Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Am Nutr Assoc ; 43(5): 437-451, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38305833

ABSTRACT

OBJECTIVE: Previous studies have shown that oyster peptides (OPs) have antioxidant and anti-fatigue activities. This study aimed to investigate the effects of OPs on swimming endurance in mice and the underlying mechanisms. METHODS: The mice were subjected to gavage with OPs and subjected to exercise training. After 14 days, various biochemical indicators in the blood and gastrocnemius muscle of mice were assessed, and real-time PCR was utilized to detect the level of signal pathway regulation by OPs in the gastrocnemius muscle. Molecular docking technology was employed to observe the potential active components in OPs that regulate signal pathways. RESULTS: In this study, OPs supplementation combined with and without exercise significantly extended swimming time compared to the sedentary group. OPs supplementation with exercise also increased glycogen levels and decreased blood urea nitrogen, lactate dehydrogenase, and lactic acid levels. Additionally, mice in the exercise with OPs group exhibited higher activities of antioxidant enzymes. OPs can upregulate metabolic regulatory factors such as AMP-activated protein kinase, peroxisome proliferator-activated receptor gamma coactivator-1 alpha, peroxisome proliferator-activated receptor delta, and glucose transporter 4, thereby increasing energy supply during exercise. Additionally, OPs enhances the expression of heme oxygenase 1 and superoxide dismutase 2, thereby reducing oxidative stress during physical activity. Molecular docking analyses revealed that peptides found in OPs formed hydrogen bonds with AMPK and HO-1, indicating that they can exert bioactivity by activating target proteins such as AMPK and HO-1. CONCLUSIONS: OPs supplementation improved energy reserves, modulated energy metabolism pathways, and coordinated antioxidative stress responses, ultimately enhancing swimming endurance. These findings suggest that OPs have the potential to improve exercise levels by promoting metabolism and improving energy utilization efficiency.


Subject(s)
AMP-Activated Protein Kinases , Heme Oxygenase-1 , Muscle, Skeletal , Physical Conditioning, Animal , Physical Endurance , Swimming , Animals , AMP-Activated Protein Kinases/metabolism , Mice , Heme Oxygenase-1/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Physical Endurance/drug effects , Male , Peptides/pharmacology , Molecular Docking Simulation , Glycogen/metabolism , Ostreidae , Antioxidants/pharmacology , Antioxidants/metabolism , Superoxide Dismutase/metabolism , Dietary Supplements , Membrane Proteins
2.
J Transl Med ; 21(1): 14, 2023 01 10.
Article in English | MEDLINE | ID: mdl-36627673

ABSTRACT

BACKGROUND: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal disease with high morbidity and mortality, affecting preterm infants especially those with very low and extremely low birth weight. ß-glucan has manifested multiple biological effects including anti-inflammatory, regulation of gut microbiota, and immunomodulatory activities. This study aimed to investigate the effects of ß-glucan on NEC. METHODS: Neonatal C57BL/6 mice were randomly divided into three groups: Control group, NEC group and ß-glucan group. Newborn 3-day-old mice were gavaged with either 1 mg/ml ß-glucan or phosphate buffer saline at 0.03 ml/g for 7 consecutive days before NEC induction and a NEC model was established with hypoxia combined with cold exposure and formula feeding. All the pups were killed after 72-h modeling. Hematoxylin-eosin staining was performed to assess the pathological injury to the intestines. The mRNA expression levels of inflammatory factors in intestinal tissues were determined using quantitative real-time PCR. The protein levels of TLR4, NF-κB and tight junction proteins in intestinal tissues were evaluated using western blotting and immunohistochemistry. 16S rRNA sequencing was performed to determine the structure of the gut microbiota. RESULTS: ß-glucan administration ameliorated intestinal injury of NEC mice; reduced the intestinal expression of TLR4, NF-κB, IL-1ß, IL-6, and TNF-α; increased the intestinal expression of IL-10; and improved the expression of ZO-1, Occludin and Claudin-1 within the intestinal barrier. Pre-treatment with ß-glucan also increased the proportion of Actinobacteria, Clostridium butyricum, Lactobacillus johnsonii, Lactobacillus murinus, and Lachnospiraceae bacterium mt14 and reduced the proportion of Klebsiella oxytoca g Klebsiella in the NEC model. CONCLUSION: ß-glucan intervention prevents against NEC in neonatal mice, possibly by suppressing the TLR4-NF-κB signaling pathway, improving intestinal barrier function, and partially regulating intestinal microbiota.


Subject(s)
Enterocolitis, Necrotizing , Gastrointestinal Microbiome , Infant, Newborn , Humans , Animals , Mice , Animals, Newborn , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Enterocolitis, Necrotizing/prevention & control , Enterocolitis, Necrotizing/metabolism , Enterocolitis, Necrotizing/pathology , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Infant, Premature , Mice, Inbred C57BL , Inflammation/pathology , Disease Models, Animal , Intestinal Mucosa/pathology
3.
Article in English | MEDLINE | ID: mdl-36673982

ABSTRACT

Breast cancer, with an overall poor clinical prognosis, is one of the most heterogeneous cancers. DNA damage repair (DDR) and epithelial-mesenchymal transition (EMT) have been identified to be associated with cancer's progression. Our study aimed to explore whether genes with both functions play a more crucial role in the prognosis, immune, and therapy response of breast cancer patients. Based on the Cancer Genome Atlas (TCGA) cancer database, we used LASSO regression analysis to identify the six prognostic-related genes with both DDR and EMT functions, including TP63, YWHAZ, BRCA1, CCND2, YWHAG, and HIPK2. Based on the six genes, we defined the risk scores of the patients and reasonably analyzed the overall survival rate between the patients with the different risk scores. We found that overall survival in higher-risk-score patients was lower than in lower-risk-score patients. Subsequently, further GO and KEGG analyses for patients revealed that the levels of immune infiltration varied for patients with high and low risk scores, and the high-risk-score patients had lower immune infiltration's levels and were insensitive to treatment with chemotherapeutic agents. Furthermore, the Gene Expression Omnibus (GEO) database validated our findings. Our data suggest that TP63, YWHAZ, BRCA1, CCND2, YWHAG, and HIPK2 can be potential genetic markers of prognostic assessment, immune infiltration and chemotherapeutic drug sensitivity in breast cancer patients.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Epithelial-Mesenchymal Transition , Breast , DNA Repair , Databases, Factual , Carrier Proteins , Protein Serine-Threonine Kinases , 14-3-3 Proteins
4.
Front Cell Infect Microbiol ; 12: 1064462, 2022.
Article in English | MEDLINE | ID: mdl-36519131

ABSTRACT

Background: Necrotizing enterocolitis (NEC) is the most prevalent gastrointestinal disorder that predominantly threatens preterm newborns. Succinate is an emerging metabolic signaling molecule that was recently studied in relation to the regulation of intestinal immunity and homeostasis. We aimed to investigate the relationship between NEC and gut luminal succinate and preliminarily explored the effect of succinate on NEC pathogenesis. Methods: Fecal samples from human neonates and mouse pups were analyzed by HPLC - MS/MS and 16S rRNA gene sequencing. C57BL/6 mice were randomly divided into four groups: control, NEC, Lsuc, and Hsuc. The mortality, weight gain, and intestinal pathological changes in four mouse groups were observed. Inflammatory cytokines and markers of macrophages were identified by quantitative real-time PCR. Succinate receptor 1 (SUCNR1) localization was visualized by immunohistochemistry. The protein levels of SUCNR1 and hypoxia-inducible factor 1a (HIF-1a) were quantified by western blotting. Results: The levels of succinate in feces from NEC patients were higher than those in feces from non-NEC patients (P <0.05). In the murine models, succinate levels in intestinal content samples were also higher in the NEC group than in the control group (P <0.05). The change in succinate level was closely related to intestinal flora composition. In samples from human neonates, relative to the control group, the NEC group showed a higher abundance of Enterobacteriaceae and a lower abundance of Lactobacillaceae and Lactobacillus (P <0.05). In the murine models, relative to the control group, increased abundance was observed for Clostridiaceae, Enterococcaceae, Clostridium_sensu_stricto_1, and Enterococcus, whereas decreased abundance was observed for Lactobacillaceae and Lactobacillus (P <0.05). Increased succinate levels prevented mice from gaining weight, damaged their intestines, and increased their mortality; upregulated the gene expression of interleukin-1ß (IL-1ß), IL-6, IL-18 and tumor necrosis factor (TNF); and downregulated the gene expression of IL-10 and transforming growth factor (TGF)-ß. Exogenous succinic acid increased inducible nitric oxide synthase (iNOS) gene expression but decreased Arginase-1 (Arg1) gene expression; and increased the protein expression of SUCNR1 and HIF-1a. Conclusion: Succinate plays an important role in the development of necrotizing enterocolitis severity, and the activation of the HIF-1a signaling pathway may lead to disease progression.


Subject(s)
Enterocolitis, Necrotizing , Intestinal Diseases , Animals , Mice , Animals, Newborn , Disease Models, Animal , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , RNA, Ribosomal, 16S/genetics , Succinic Acid , Tandem Mass Spectrometry , Humans , Infant, Newborn
5.
Front Neurosci ; 16: 849372, 2022.
Article in English | MEDLINE | ID: mdl-35573292

ABSTRACT

Very preterm infants who survive are at high risk of white matter injury (WMI). With a greater understanding of the pathogenesis of WMI, the gut microbiota has recently drawn increasing attention in this field. This review tries to clarify the possible mechanisms behind the communication of the gut bacteria and the immature brain via the gut-brain axis. The gut microbiota releases signals, such as microbial metabolites. These metabolites regulate inflammatory and immune responses characterized by microglial activation, which ultimately impact the differentiation of pre-myelinating oligodendrocytes (pre-OLs) and lead to WMI. Moreover, probiotics and prebiotics emerge as a promising therapy to improve the neurodevelopmental outcome. However, future studies are required to clarify the function of these above products and the optimal time for their administration within a larger population. Based on the existing evidence, it is still too early to recommend probiotics and prebiotics as effective treatments for WMI.

SELECTION OF CITATIONS
SEARCH DETAIL