differ among the groups. Compared to the control group, cows supplemented with 2 g/d RPGSH had lower β-hydroxybutyrate levels and higher glucose levels on days 14 and 21 postpartum (p < 0.05). From days 14 to 21 postpartum, RPGSH supplementation increased blood GSH, serum catalase, and total antioxidant capacity while reducing malondialdehyde, reactive oxygen species, haptoglobin, cortisol, C-reactive protein, and interleukin−6 levels compared with the control group (p < 0.05). The supplementation of 2 g/d RPGSH showed relatively better effects. RPGSH supplementation at 2 g/d improved lactation performance, nutrient metabolism, oxidative stress, and inflammation status in dairy cows, playing a crucial role in maintaining their health. To our knowledge, this is the first report on the effects of supplementing RPGSH additive in Holstein cows. Full article
(This article belongs to the Section Veterinary Internal Medicine)
Show Figures

Figure 1

Figure 1
<p>Effects of rumen-protected glutathione (RPGSH) on glutathione (GSH, (<b>A</b>)), catalase (CAT, (<b>B</b>)), total antioxidant capacity (T-AOC, (<b>C</b>)), malondialdehyde (MDA, (<b>D</b>)), and reactive oxygen species (ROS, (<b>E</b>)). Treatment: T1 = basal diet + RPGSH 1.5 g/d (shown as ●), T2 = basal diet + RPGSH 2 g/d (shown as ■), T3 = basal diet + RPGSH 3 g/d (shown as ▲), and control = basal diet (shown as ⯁); Lin = linear; Quad = quadratic. Error bars indicate the SEM. Different lowercase letters indicate significant differences between peers (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>Effects of rumen-protected glutathione (RPGSH) on haptoglobin (HP, (<b>A</b>)), cortisol (COR, (<b>B</b>)), C-reactive protein (CRP, (<b>C</b>)), and interleukin-6 (IL-6, (<b>D</b>)). Treatments: T1 = basal diet + RPGSH 1.5 g/d (shown as ●), T2 = basal diet + RPGSH 2 g/d (shown as ■), T3 = basal diet + RPGSH 3 g/d (shown as ▲), and control = basal diet (shown as ⯁); Lin = linear; Quad = quadratic. Error bars indicate the SEM. Different lowercase letters indicate significant differences between peers (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
16 pages, 1125 KiB  
Article
Impact of L-Citrulline Supplementation and HIIT on Lipid Profile, Arterial Stiffness, and Fat Mass in Obese Adolescents with Metabolic-Dysfunction-Associated Fatty Liver Disease: A Randomized Clinical Trial
by Alan Arturo Rodríguez-Carrillo, Mario Ramón Espinoza-Vargas, Katya Vargas-Ortiz, Lorena del Rocío Ibarra-Reynoso, Monserrat Olvera-Juárez, Armando Gómez-Ojeda, Ma. Eugenia Garay-Sevilla and Arturo Figueroa
Nutrients 2025, 17(3), 402; https://doi.org/10.3390/nu17030402 - 23 Jan 2025
Viewed by 410
Abstract
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) and obesity contribute to vascular dysfunction through oxidative stress, heightening cardiovascular risk. Oral supplementation with L-citrulline (L-cit), a precursor of L-arginine (L-arg) and nitric oxide, and high-intensity interval training (HIIT) may improve vascular function and cardiometabolic health. [...] Read more.
Background: Metabolic-dysfunction-associated steatotic liver disease (MASLD) and obesity contribute to vascular dysfunction through oxidative stress, heightening cardiovascular risk. Oral supplementation with L-citrulline (L-cit), a precursor of L-arginine (L-arg) and nitric oxide, and high-intensity interval training (HIIT) may improve vascular function and cardiometabolic health. Objectives: This study aimed to evaluate the combined effects of L-cit supplementation and HIIT on arterial stiffness, body composition, glucose metabolism, lipid profile, and blood pressure (BP) in adolescents with MASLD and obesity. Methods: In this double-blind, placebo-controlled, randomized clinical trial (ClinicalTrials.gov (NCT05778266), 44 adolescents (15–19 years) with MASLD and obesity were assigned to HIIT + L-cit (n = 14), HIIT + placebo (n = 14), or L-cit (n = 15) for 12 weeks. HIIT sessions (85% and 60% peak heart rate during intense and recovery periods) occurred thrice weekly. Training volume progressively increased, and participants performed 20 min of HITT per session in the last 8 weeks. Results: Outcomes included pulse wave velocity (PWV), augmentation index (Aix75), VO2peak, body composition, BP, glucose and lipid profiles, and hepatic steatosis. Compared to L-cit, HIIT + L-cit improved non-high-density lipoprotein cholesterol (p = 0.04), very-low-density lipoprotein cholesterol (p = 0.01), triglycerides (p = 0.02), and VO2peak (p = 0.001). No significant between-group changes were found in PWV, AIx75, hepatic steatosis, and body composition. HIIT + placebo improved VO2peak (p = 0.002), and L-cit decreased the degree of steatosis (p = 0.038). Conclusions: HIIT + L-cit supplementation enhanced lipid profile and cardiorespiratory fitness, while HIIT + placebo improved cardiorespiratory capacity, and L-cit alone decreased hepatic steatosis. Thus, L-cit could be an adjuvant strategy to manage obesity-related MASLD in adolescents. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>CONSORT flow diagram.</p>
Full article ">Figure 2
<p>NS: not significant; VO2peak: peak oxygen consumption; VLDL: very-low-density lipoprotein; HIIT: high-intensity interval training; L-cit: L-citrulline; Pla: placebo. Absolute changes in (<b>A</b>) VO2peak, (<b>B</b>) VLDL-C, (<b>C</b>) non-HDL-C, and (<b>D</b>) triglycerides after 12 weeks of intervention. Analysis performed with one-way ANOVA (blue) or the Kruskal–Wallis test (green) according to normality of data. Post hoc comparisons were conducted using the Bonferroni test: a: <span class="html-italic">p</span> &lt; 0.05 vs. L-cit; b: <span class="html-italic">p</span> &lt; 0.01 vs HIIT + placebo; c: <span class="html-italic">p</span> &lt; 0.01 vs. L-cit; d: <span class="html-italic">p</span> &lt; 0.05 vs. HIIT + placebo.</p>
Full article ">
13 pages, 2822 KiB  
Article
Impact of Calcium Propionate Supplementation on the Lactation Curve and Milk Metabolomic Analysis on Rambouillet Ewes
by Luis Fernando Pérez Segura, Hector A. Lee-Rangel, Rogelio Flores Ramirez, Juan Carlos García-López, Gregorio Álvarez-Fuentes, Anayeli Vázquez Valladolid, Pedro A. Hernández-García, Octavio Negrete Sanchez and Juan Antonio Rendon Huerta
Vet. Sci. 2025, 12(2), 79; https://doi.org/10.3390/vetsci12020079 - 22 Jan 2025
Viewed by 417
Abstract
In lactating ewes, energy demand increases for milk production, reserve mobilizations, and body weight maintenance. For reconversion to energy, ruminants require ruminal propionate production because it is the most predominant substrate for gluconeogenesis and one of the most relevant pathways since it allows [...] Read more.
In lactating ewes, energy demand increases for milk production, reserve mobilizations, and body weight maintenance. For reconversion to energy, ruminants require ruminal propionate production because it is the most predominant substrate for gluconeogenesis and one of the most relevant pathways since it allows an adequate supply of glucose. Calcium propionate supplementation is an alternative to increase glucose production by an external additive. Thus, the objective was to evaluate the effect of calcium propionate (CaPr) on milk production and milk metabolomic profile on lactating ewes. Sixteen Rambouillet (65.3 ± 6.2 kg BW; three years old) were randomly assigned one of two experimental treatments: (a) basal diet without supplementation (CP/0S) and (b) basal diet + 30 g d−1 of CaPr (CP/30S). The experimental period was from parturition day until day 60 (baby lamb weaning). A completely randomized design was used and analyzed with a mixed model. Initial and final lactating weight and milk production differed statistically (p < 0.05) between treatments. CP/30S led to differential changes (p < 0.05) in the lactation curve, showing significant milk production over eight-week measurements. Lactation peak (mL), maximum production (mL), and lactational persistency (d) were superior (p < 0.05) for supplemented ewes. An 11.4% variability was shown in a principal component analysis between treatments. For CP/0S, 63 bioactive compounds were detected, and 55 for CP/30S treatment. The metabolites detected in CP/0S showed that only fatty acid biosynthesis, biosynthesis of unsaturated fatty acids, and fatty acid elongation pathways were affected (p < 0.05) in milk. However, for CP/30S, metabolic pathways related (p < 0.05) were fatty acid biosynthesis, biosynthesis of unsaturated fatty acids, fatty acid elongation, phenylalanine metabolism, and steroid metabolism in milk samples. Calcium propionate supplementation increases milk performance and lactation persistency-induced changes in specific metabolic milk production pathways. Full article
(This article belongs to the Section Nutritional and Metabolic Diseases in Veterinary Medicine)
Show Figures

Figure 1

Figure 1
<p>Lactation curves for daily milk production in ewes supplemented with 30 g/day of calcium propionate (CP/30S) and nonsupplemented ewes (CP/0S) over 60 days from the parturition.</p>
Full article ">Figure 2
<p>Partial least squares discriminant analysis (PLS-DA) loading map for volatile CP/0S and CP/30S.</p>
Full article ">Figure 3
<p>Variable importance in projection test for CP/S0 and CP/30S ewe milk compounds.</p>
Full article ">Figure 4
<p>Spearman’s rank correlation coefficient reports the correlation of 25 most important compounds between treatments.</p>
Full article ">Figure 5
<p>Volcano plot of ewe milk biocompound analysis from CP/S0 and CP/S30 treatments; red dots are upregulated, and purple dots are downregulated metabolites identified.</p>
Full article ">Figure 6
<p>Pathway topology (functional analysis) according to KEEG pathway folder; identification of differential metabolites in ewe milk supplemented with calcium propionate.</p>
Full article ">
19 pages, 862 KiB  
Article
Effect of Dietary Lactobacillus plantarum Supplementation on the Growth Performance, Intestinal Health, Antioxidant Capacity, and mTOR Signaling Pathway of Juvenile Coho Salmon (Oncorhynchus kisutch)
by Qin Zhang, Lan Li, Rongxin Qin, Liuqing Meng, Dongsheng Liu, Tong Tong, Lixiao Xu, Yongqiang Liu and Weiguang Kong
Int. J. Mol. Sci. 2025, 26(3), 907; https://doi.org/10.3390/ijms26030907 - 22 Jan 2025
Viewed by 383
Abstract
This study investigates the effect of dietary Lactobacillus plantarum supplementation on juvenile coho salmon (Oncorhynchus kisutch). Four groups of the juveniles (initial weight 103.87 ± 2.65 g) were fed for 10 weeks with four diets containing 0 (control diet), 105 [...] Read more.
This study investigates the effect of dietary Lactobacillus plantarum supplementation on juvenile coho salmon (Oncorhynchus kisutch). Four groups of the juveniles (initial weight 103.87 ± 2.65 g) were fed for 10 weeks with four diets containing 0 (control diet), 105 (T1), 107 (T2), and 109 (T3) cfu/g of L. plantarum. The main results are as follows: Compared with the control diet, the final weight, specific growth rate (SGR), and weight gain rate (WGR) of the juveniles fed the T1, T2, and T3 diet significantly (p < 0.05) increased, while the feed coefficient ratio (FCR) expressed an opposite trend. The activities of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GSH-PX) in the serum of the juveniles fed the T2 diet significantly (p < 0.05) increased, while the malondialdehyde (MDA) expressed an opposite trend. The expression of phosphatidylinositol 4,5-bisphosphate 3-kinase (pi3k), AKT-interacting protein (akt), mechanistic target of rapamycin kinase (mtor), glucose-6-phosphate dehydrogenase (g6pd), sod, cat, and gsh-px genes in the liver of the juveniles fed the T2 diet significantly (p < 0.05) increased. In conclusion, the T2 diet significantly improved the growth performance, antioxidant capacity, and upregulated key mTOR pathway genes in juvenile coho salmon. Full article
(This article belongs to the Special Issue Fish Nutrition, Metabolism and Physiology)
Show Figures

Figure 1

Figure 1
<p>Effect of dietary <span class="html-italic">L. plantarum</span> supplementation on intestinal tissue morphology of juvenile coho salmon. VH means villi height, VW means villi width, and IWT means intestine wall thickness.</p>
Full article ">Figure 2
<p>Effect of dietary <span class="html-italic">L. plantarum</span> supplementation on the expression of phosphatidylinositol 4,5-bisphosphate 3-kinase (<span class="html-italic">pi3k</span>), AKT-interacting protein (<span class="html-italic">akt</span>), mechanistic target of rapamycin kinase (<span class="html-italic">mtor</span>), glucose-6-phosphate dehydrogenase (<span class="html-italic">g6pd</span>), superoxide dismutase (<span class="html-italic">sod</span>), catalase (<span class="html-italic">cat</span>), and glutathione peroxidase (<span class="html-italic">gsh-px</span>) genes in the liver of juvenile coho salmon. All data are mean ± SE (<span class="html-italic">n</span> = 3), and in the same row, the values with different small letter superscripts are significantly different (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">
14 pages, 2875 KiB  
Article
Glucosyl Hesperidin Supplementation Prevents Tubulointerstitial Fibrosis and Immune Activation in Diabetic Nephropathy in Mice
by Kotaro Hashimoto, Yuki Yoshida, Mion Kamesawa, Nao Yazawa, Hikaru Tominaga, Rahmawati Aisyah, Siyi Chen, Chanikan Bumrungkit, Seiji Kawamoto, Thanutchaporn Kumrungsee and Noriyuki Yanaka
Nutrients 2025, 17(3), 383; https://doi.org/10.3390/nu17030383 - 21 Jan 2025
Viewed by 552
Abstract
Background: Diabetic nephropathy (DN) is a serious condition that can result in end-stage renal failure. Recent evidence has focused on the dietary effects of polyphenols on blood glucose levels and the complications of diabetes. Objectives: In this study, we investigated the protective effect [...] Read more.
Background: Diabetic nephropathy (DN) is a serious condition that can result in end-stage renal failure. Recent evidence has focused on the dietary effects of polyphenols on blood glucose levels and the complications of diabetes. Objectives: In this study, we investigated the protective effect of glucosyl hesperidin (G-Hes), composed of glucose and hesperidin, against streptozotocin (STZ)-induced nephropathy in mice. Methods: We used an STZ-induced diabetic mouse model to investigate the preventive effect of G-Hes on renal pathology. After G-Hes supplementation for 4 weeks, we investigated the renal gene expression profiles using DNA microarray analysis and renal histology to examine the underlying molecular mechanism. Results: G-Hes suppressed the increase in kidney weight without any change in the blood glucose levels. This study identified 511 genes whose expression levels were substantially increased during DN development but were downregulated by G-Hes supplementation. G-Hes prevented mRNA expression associated with renal tubule injury, fibrosis, and immune responses. Notably, G-Hes supplementation considerably decreased the complement component C3 at the mRNA and protein levels in the glomeruli and ameliorated glomerular and mesangial matrix expansion in diabetic nephropathy. Conclusions: G-Hes supplementation is useful in preventing tubulointerstitial fibrosis and inflammation in a mouse model of DN, without exhibiting a hypoglycemic effect. Full article
(This article belongs to the Section Nutrition and Diabetes)
Show Figures

Figure 1

Figure 1
<p>The preventive effect of G-Hes supplementation on an increased kidney weight without any change in the blood glucose level. (<b>A</b>,<b>B</b>) Body weight and blood glucose level showed no difference between the DM group (<span class="html-italic">n</span> = 8) and G-Hes group (<span class="html-italic">n</span> = 7). (<b>C</b>,<b>D</b>) Kidney weight and relative kidney weight of the G-Hes group were lower than the DM group. All values are expressed as the means ± S.E. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> and ## <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> and #### <span class="html-italic">p</span> &lt; 0.0001 as determined by the Student’s <span class="html-italic">t</span>-test (control, <span class="html-italic">n</span> = 5; DM, <span class="html-italic">n</span> = 8; G-Hes, <span class="html-italic">n</span> = 7).</p>
Full article ">Figure 2
<p>Molecular cues for the preventive effect of G-Hes supplementation. (<b>A</b>) Venn diagram shows the renal genes that were upregulated in the DM group and downregulated by G-Hes supplementation. (<b>B</b>) Selected gene expression related to renal pathologies by hyperglycemia.</p>
Full article ">Figure 3
<p>The preventive effect of G-Hes supplementation on renal fibrosis. (<b>A</b>–<b>F</b>) Relative mRNA expression of renal fibrosis-related genes in three groups. (<b>G</b>) Kidney tissues were isolated and paraffin-embedded sections were subjected to Azan-Mallory (AZM) staining. Tubulointerstitial fibrosis in the DM group (the white arrow). Values are the means ± SEM. Statistical analysis was performed with the Student’s <span class="html-italic">t</span>-test. * <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 (control, <span class="html-italic">n</span> = 5; DM, <span class="html-italic">n</span> = 8; G-Hes, <span class="html-italic">n</span> = 7). Scale bars represent 100 μm.</p>
Full article ">Figure 4
<p>The preventive effect of G-Hes supplementation on C3 protein accumulation and the glomerular area. (<b>A</b>) Relative mRNA expression of the renal C3 gene in three groups. (<b>B</b>) Immunohistochemical localization of the C3 protein in the mouse kidney. Kidney tissues of three groups were isolated, and paraffin-embedded sections were subjected to anti-C3 staining. (<b>C</b>,<b>D</b>) Quantification of the average glomerular cross-sectional area. Representative image of the kidney histological staining with H&amp;E reagents. Values are the means ± SEM. Statistical analysis was performed with the Student’s <span class="html-italic">t</span>-test. * <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; 0.005; **** <span class="html-italic">p</span> &lt; 0.001 (control, <span class="html-italic">n</span> = 5; DM, <span class="html-italic">n</span> = 8; G-Hes, <span class="html-italic">n</span> = 7). Scale bars represent 20 μm.</p>
Full article ">Figure 5
<p>Response of renal mRNA expressions to G-Hes supplementation. (<b>A</b>) Total RNA from the DMA and G-Hes groups (<span class="html-italic">n</span> = 15) was subjected to quantitative PCR to examine the renal mRNA expression level of the <span class="html-italic">C3</span> and <span class="html-italic">PDGFRβ</span> genes. The relative mRNA expression level of each gene was analyzed with the Pearson’s correlation coefficient. (<b>B</b>) The relative mRNA expression level of PDGFRβ was determined by quantitative PCR. (<b>C</b>) Kidney tissues of the three groups were isolated, and paraffin-embedded sections were subjected to anti-αSMA staining to quantify the average mesangial cell area in the glomerulus. (<b>D</b>–<b>F</b>) Relative mRNA expression of the renal IFN signaling-related genes in the three groups. All values are expressed as the means ± S.E. * <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; 0.005; **** <span class="html-italic">p</span> &lt; 0.001 (control, <span class="html-italic">n</span> = 5; DM, <span class="html-italic">n</span> = 8; G-Hes, <span class="html-italic">n</span> = 7).</p>
Full article ">Figure 6
<p>Hesperetin inhibits EMT in TGF-β-treated HK-2 cells. Protein expressions of E-cadherin, N-cadherin, snail, and ZEB1 were assessed after 48 h of treatment with TGF-β and hesperetin (Hesp, 30 μM). E-cadherin mRNA expression was examined after 48 h of treatment with TGF-β and hesperetin (Hesp). Error bars indicate the S.E.; ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.001, compared to the control group. <span class="html-italic">n</span> = 6.</p>
Full article ">
18 pages, 972 KiB  
Article
Chia (Salvia hispanica) Seed Oil Modulates the Haemato-Immunological Response, Antioxidative Status and Cytokine Gene Expression of Tropical Freshwater Teleost, Labeo rohita
by Sanjay Kumar Gupta, Rajan Gupta, Akruti Gupta, Md Javed Foysal and Kishore Kumar Krishnani
Biology 2025, 14(1), 95; https://doi.org/10.3390/biology14010095 - 18 Jan 2025
Viewed by 404
Abstract
The present investigation attempts to evaluate the impact of the dietary inclusion of chia (Salvia hispanica) seed oil (CSO) on the indices of haemato-immunology, metabolic enzymes, and expression of immune-responsive cytokine genes in Labeo rohita (rohu) fingerlings. The responses were observed [...] Read more.
The present investigation attempts to evaluate the impact of the dietary inclusion of chia (Salvia hispanica) seed oil (CSO) on the indices of haemato-immunology, metabolic enzymes, and expression of immune-responsive cytokine genes in Labeo rohita (rohu) fingerlings. The responses were observed in a 60-day feeding trial, set up with a total of 180 rohu fingerlings (19.74 ± 0.33 g) randomly allocated to four treatment groups with three replicates each. The groups were fed with a basal diet incorporated with 0%, 1.0%, 2.0%, and 3.0% CSO, denoted as control, CSO (1), CSO (2), and CSO (3), respectively. Significant (p < 0.05) augmentation of hematological indices such as total protein and globulin levels was observed in the group fed a 1.0% CSO-supplemented diet. Serum glucose, cholesterol, triglycerides, and complement reactive protein levels declined, whereas marker anti-oxidative enzymes (SOD, CAT, and GST) and protein metabolic enzymes (ALT and AST) increased (p < 0.05) in the lowest CSO-supplemented group. A significant upregulation of inflammatory cytokine viz. IL-1β, IFN-γ, TNF-α, and TLR22 alongside downregulation of IL-10 was noted in various tissues. The results support the inclusion of 1.0% CSO as a prospective dietary vegan substitute to fish oil in rohu aquaculture. Full article
(This article belongs to the Section Physiology)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Expression of IL-1β gene (<b>a</b>), TNF-α (<b>b</b>), IFN-γ (<b>c</b>), TLR22 (<b>d</b>), and IL-10 (<b>e</b>) in the liver, kidney, and intestine relative to β-actin after 60 days of dietary trial with CSO supplementation in <span class="html-italic">Labeo rohita</span> fingerlings. Statistically significant modulation in the expression of genes relative to the control group and CSO-supplemented dietary groups is displayed. Different superscripts (a, b, c, and d) on the uppermost part of error bars show significant differences (<span class="html-italic">p</span> &lt; 0.05). Values are represented as mean ± SE, n = 3.</p>
Full article ">
20 pages, 4651 KiB  
Article
Reduced Glutathione Promoted Growth Performance by Improving the Jejunal Barrier, Antioxidant Function, and Altering Proteomics of Weaned Piglets
by Zhimei Tian, Yiyan Cui, Miao Yu, Dun Deng, Zhenming Li, Xianyong Ma and Mingren Qu
Antioxidants 2025, 14(1), 107; https://doi.org/10.3390/antiox14010107 - 17 Jan 2025
Viewed by 338
Abstract
Reduced glutathione (GSH) is a main nonenzymatic antioxidant, but its effects and underlying mechanisms on growth and intestinal health in weaned piglets still require further assessment. A total of 180 weaned piglets were randomly allotted to 5 groups: a basal diet (CON), and [...] Read more.
Reduced glutathione (GSH) is a main nonenzymatic antioxidant, but its effects and underlying mechanisms on growth and intestinal health in weaned piglets still require further assessment. A total of 180 weaned piglets were randomly allotted to 5 groups: a basal diet (CON), and a basal diet supplemented with antibiotic chlortetracycline (ABX), 50 (GSH1), 65 (GSH2), or 100 mg/kg GSH (GSH3). Results revealed that dietary GSH1, GSH2, and ABX improved body weight and the average daily gain of weaned piglets, and ABX decreased albumin content but increased aspartate aminotransferase (AST) activity and the ratio of AST to alanine transaminase levels in plasma. GSH2 significantly decreased glucose content but increased the content of triglyceride and cholesterol in the plasma. Both GSH1 and GSH2 improved the jejunal mucosa architecture (villus height, crypt depth, and the ratio of villus height to crypt depth), tight junction protein (ZO-1 and Occludin), and antioxidant capacity (CAT and MDA), and the effects were superior to ABX. Dietary GSH improved the jejunal barrier by probably inhibiting the myosin light chain kinas pathway to up-regulate the transcript expression of tight junction protein (ZO-1 and Occludin) and Mucins. Through the proteomics analysis of the jejunal mucosa using 4D-DIA, the KEGG pathway enrichment analysis showed that differentiated proteins were significantly enriched in redox homeostasis-related pathways such as glutathione metabolism, cytochrome P450, the reactive oxygen species metabolic pathway, the oxidative phosphorylation pathway, and the phosphatidylinositol 3-kinase-serine/threonine kinase pathway in GSH2 vs. CON and in GSH2 vs. ABX. The results of proteomics and qRT-PCR showed that GSH supplementation might dose-dependently promote growth performance and that it alleviated the weaning stress-induced oxidative injury of the jejunal mucosa in piglets by activating SIRTI and Akt pathways to regulate GPX4, HSP70, FoxO1. Therefore, diets supplemented with 50–65 mg/kg GSH can promote the growth of and relieve intestinal oxidative injury in weaned piglets. Full article
Show Figures

Figure 1

Figure 1
<p>Effect of GSH on histomorphology of the jejunal mucosa in weaned piglets. (<b>A</b>) Histology images of jejunum by H&amp;E stains (scale bar, 500 µm). (<b>B</b>–<b>D</b>) The morphometry of the jejunal mucosa. Note: Values are shown as mean ± SEM. * <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. ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH1, a basal diet supplemented with 50 mg/kg GSH; GSH2, a basal diet supplemented with 65 mg/kg GSH; GSH3, a basal diet supplemented with 100 mg/kg GSH in the basal diet.</p>
Full article ">Figure 2
<p>Effect of reduced glutathione on the epithelial barrier of jejunum in weaned piglets. Note: Values are shown as mean ± SEM. * <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. ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH1, a basal diet supplemented with 50 mg/kg GSH; GSH2, a basal diet supplemented with 65 mg/kg GSH; GSH3, a basal diet supplemented with 100 mg/kg GSH in the basal diet. ZO-1, zonula occludens-1; Muc1/2, mucin 1/2; MLCK, myosin light chain kinas.</p>
Full article ">Figure 3
<p>Effects of reduced glutathione on jejunal antioxidant status in weaned piglets. Note: Values are shown as mean ± SEM. * <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. ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH1, a basal diet supplemented with 50 mg/kg GSH; GSH2, a basal diet supplemented with 65 mg/kg GSH; GSH3, a basal diet supplemented with 100 mg/kg GSH in the basal diet. MDA, malondialdehyde; GSH, reduced glutathione, GPX, glutathione peroxidase; CAT, catalase; T-AOC, total antioxidant capacity; T-SOD, total superoxide dismutase.</p>
Full article ">Figure 4
<p>Proteomics analysis of the jejunal mucosa in weaned piglets. (<b>A</b>) Qualitative and quantitative analysis of identified proteins; (<b>B</b>) Differentially expressed proteins; (<b>C</b>) Two-dimensional principal component analysis; (<b>D</b>–<b>F</b>) KEGG pathway enrichment from GSH2 vs. CON, GSH2 vs. ABX, and ABX vs. CON, respectively. KEGG: Kyoto encyclopedia of genes and genomes. Note: ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH2, a basal diet supplemented with 65 mg/kg GSH in the basal diet.</p>
Full article ">Figure 4 Cont.
<p>Proteomics analysis of the jejunal mucosa in weaned piglets. (<b>A</b>) Qualitative and quantitative analysis of identified proteins; (<b>B</b>) Differentially expressed proteins; (<b>C</b>) Two-dimensional principal component analysis; (<b>D</b>–<b>F</b>) KEGG pathway enrichment from GSH2 vs. CON, GSH2 vs. ABX, and ABX vs. CON, respectively. KEGG: Kyoto encyclopedia of genes and genomes. Note: ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH2, a basal diet supplemented with 65 mg/kg GSH in the basal diet.</p>
Full article ">Figure 4 Cont.
<p>Proteomics analysis of the jejunal mucosa in weaned piglets. (<b>A</b>) Qualitative and quantitative analysis of identified proteins; (<b>B</b>) Differentially expressed proteins; (<b>C</b>) Two-dimensional principal component analysis; (<b>D</b>–<b>F</b>) KEGG pathway enrichment from GSH2 vs. CON, GSH2 vs. ABX, and ABX vs. CON, respectively. KEGG: Kyoto encyclopedia of genes and genomes. Note: ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH2, a basal diet supplemented with 65 mg/kg GSH in the basal diet.</p>
Full article ">Figure 5
<p>Effect of reduced glutathione on oxidative stress-related genes in the jejunal mucosa of weaned piglets. Note: Values are shown as mean ± SEM. * <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. ABX, a basal diet supplemented with chlortetracycline; CON, basal diet; GSH1, a basal diet supplemented with 50 mg/kg GSH; GSH2, a basal diet supplemented with 65 mg/kg GSH; GSH3, a basal diet supplemented with 100 mg/kg GSH in the basal diet. GPX4, glutathione peroxidase 4; Hsp70, heat shock protein 70 kDa; Hsp90, heat shock protein 90 kDa; SIRT1, sirtuin 1; FoxO1, forkhead box protein O1; Akt1, serine/threonine kinase 1.</p>
Full article ">
20 pages, 1087 KiB  
Article
Coffee Pulp from Azores: A Novel Phytochemical-Rich Food with Potential Anti-Diabetic Properties
by Anabela S. G. Costa, Juliana A. Barreto Peixoto, Susana Machado, Liliana Espírito Santo, Thiago F. Soares, Nelson Andrade, Rui Azevedo, Agostinho Almeida, Helena S. Costa, Maria Beatriz Prior Pinto Oliveira, Fátima Martel, Jesus Simal-Gandara and Rita C. Alves
Foods 2025, 14(2), 306; https://doi.org/10.3390/foods14020306 - 17 Jan 2025
Viewed by 594
Abstract
Coffee pulp, a by-product of wet coffee processing, shows significant potential in the food and health domains, but its real applications remain underexplored. This work investigated the chemical composition and bioactive properties of coffee pulp from São Miguel Island (Azores, Portugal). The studied [...] Read more.
Coffee pulp, a by-product of wet coffee processing, shows significant potential in the food and health domains, but its real applications remain underexplored. This work investigated the chemical composition and bioactive properties of coffee pulp from São Miguel Island (Azores, Portugal). The studied coffee pulp exhibited high fiber content (52% dw), mostly insoluble; notable mineral levels (10.6%), mainly K, Ca, and Mg; and 6% dw of total amino acids, with hydroxyproline, aspartic acid, glutamic acid, and leucine in higher amounts. Despite containing low fat (1.6% dw), mainly saturated, it also showed considerable amounts of polyunsaturated fatty acids with a favorable n6/n3 ratio (1.40) and vitamin E (α-, β-, and γ-tocopherols). Its antioxidant capacity can be partially explained by the chlorogenic acid content (9.2 mg/g dw), and caffeine (0.98%) was present in similar amounts to those observed in some arabica coffee beans. A decrease in glucose uptake in Caco-2 cells was found, but not in fructose, suggesting selective inhibition of SGLT1 and potential antidiabetic effects. These results show that Azorean coffee pulp has potential as a sustainable and bioactive ingredient for incorporation into functional foods or dietary supplements. Full article
Show Figures

Figure 1

Figure 1
<p>Total and free amino acid profiles of Azorean coffee pulp. The results are expressed as mean ± standard deviation (<span class="html-italic">n</span> = 3), in dry weight. n.d., not detected (For total amino acids, conversion of asparagine and glutamine into aspartic acid and glutamic acid occurs during acid hydrolysis, respectively).</p>
Full article ">Figure 2
<p>Effect of coffee pulp extracts on (<b>a</b>) <sup>3</sup>H-DG and (<b>b</b>) <sup>14</sup>C-FRU uptake by Caco-2 cells in comparison with control (100%, horizontal dash line). The results are expressed as average ± SEM (<span class="html-italic">n</span> = 9); ****, <span class="html-italic">p</span> &lt; 0.0001 significantly different from control by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">Figure 3
<p>Effect of coffee pulp extracts on (<b>a</b>) Caco-2 cell viability (extracellular LDH activity), and (<b>b</b>) culture mass (SRB assay), in comparison with control (100%, horizontal dash line). The results are expressed as average ± SEM (<span class="html-italic">n</span> = 9). *, <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 significantly different from control by Student’s <span class="html-italic">t</span>-test.</p>
Full article ">
20 pages, 1936 KiB  
Article
Utilizing Invasive Pterygoplichthys pardalis as a Sustainable Fish Meal Substitute and Euphorbia hirta Extract Supplement: Effects on Growth Performance, Organosomatic Indices, Hematological Profiles, and Serum Biochemistry in Chinese Bullfrogs (Hoplobatrachus chinensis)
by Sontaya Sookying, Phanit Srisuttha, Vipada Rodprasert, Chanthima Chaodon, Wikit Phinrub, Nantaporn Sutthi and Paiboon Panase
Life 2025, 15(1), 115; https://doi.org/10.3390/life15010115 - 16 Jan 2025
Viewed by 655
Abstract
This research examined the efficacy of substituting commercial fish meal (CFM) with Pterygoplichthys pardalis meal (PPM) in Hoplobatrachus chinensis diets, with and without Euphorbia hirta extract (EHE) supplementation. The study utilized six dietary treatments: a control diet (0% PPM, no EHE) and five [...] Read more.
This research examined the efficacy of substituting commercial fish meal (CFM) with Pterygoplichthys pardalis meal (PPM) in Hoplobatrachus chinensis diets, with and without Euphorbia hirta extract (EHE) supplementation. The study utilized six dietary treatments: a control diet (0% PPM, no EHE) and five experimental diets with varying PPM levels (0%+, 25%+, 50%+, 75%+, and 100%+), each fortified with 300 mg/kg EHE. The experiment spanned 90 days. The analysis revealed that PPM exhibited superior amino acid profiles compared to CFM, both in quality and quantity, while CFM demonstrated higher fatty acid content. The growth metrics showed a significant decline only in the group receiving 100% PPM replacement with EHE supplementation. Most organosomatic indices remained consistent across the treatments, with the exception of intraperitoneal fat, which decreased in all EHE-supplemented groups. Blood parameters, including white blood cells, red blood cells, and hematocrit, along with serum proteins (total protein, globulin, and albumin), displayed an upward trend in all EHE-supplemented groups. The 50%+ and 75%+ PPM replacement groups exhibited significantly elevated serum glucose levels (p < 0.05). Liver enzymes (alanine transaminase and aspartate transaminase) showed no significant variations among the treatments. The results indicate that PPM can serve as an effective replacement for up to 75% of CFM in H. chinensis feed, without compromising their growth performance. Moreover, supplementing with EHE helps to enhance essential biochemical indices in the body, without adversely affecting liver function. This investigation offers valuable perspectives on the development of sustainable aquaculture feed and the potential application of invasive fish species in aquatic animal nutrition. Full article
(This article belongs to the Special Issue Fish Nutrition, Production, and Welfare)
Show Figures

Figure 1

Figure 1
<p>Growth performance of <span class="html-italic">Hoplobatrachus chinensis</span> fed with fish meal replacement at six different levels of <span class="html-italic">Pterygoplichthys pardalis</span> meal and supplemented with <span class="html-italic">Euphorbia hirta</span> leaf extract for 90 days; WG = weight gain (<b>a</b>), ADG = average daily growth (<b>b</b>), SGR = specific growth rate (<b>c</b>), SR = survival rate (<b>d</b>), FCR = feed conversion ratio (<b>e</b>), and PER = protein efficiency ratio (<b>f</b>). Different letters indicate significant differences (<span class="html-italic">p &lt;</span> 0.05).</p>
Full article ">Figure 2
<p>Organosomatic indices of <span class="html-italic">Hoplobatrachus chinensis</span> fed with fish meal replacement at six different levels of <span class="html-italic">Pterygoplichthys pardalis</span> meal and supplemented with <span class="html-italic">Euphorbia hirta</span> leaf extract for 90 days; HSI = hepatosomatic index (<b>a</b>), RSI = renosomatic index (<b>b</b>), ISI = intestinosomatic index (<b>c</b>), IF = intraperitoneal fat, (<b>d</b>) and SSI = spleenosomatic index (<b>e</b>). Different letters indicate significant differences (<span class="html-italic">p &lt;</span> 0.05).</p>
Full article ">Figure 3
<p>Hematological indices of <span class="html-italic">Hoplobatrachus chinensis</span> fed with fish meal replacement at six different levels of <span class="html-italic">Pterygoplichthys pardalis</span> meal and supplemented with <span class="html-italic">Euphorbia hirta</span> leaf extract for 90 days; WBC = white blood cell count (<b>a</b>), RBC = red blood cell count, (<b>b</b>) and Hct = hematocrit (<b>c</b>). Different letters indicate significant differences (<span class="html-italic">p &lt;</span> 0.05).</p>
Full article ">Figure 4
<p>Serum biochemical indices of <span class="html-italic">Hoplobatrachus chinensis</span> fed with fish meal replacement at six different levels of <span class="html-italic">Pterygoplichthys pardalis</span> meal and supplemented with <span class="html-italic">Euphorbia hirta</span> leaf extract for 90 days: total protein (<b>a</b>), albumin (<b>b</b>), globulin (<b>c</b>), glucose (<b>d</b>), ALT—alanine transaminase (<b>e</b>), and AST—aspartate transaminase (<b>f</b>). Different letters indicate significant differences (<span class="html-italic">p &lt;</span> 0.05).</p>
Full article ">
12 pages, 264 KiB  
Review
The Clinical Use of Myo-Inositol in IVF-ET: A Position Statement from the Experts Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS), the Polish Society of Andrology, and the International Scientific Association for the Support and Development of Medical Technologies
by Artur Wdowiak, Szymon Bakalczuk, Michał Filip, Antonio Simone Laganà and Vittorio Unfer
J. Clin. Med. 2025, 14(2), 558; https://doi.org/10.3390/jcm14020558 - 16 Jan 2025
Viewed by 467
Abstract
Background: Myo-inositol plays a vital role in human health, functioning as a second messenger of FSH and facilitating the transport of glucose into the cell. Consequently, myo-inositol is regularly utilized in the treatment of polycystic ovary syndrome (PCOS), wherein it acts upon metabolic [...] Read more.
Background: Myo-inositol plays a vital role in human health, functioning as a second messenger of FSH and facilitating the transport of glucose into the cell. Consequently, myo-inositol is regularly utilized in the treatment of polycystic ovary syndrome (PCOS), wherein it acts upon metabolic factors, improving insulin sensitivity and reducing total androgen levels. Patients with PCOS frequently suffer from infertility; thus, the use of myo-inositol has been explored in improving assistive reproductive technique (ART) procedures. This is by no means limited to patients with PCOS, as inositol has found applications in non-PCOS patient groups in addition to in male factor infertility. This joint statement from the Experts Group on Inositol in Basic and Clinical Research and on PCOS (EGOI-PCOS), the Polish Society of Andrology, and the International Scientific Association for the Support and Development of Medical Technologies discusses the latest evidence on this topic, with the aim of interrogating whether myo-inositol could be implemented in everyday ART patient care. Methods: The authors conducted a narrative review performed via an independent literature search between July and August 2024, using the search platforms PubMed, Web of Science, and Google Scholar. Results: In both non-PCOS and PCOS populations seeking IVF care, MI supplementation prior to ovarian stimulation may positively affect gonadotropin use and duration, oocyte and embryo quality, fertilization, and clinical pregnancy rates. Conclusions: This position statement recommends that myo-inositol be considered as a potential pretreatment strategy prior to ovarian hyperstimulation with gonadotropins. Full article
(This article belongs to the Section Reproductive Medicine & Andrology)
19 pages, 5945 KiB  
Article
The Inhibitory Effects of NCT503 and Exogenous Serine on High-Selenium Induced Insulin Resistance in Mice
by Shuo Zhan, Jianrong Wang, Mingyu Zhu, Yiqun Liu, Feng Han, Licui Sun, Qin Wang and Zhenwu Huang
Nutrients 2025, 17(2), 311; https://doi.org/10.3390/nu17020311 - 16 Jan 2025
Viewed by 361
Abstract
Objective: This study aims to identify whether the development of insulin resistance (IR) induced by high selenium (Se) is related to serine deficiency via the inhibition of the de novo serine synthesis pathway (SSP) by the administrations of 3-phosphoglycerate dehydrogenase (PHGDH) inhibitor (NCT503) [...] Read more.
Objective: This study aims to identify whether the development of insulin resistance (IR) induced by high selenium (Se) is related to serine deficiency via the inhibition of the de novo serine synthesis pathway (SSP) by the administrations of 3-phosphoglycerate dehydrogenase (PHGDH) inhibitor (NCT503) or exogenous serine in mice. Method: forty-eight male C57BL/6J mice were randomly divided into four groups: adequate-Se (0.1 mgSe/kg), high-Se (0.8 mgSe/kg), high-Se +serine (240 mg/kg/day), and high-Se +NCT503 (30 mg/kg, twice a week) for 5 months. The glucose tolerance test (GTT) and insulin tolerance test (ITT) were used to confirm the development of IR in mice with high-Se intake, and fasting blood glucose levels were measured monthly. The Se contents in plasma and tissues were detected by ICP-MS. The levels of insulin (INS), homocysteine (HCY), and serine in plasma were tested by ELISA. Western blot analyses were conducted to evaluate the protein expressions of glutathione peroxidase 1 (GPX1), selenoprotein P (SELENOP) and PHGDH, the PI3K-AKT-mTOR pathway, folate cycle (SHMT1, MTHFR), and methionine cycle (MS). Results: An IR model was developed in mice from the high-Se group with elevated fasting blood glucose and INS levels, impaired glucose tolerance, and reduced insulin sensitivity, but not in both the high-Se +serine group and the high-Se +NCT503 group. Compared with the high-Se and high-Se +serine groups, the expressions of GPX1 and SELENOP significantly decreased for the high-Se +NCT503 group in the liver, muscle, and pancreas tissues. The expression of PHGDH of high-Se group was significantly higher than that of the adequate-Se group in the liver (p < 0.05) and pancreas (p < 0.001). Also, the expected high expression of PHGDH was effectively inhibited in mice from the high-Se +serine group but not from the high-Se +NCT503 group. The expression of p-AKT (Ser-473) for the high-Se group was significantly lower than that of the adequate-Se group in the liver, muscle, and pancreas. Conclusions: The IR induced by high-Se intake in the body has been confirmed to be partially due to serine deficiency, which led to the initiation of SSP to produce endogenous serine. The supplementations of exogenous serine or inhibitors of PHGDH in this metabolic pathway could be used for the intervention. Full article
Show Figures

Figure 1

Figure 1
<p>Animal experiment flowchart.</p>
Full article ">Figure 2
<p>Effects of selenium levels (0.1, 0.8 mg Se/kg) and interventions (Ser, NCT503) on weight, glucose metabolism, GTT, and ITT in C57BL/6J mice. (<b>a</b>) Changes in body weight over 5 months. (<b>b</b>) Fasting blood glucose levels measured monthly. (<b>c</b>) Glucose tolerance assessed at the end of the fourth month. (<b>d</b>) Insulin tolerance evaluated at the end of the fourth month. (Data are expressed as mean ± SD, n = 10–12). (* <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; two-way ANOVA). Both Ser and NCT503 effectively mitigate Se-induced metabolic disturbances, with Ser showing a more substantial improvement in body weight and blood glucose regulation.</p>
Full article ">Figure 3
<p>Effects of selenium levels (0.1 mg and 0.8 mg Se/kg) and interventions (Ser, NCT503) on plasma biochemical markers in C57BL/6J mice. (<b>a</b>–<b>c</b>) Changes in plasma biochemical markers: (<b>a</b>) insulin (INS, mIU/L), (<b>b</b>) homocysteine (Hcy, µmol/L), amd (<b>c</b>) serine (Ser, µmol/L). (<b>d</b>) Selenium levels in plasma, liver, muscle, and pancreas. (<b>e</b>) PHGDH enzyme activity in plasma, liver, muscle, and pancreas. (Data are expressed as mean ± SD, n = 7). (*** <span class="html-italic">p</span> &lt; 0.001, **** <span class="html-italic">p</span> &lt; 0.0001; two-way ANOVA). Supplementation with Ser and NCT503 modulates biochemical markers and PHGDH enzyme activity. Ser supplementation lowers INS and HCY levels, while NCT503 decreases INS and serine levels, and increases HCY levels. The NCT503 inhibitor has a stronger inhibitory effect on PHGDH enzyme activity.</p>
Full article ">Figure 4
<p>Effect of 0.1, 0.8 mg Se/kg and Ser or NCT503 intervention group on the Expressions of selenoproteins and enzymes in Liver Tissue. (<b>a</b>) WB images. (<b>b</b>–<b>g</b>) grayscale analysis. (Mean ± SD, n = 3, * <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; two-way ANOVA). NCT503 and Ser differentially modulate liver enzyme expression, with NCT503 enhancing PHGDH activity and reducing SELENOP, while Ser primarily decreases MS expression.</p>
Full article ">Figure 5
<p>Effect of 0.1, 0.8 mg Se/kg and Ser or the NCT503 intervention group on the expressions of selenoproteins and enzymes in muscle Tissue. (<b>a</b>) WB images. (<b>b</b>–<b>g</b>) grayscale analysis. (Mean ± SD, n = 3, * <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; two-way ANOVA). NCT503 reduces GPX1 and SELENOP expression in muscle, while Ser mainly reduces MS expression.</p>
Full article ">Figure 6
<p>Effect of 0.1, 0.8 mg Se/kg and Ser or the NCT503 intervention group on the expression of selenoproteins and enzymes in Mouse pancreas Tissue. (<b>a</b>) WB images. (<b>b</b>–<b>f</b>) grayscale analysis. (Mean ± SD, n = 3, * <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; two-way ANOVA). NCT503 increases PHGDH expression in the pancreas, while Ser primarily reduces SHMT1 expression.</p>
Full article ">Figure 7
<p>Effects of 0.1, 0.8 mg Se/kg, and Ser or the NCT503 intervention group on the PI3K-AKT-mTOR pathway in mice liver. (<b>a</b>) WB images. (<b>b</b>–<b>f</b>) Pathway grayscale analysis (Mean ± SD, n = 3, * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001; two-way ANOVA). Ser and NCT503 supplementation modulate mTOR expression in the liver, with NCT503 and Ser both enhancing mTOR signaling.</p>
Full article ">Figure 8
<p>Effects of 0.1, 0.8 mg Se/kg, and Ser or the NCT503 intervention group on the PI3K-AKT-mTOR pathway in mice muscle. (<b>a</b>) WB images. (<b>b</b>–<b>f</b>) Pathway grayscale analysis (Mean ± SD, n = 3, * <span class="html-italic">p</span> &lt; 0.05; two-way ANOVA). While Ser and NCT503 supplementation slightly increased Akt/mTOR pathway markers in muscle, these changes were not significant.</p>
Full article ">Figure 9
<p>Effects of 0.1, 0.8 mg Se/kg, and Ser or the NCT503 intervention group on the PI3K-AKT-mTOR pathway in mice muscle pancreas. (<b>a</b>) WB images. (<b>b</b>–<b>f</b>) Pathway grayscale analysis (Mean ± SD, n = 3, * <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; two-way ANOVA). Both Ser and NCT503 supplementation restored Akt/mTOR signaling in the pancreas, with significant increases in mTOR and phosphorylated Akt levels compared to the high-Se group.</p>
Full article ">Figure 10
<p>The SSP supports selenium detoxification and metabolic balance. The SSP plays a pivotal role in maintaining redox balance, supporting methylation reactions, and facilitating selenium detoxification under high selenium conditions. Increased PHGDH expression drives serine production, replenishing SAM for methylation and promoting GSH synthesis to combat oxidative stress. Excessive SSP activation burdens the methionine and folate cycles, depleting SAM and disrupting methylation homeostasis. Serine supplementation alleviates these effects by restoring one-carbon metabolism and enhancing selenium detoxification. Similarly, pharmacological inhibition of PHGDH (e.g., NCT-503) reduces SSP overactivation, highlighting its potential as a therapeutic target in Se-induced metabolic dysregulation.</p>
Full article ">Figure 11
<p>Integration of mTOR signaling, oxidative stress, and IR. The figure delineates the intricate mechanisms by which mTOR signaling integrates oxidative stress, energy homeostasis, and insulin sensitivity. Oxidative stress activates AMPK, which inhibits mTORC1 via TSC2 phosphorylation and suppresses Rheb, a key mTOR activator. Reduced mTOR signaling impairs AKT activation, disrupting GLUT4 translocation and diminishing glucose uptake. Concurrently, impaired AKT fails to suppress FOXO1 nuclear activity, leading to enhanced gluconeogenesis through the upregulation of key enzymes like PEPCK. Elevated oxidative stress and inflammation further aggravate IR by activating pathways, inducing aberrant IRS1 phosphorylation, and reducing insulin receptor sensitivity. These combined effects underscore the centrality of mTOR in maintaining redox balance, metabolic stability, and insulin signaling integrity.</p>
Full article ">
19 pages, 1921 KiB  
Article
The Effect of the 14:10-Hour Time-Restricted Feeding (TRF) Regimen on Selected Markers of Glucose Homeostasis in Diet-Induced Prediabetic Male Sprague Dawley Rats
by Sthembiso Msane, Andile Khathi and Aubrey Mbulelo Sosibo
Nutrients 2025, 17(2), 292; https://doi.org/10.3390/nu17020292 - 15 Jan 2025
Viewed by 493
Abstract
Background: Prediabetes is a condition that often precedes the onset of type 2 diabetes mellitus (T2DM). Literature evidence indicates that prediabetes is reversible, making it an important therapeutic target for preventing the progression to T2DM. Several studies have investigated intermittent fasting as a [...] Read more.
Background: Prediabetes is a condition that often precedes the onset of type 2 diabetes mellitus (T2DM). Literature evidence indicates that prediabetes is reversible, making it an important therapeutic target for preventing the progression to T2DM. Several studies have investigated intermittent fasting as a possible method to manage or treat prediabetes. Objectives: This study evaluated the impact of a 14:10-hour time-restricted feeding (TRF) regimen on leptin concentration, insulin sensitivity and selected markers associated with the insulin signalling pathway and glucose homeostasis in diet-induced prediabetic rats. Methods: Twenty-four male Sprague Dawley rats were obtained and randomly divided into two dietary groups: group 1 (n = 6) received a standard diet and water, while group 2 (n = 18) was provided a high-fat, high-carbohydrate (HFHC) diet supplemented with 15% fructose for a period of 20 weeks to induce prediabetes. After confirming prediabetes, an intermittent fasting (IF) regimen was assigned to the rats while also having untreated and metformin-treated prediabetic rats serving as controls. Results: Both IF and HFHC-Met groups yield significantly lower blood glucose, leptin and BMI results compared to the prediabetic group. The IF group yielded significantly lower insulin, HOMA-IR and HbA1C than both controls. Conclusions: The study showed the potential of IF in alleviating prediabetes-induced dysregulation of glucose homeostasis and therefore warrants further investigations into its use in the management of prediabetes. Full article
(This article belongs to the Special Issue Nutritional and Dietary Approaches to Diabetes)
Show Figures

Figure 1

Figure 1
<p>Experimental protocol showing prediabetes induction, intermittent fasting and HFHC-Met implementation on male Sprague Dawley rats. ADA criteria were used to identify prediabetes.</p>
Full article ">Figure 2
<p>Effect of the 14:10-hour TRF regimen on the 2-h oral glucose tolerance test in prediabetic rats during the 12-week experiment. ∆ = <span class="html-italic">p</span> value &lt; 0.05 denotes comparison with NPD and β = <span class="html-italic">p</span> value &lt; 0.05 denotes a comparison with PD.</p>
Full article ">Figure 2 Cont.
<p>Effect of the 14:10-hour TRF regimen on the 2-h oral glucose tolerance test in prediabetic rats during the 12-week experiment. ∆ = <span class="html-italic">p</span> value &lt; 0.05 denotes comparison with NPD and β = <span class="html-italic">p</span> value &lt; 0.05 denotes a comparison with PD.</p>
Full article ">Figure 3
<p>Effect of the 14:10-hour TRF regimen on the plasma leptin concentration in prediabetic rats at week 12 of the experiment. Data are presented as mean values. ∆ = <span class="html-italic">p</span> value &lt; 0.05 denotes comparison with NPD and β = <span class="html-italic">p</span> value &lt; 0.05 denotes a comparison with PD.</p>
Full article ">Figure 4
<p>Effect of the 14:10-hour TRF regimen on the IRS1, IRS2, Akt, PI3K, mTORC1 and GLUT 4 in prediabetic rats at week 12 of the experiment. ∆ = <span class="html-italic">p</span> value &lt; 0.05 denotes comparison with NPD and β = <span class="html-italic">p</span> value &lt; 0.05 denotes a comparison with PD.</p>
Full article ">Figure 5
<p>Effect of the 14:10-hour TRF regimen on the glycogen concentration on liver and skeletal tissues in prediabetic rats at week 12 of the experiment. ∆= <span class="html-italic">p</span> value &lt; 0.05 denotes comparison with NPD and β = <span class="html-italic">p</span> value &lt; 0.05 denotes a comparison with PD.</p>
Full article ">
Back to TopTop