Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters











Publication year range
1.
J Agric Food Chem ; 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39249130

ABSTRACT

Flavonoid natural products are emerging as a promising approach for treating Ulcerative Colitis (UC) due to their natural origin and minimal toxicity. This study investigates the effects of Neohesperidin (NEO), a natural flavonoid, on Dextran Sodium Sulfate (DSS)-induced UC in mice, focusing on the underlying molecular mechanisms. Early intervention with NEO (25 and 50 mg/kg) mitigated colon shortening, restored damaged barrier proteins, and significantly reduced the inflammatory cytokine levels. Moreover, NEO inhibited the MAPK/NF-κB signaling pathway and enhanced the levels of intestinal barrier proteins (Claudin-3 and ZO-1). Additionally, NEO increased beneficial intestinal probiotics (S24-7 and Lactobacillaceae) while reducing harmful bacteria (Erysipelotrichi, Enterobacteriaceae). Fecal microbial transplantation (FMT) results demonstrated that NEO (50 mg/kg) markedly improved UC symptoms. In conclusion, early NEO intervention may alleviate DSS-induced UC by inhibiting inflammatory responses, preserving intestinal barrier integrity and modulating gut microbiota.

2.
J Dairy Sci ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38876213

ABSTRACT

High-yield dairy cows typically undergo intense cellular metabolism, leading to oxidative stress in their mammary tissues. Our study found that these high-yield cows had significantly elevated levels of hydrogen peroxide (H2O2), lipoperoxidase, and total antioxidant capacity in their blood, compared with ordinary cows. This increased oxidative stress is associated with heightened expression of genes such as GCLC, GCLM and SIRT1 and proteins such as SIRT1 in the mammary tissue of high-yield cows. MAC-T cells were stimulated with H2O2 at a concentration equal to the average H2O2 level in the serum of ethically high-yielding cows, as detected by an assay kit. Our observations revealed that short-term exposure (12 h) to H2O2 upregulated the expression of SIRT1 gene and protein. It also increased gene expression for SOD2, CAT, GCLC, GCLM, PGC-1α, and NQO1, elevated the phosphorylation of AMPK, and enhanced protein expression of PGC-1α, NQO1, Nrf2, and HO-1, while reducing the phosphorylation of NF-κB. Additionally, short-term H2O2 stimulation resulted in increased total antioxidant capacity, SOD, GSH, and CAT levels in the mammary epithelial cells of dairy cows. In contrast, prolonged exposure to H2O2 (24 h) yielded opposite results, indicating reduced antioxidant capacity. Further investigation showed that SIRT1 inhibitor (EX 527) could reverse the enhanced cellular antioxidant capacity triggered by short-term oxidative stress. However, it is crucial to note that while 12 h H2O2 stimulation improved antioxidant capacity, reactive oxygen species (ROS) and malondialdehyde (MDA) levels inside the cell gradually increased over time, suggesting greater damage under long-term stimulation. Conversely, the SIRT1 activator (SRT 2104) could reverse the reduced cellular antioxidant capacity caused by long-term oxidative stress and significantly inhibit the accumulation of ROS and MDA. Notably, SRT 2104 demonstrated similar effects in MAC-T cells during lactation. In summary, SIRT1 plays a crucial role in regulating the antioxidant capacity of mammary epithelial cells in dairy cows. This discovery provides valuable insights into the antioxidant mechanisms of mammary cells, which can serve as a theoretical foundation for future mammary health strategies.

3.
Phytomedicine ; 130: 155730, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38759313

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a prolonged inflammatory disease of the gastrointestinal tract. Current therapeutic options remain limited, underscoring the imperative to explore novel therapeutic strategies. Narirutin (NR), a flavonoid naturally present in citrus fruits, exhibits excellent anti-inflammatory effects in vitro, yet its in vivo efficacy, especially in UC, remains underexplored. OBJECTIVE: This work examined the effect of NR on dextrose sodium sulfate (DSS)-induced UC in mice in vivo, with a specific focus on the role of gut flora in it. METHODS: The effects of NR (10, 20, and 40 mg/kg) on DSS-induced UC in mice were investigated by monitoring changes in body weight, disease activity index (DAI) scores, colon length, and histological damage. Colonic levels of pro-inflammatory mediators, tight junction (TJ) proteins, and inflammation-related signaling pathway proteins were analyzed via enzyme-linked immunosorbent assay, western blot, and immunofluorescence. The role of gut microbiota in NR against colitis was analyzed through 16S rRNA sequencing, flora clearance assays, and fecal microbiota transplantation (FMT) assays. RESULTS: NR administration suppressed DSS-induced colitis as reflected in a decrease in body weight loss, DAI score, colon length shortening, and histological score. Furthermore, NR administration preserved the integrity of the DSS-induced intestinal barrier by inhibiting the reduction of TJ proteins (claudin3, occludin, and zonula occludens-1). Moreover, NR administration markedly repressed the activation of the toll-like receptor 4-mitogen-activated protein kinase/nuclear factor-κB pathway and reduced the amount of pro-inflammatory mediators in the colon. Importantly, the results of 16S rRNA sequencing showed that the intestinal flora of mice with colitis exhibited richer microbial diversity following NR administration, with elevated abundance of Lactobacillaceae (Lactobacillus) and decreased abundance of Bacteroidaceae (Bacteroides) and Shigella. In addition, the anti-colitis effect of NR almost disappeared after gut flora clearance. Further FMT assay also validated this gut flora-dependent protective mechanism of NR. CONCLUSION: Our findings suggest that NR is a prospective natural compound for the management of UC by modulating intestinal flora.


Subject(s)
Colon , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Gastrointestinal Microbiome/drug effects , Mice , Male , Colon/drug effects , Colon/pathology , Glucose/metabolism , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Dextran Sulfate , Flavanones/pharmacology , Anti-Inflammatory Agents/pharmacology , Disease Models, Animal , NF-kappa B/metabolism , Fecal Microbiota Transplantation , Colitis/chemically induced , Colitis/drug therapy , Citrus/chemistry , Tight Junction Proteins/metabolism , Sulfates/pharmacology
4.
Theor Appl Genet ; 137(6): 126, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727833

ABSTRACT

KEY MESSAGE: The gene controlling pink flesh in watermelon was finely mapped to a 55.26-kb region on chromosome 6. The prime candidate gene, Cla97C06G122120 (ClPPR5), was identified through forward genetics. Carotenoids offer numerous health benefits; while, they cannot be synthesized by the human body. Watermelon stands out as one of the richest sources of carotenoids. In this study, genetic generations derived from parental lines W15-059 (red flesh) and JQ13-3 (pink flesh) revealed the presence of the recessive gene Clpf responsible for the pink flesh (pf) trait in watermelon. Comparative analysis of pigment components and microstructure indicated that the disparity in flesh color between the parental lines primarily stemmed from variations in lycopene content, as well as differences in chromoplast number and size. Subsequent bulk segregant analysis (BSA-seq) and genetic mapping successfully narrowed down the Clpf locus to a 55.26-kb region on chromosome 6, harboring two candidate genes. Through sequence comparison and gene expression analysis, Cla97C06G122120 (annotated as a pentatricopeptide repeat, PPR) was predicted as the prime candidate gene related to pink flesh trait. To further investigate the role of the PPR gene, its homologous gene in tomato was silenced using a virus-induced system. The resulting silenced fruit lines displayed diminished carotenoid accumulation compared with the wild-type, indicating the potential regulatory function of the PPR gene in pigment accumulation. This study significantly contributes to our understanding of the forward genetics underlying watermelon flesh traits, particularly in relation to carotenoid accumulation. The findings lay essential groundwork for elucidating mechanisms governing pigment synthesis and deposition in watermelon flesh, thereby providing valuable insights for future breeding strategies aimed at enhancing fruit quality and nutritional value.


Subject(s)
Chromosome Mapping , Citrullus , Fruit , Phenotype , Pigmentation , Plant Proteins , Citrullus/genetics , Citrullus/metabolism , Pigmentation/genetics , Plant Proteins/genetics , Plant Proteins/metabolism , Fruit/genetics , Genes, Plant , Carotenoids/metabolism , Genes, Recessive , Gene Expression Regulation, Plant , Chromosomes, Plant/genetics , Lycopene/metabolism
5.
Phytomedicine ; 130: 155741, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38772182

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is a chronic recurrent intestinal disease lacking effective treatments. ß-arbutin, a glycoside extracted from the Arctostaphylos uva-ursi leaves, that can regulate many pathological processes. However, the effects of ß-arbutin on UC remain unknown. PURPOSE: In this study, we investigated the role of ß-arbutin in relieving colitis and explored its potential mechanisms in a mouse model of dextran sulfate sodium (DSS)-induced colitis. METHODS: In C75BL/6 J mice, DSS was used to induce colitis and concomitantly ß-arbutin (50 and 100 mg/kg) was taken orally to evaluate its curative effect by evaluating disease activity index (DAI) score, colon length and histopathology. Alcian blue periodic acid schiff (AB-PAS) staining, immunohistochemistry (IHC), immunofluorescence (IF) and TdT-mediated dUTP Nick-End Labeling (Tunel) staining were used to assess intestinal barrier function. Flow cytometry, double-IF and western blotting (WB) were performed to verify the regulatory mechanism of ß-arbutin on neutrophil extracellular traps (NETs) in vivo and in vitro. NETs depletion experiments were used to demonstrate the role of NETs in UC. Subsequently, the 16S rRNA gene sequencing was used to analyze the intestinal microflora of mouse. RESULTS: Our results showed that ß-arbutin can protect mice from DSS-induced colitis characterized by a lower DAI score and intestinal pathological damage. ß-arbutin reduced inflammatory factors secretion, notably regulated neutrophil functions, and inhibited NETs formation in an ErK-dependent pathway, contributing to the resistance to colitis as demonstrated by in vivo and in vitro experiments. Meanwhile, remodeled the intestinal flora structure and increased the diversity and richness of intestinal microbiota, especially the abundance of probiotics and butyric acid-producing bacteria. It further promoted the protective effect in the resistance of colitis. CONCLUSION: ß-arbutin promoted the maintenance of intestinal homeostasis by inhibiting NETs formation, maintaining mucosal-barrier integrity, and shaping gut-microbiota composition, thereby alleviating DSS-induced colitis. This study provided a scientific basis for the rational use of ß-arbutin in preventing colitis and other related diseases.


Subject(s)
Arbutin , Dextran Sulfate , Disease Models, Animal , Extracellular Traps , Gastrointestinal Microbiome , Mice, Inbred C57BL , Animals , Extracellular Traps/drug effects , Gastrointestinal Microbiome/drug effects , Mice , Arbutin/pharmacology , Male , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/chemically induced , Neutrophils/drug effects , Colitis/drug therapy , Colitis/chemically induced , Colon/drug effects , Colon/pathology
6.
Life Sci ; 342: 122533, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38428570

ABSTRACT

The World Health Organization recommends breastfeeding for 6 months, but mastitis, a common disease during lactation, presents a major obstacle to fulfilling this recommendation. Maternal nutrient intake during lactation has been shown to be related to mastitis. Therefore, this study aimed to explore the effect of hesperetin, a phytonutrient, on mastitis. The oral administration of hesperetin to lipopolysaccharide (LPS)-induced mastitis mice alleviated their pathological damage, reduced the secretion of pro-inflammatory cytokines, and maintained the integrity of their blood-milk barrier. Moreover, our results showed that oral administration of hesperetin regulates the composition of the intestinal flora of mice. Fecal microbial transplantation (FMT) from the mice of hesperetin group alleviated LPS-induced mastitis in recipient mice. In additional, hesperetin attenuated the inflammatory response and increased the expression of tight junction proteins (TJs) in LPS-stimulated mouse mammary epithelial cells (mMECs). Through network pharmacological analysis and further research, we demonstrated hesperetin inhibits the expression of TLR4 and the activation of NF-κB signaling. In conclusion, hesperetin protects the blood-milk barrier and improve mastitis by regulating intestinal flora and inhibiting the activation of TLR4/NF-κB signaling axis. This study provides a theoretical basis for lactating females to consume hesperetin as a supplement to prevent mastitis and maintain mammary health.


Subject(s)
Gastrointestinal Microbiome , Hesperidin , Mastitis , Humans , Female , Animals , Mice , NF-kappa B/metabolism , Toll-Like Receptor 4/metabolism , Milk/metabolism , Lactation , Lipopolysaccharides/adverse effects , Mastitis/prevention & control , Mastitis/metabolism , Mastitis/pathology , Mammary Glands, Animal/metabolism
7.
Cells ; 13(5)2024 Mar 03.
Article in English | MEDLINE | ID: mdl-38474407

ABSTRACT

Inflammatory bowel disease (IBD) refers to a cluster of intractable gastrointestinal disorders with an undetermined etiology and a lack of effective therapeutic agents. Amygdalin (Amy) is a glycoside extracted from the seeds of apricot and other Rosaceae plants and it exhibits a wide range of pharmacological properties. Here, the effects and mechanisms of Amy on colitis were examined via 16S rRNA sequencing, ELISA, transmission electron microscopy, Western blot, and immunofluorescence. The results showed that Amy administration remarkably attenuated the signs of colitis (reduced body weight, increased disease activity index, and shortened colon length) and histopathological damage in dextran sodium sulfate (DSS)-challenged mice. Further studies revealed that Amy administration significantly diminished DSS-triggered gut barrier dysfunction by lowering pro-inflammatory mediator levels, inhibiting oxidative stress, and reducing intestinal epithelial apoptosis and ferroptosis. Notably, Amy administration remarkably lowered DSS-triggered TLR4 expression and the phosphorylation of proteins related to the NF-κB and MAPK pathways. Furthermore, Amy administration modulated the balance of intestinal flora, including a selective rise in the abundance of S24-7 and a decline in the abundance of Allobaculum, Oscillospira, Bacteroides, Sutterella, and Shigella. In conclusion, Amy can alleviate colitis, which provides data to support the utility of Amy in combating IBD.


Subject(s)
Amygdalin , Colitis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Animals , Mice , RNA, Ribosomal, 16S , Cell Death , Dextran Sulfate
8.
Int J Biol Macromol ; 254(Pt 3): 127786, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37918588

ABSTRACT

Valine, a branched-chain amino acid found in dairy cows, has been recognized for its critical role in milk synthesis. However, the precise effect of valine on lactation in dairy cows remains an area of investigation. In our study, bovine mammary epithelial cells (BMECs) were isolated to explore the mechanism through which valine enhances milk synthesis. The results showed that 100 µM valine significantly boosted the milk synthesis via TAS1R1-mTOR-DDX39B signaling pathway in BMECs. Subsequent investigations revealed that DDX39B governs the accumulation of PKM2 in the nuclei of BMECs. This nuclear buildup of PKM2 weakened the interaction between HDAC3 and histone H3, leading to an increase in the acetylation levels of histone H3. In an vivo context, the 0.25 % valine-enriched drinking water notably elevated in the expression of milk protein and fat in these mice. Further examination showed that 0.25 % valine drinking water considerably augmented the protein expression levels of DDX39B, PKM2, and p-mTOR in the mice mammary glands. In summary, our results suggest that valine, by modulating the TAS1R1-mTOR-DDX39B signaling pathway, directs the accumulation of PKM2 in the nucleus. This, in turn, escalates the acetylation levels of histone H3, promoting the synthesis of both milk protein and fat.


Subject(s)
Drinking Water , Histones , Female , Animals , Cattle , Mice , Histones/metabolism , Valine/metabolism , Acetylation , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Milk Proteins/metabolism , Epithelial Cells
9.
J Agric Food Chem ; 71(43): 16043-16056, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37856155

ABSTRACT

Phlorizin (PHZ) is the main active component of apple peel and presents a potential application value. In the past few years, some reports have suggested that PHZ may have antioxidant and anti-inflammatory effects. Herein, we have attempted to assess the protective effects of PHZ on dextran sodium sulfate (DSS)-induced colitis in mice and to determine the underlying molecular mechanisms. Our results suggested that early intervention with PHZ (20, 40, and 80 mg/kg) significantly reduced the severity of DSS-induced colitis in mice, as presented by a longer colon, improved tight junction protein, decreased disease activity index, and attenuated inflammatory factors. Additionally, early intervention with + (20, 40, and 80 mg/kg) significantly inhibited ferroptosis by decreasing the surrogate ferroptosis marker levels (MDA and Iron Content). Additionally, PHZ (80 mg/kg) increased the diversity of intestinal flora in colitic mice by elevating the levels of beneficial bacteria (Lactobacillaceae and Muribaculaceae) and reducing the levels of harmful bacteria (Lachnospiraceae). This indirectly led to an increase in the amount of short-chain fatty acids. A fecal microbial transplantation (FMT) test was conducted to show that PHZ (80 mg/kg) ameliorated ulcerative colitis (UC) by regulating gut dysbiosis. In conclusion, early intervention with PHZ decreased DSS-induced colitis in mice by preserving their intestinal barrier and regulating their intestinal flora.


Subject(s)
Colitis, Ulcerative , Colitis , Ferroptosis , Gastrointestinal Microbiome , Animals , Mice , Phlorhizin , Dextran Sulfate/adverse effects , Colitis/chemically induced , Colitis/drug therapy , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colon , Mice, Inbred C57BL , Disease Models, Animal
10.
Int Immunopharmacol ; 122: 110551, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37406397

ABSTRACT

Mastitis occurs frequently in breastfeeding women and not only affects the women's health but also hinders breastfeeding. Maslinic acid is a type of pentacyclic triterpenoid widely found in olives that has good anti-inflammatory activity. This study aims to discuss the protective function of maslinic acid against mastitis and its underlying mechanism. For this, mice models of mastitis were established using lipopolysaccharide (LPS). The results revealed that maslinic acid reduced the pathological lesions in the mammary gland. In addition, it reduced the generation of pro-inflammatory factors and enzymes (IL-6, IL-1ß, TNF-α, iNOS, and COX2) in both mice mammary tissue and mammary epithelial cells. The high-throughput 16S rDNA sequencing of intestinal flora showed that in mice with mastitis, maslinic acid treatment altered ß-diversity and regulated microbial structure by increasing the abundance of probiotics such as Enterobacteriaceae and downregulating harmful bacteria such as Streptococcaceae. In addition, maslinic acid protected the blood-milk barrier by maintaining tight-junction protein expression. Furthermore, maslinic acid downregulated mammary inflammation by inhibiting the activation of NLRP3 inflammasome, AKT/NF-κB, and MAPK signaling pathways. Thus, in a mice model of LPS-induced mastitis, maslinic acid can inhibit the inflammatory response, protect the blood-milk barrier, and regulate the constitution of intestinal flora.


Subject(s)
Gastrointestinal Microbiome , Mastitis , Humans , Female , Animals , Mice , Lipopolysaccharides/pharmacology , Milk/metabolism , Mastitis/chemically induced , Mastitis/drug therapy , Mastitis/metabolism , NF-kappa B/metabolism , Mammary Glands, Animal/pathology
11.
Vet Microbiol ; 280: 109697, 2023 May.
Article in English | MEDLINE | ID: mdl-36827937

ABSTRACT

Mitophagy occurs in a variety of pathogenic infections. However, the role of mitophagy in the intracellular survival of Staphylococcus aureus (S.aureus) within bovine mammary epithelial cells (BMECs) and which molecules specifically mediate the induction of mitophagy remains unclear. Therefore, this study aims to investigate the role and mechanism of mitophagy in the intracellular survival of S.aureus. Here, we reported that S.aureus induced complete mitophagy to promote its survival within BMECs. The further mechanistic study showed that S. aureus induced mitophagy by activating the p38-PINK1-Parkin signaling pathway. These findings expand our knowledge of the intracellular survival mechanism of S.aureus in the host and provide a desirable therapeutic strategy against S.aureus and other intracellular infections.


Subject(s)
Cattle Diseases , Staphylococcal Infections , Cattle , Animals , Staphylococcus aureus , Mitophagy , Signal Transduction , Epithelial Cells/metabolism , Staphylococcal Infections/veterinary , Staphylococcal Infections/drug therapy , Ubiquitin-Protein Ligases/metabolism , Cattle Diseases/metabolism
12.
Int Immunopharmacol ; 115: 109698, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36634417

ABSTRACT

Our previous study showed that α-Cyperone inhibited the inflammatory response triggered by activated microglia and protected dopaminergic neuron in in vitro cell model of Parkinson's disease (PD). It is unclear the effect of α-Cyperone in animal models of PD. In this study, our results indicated that α-Cyperone ameliorated motor dysfunction, protected dopaminergic neurons, and inhibited the reduction of dopamine and its metabolites in lipopolysaccharide (LPS)-induced PD rat model. Moreover, α-Cyperone suppressed the activation of microglia and the expression of neuroinflammatory factor (TNF-α, IL-6, IL-1ß, iNOS, COX-2 and ROS). Furthermore, the molecular mechanism research revealed that α-Cyperone inhibited neuroinflammation and oxidative stress to exert protective effect in microglia by activating Nrf2/HO-1 and suppressing NF-κB signaling pathway. Moreover, α-Cyperone upregulated the expression of antioxidant enzymes (GCLC, GCLM and NQO1) in microglia. In conclusion, our study demonstrates α-Cyperone alleviates dopaminergic neurodegeneration by inhibiting neuroinflammation and oxidative stress in LPS-induced PD rat model via activating Nrf2/HO-1 and suppressing NF-κB signaling pathway.


Subject(s)
NF-kappa B , Parkinson Disease , Rats , Animals , NF-kappa B/metabolism , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , Lipopolysaccharides/pharmacology , Dopaminergic Neurons , NF-E2-Related Factor 2/metabolism , Neuroinflammatory Diseases , Anti-Inflammatory Agents/pharmacology , Signal Transduction , Microglia
13.
J Therm Biol ; 110: 103375, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36462887

ABSTRACT

Lysine, as the first limiting amino acid in dairy cows, has been shown to play an important role in milk synthesis and cell proliferation. However, the underlying mechanism remains unclear. In this study, we isolated bovine primary mammary epithelial cells (BMECs) and studied the mechanism in which lysine promotes cell proliferation and ß-casein synthesis through overexpression and knockdown of CDK1 and supplements BCH, U0126, and rapamycin in BMECs. Results show that 0.7 mM lysine can significantly promote cell proliferation and the synthesis of ß-casein in BMECs. In addition, lysine activates the ERK signaling pathway to promote the expression of CDK1. Further studies have shown that CDK1 can promote cell proliferation and the synthesis of ß-casein through the mTOR signaling pathway in BMECs. Lastly, lysine can promote cell proliferation and the synthesis of ß-casein through SLC6A14 in BMECs. The above results indicate that lysine promotes cell proliferation and the synthesis of ß-casein through the SLC6A14-ERK-CDK1-mTOR signaling pathway in BMECs.


Subject(s)
Caseins , MAP Kinase Signaling System , Female , Cattle , Animals , Lysine , Signal Transduction , Epithelial Cells , Cell Proliferation , TOR Serine-Threonine Kinases
14.
Metabolites ; 12(11)2022 Oct 28.
Article in English | MEDLINE | ID: mdl-36355118

ABSTRACT

Dairy cow mammary gland fibrosis causes huge economic losses to livestock production, however, research on dairy cow mammary gland fibrosis is in its infancy and it lacks effective treatments. Therefore, the purpose of this experiment was to explore the correlation between mastitis and fibrosis and mitochondrial damage, and to further explore its pathogenesis. In vivo, mammary tissue and milk samples were collected from healthy cows (n = 10) and mastitis cows (n = 10). The results of the study showed that compared with the control group, the mastitis tissue showed tissue damage, accumulation of collagen fibers, and the content of TGF-ß1 in mammary tissue and milk was significantly increased; the level of inflammatory mediators was significantly increased; the fibrotic phenotype, collagen 1, α-SMA, vimentin gene, and protein levels were significantly increased, while the E-cadherin gene and protein levels were significantly decreased. In vitro, based on TGF-ß1-induced bMECs, the above experimental results were further confirmed, and TGF-ß1 significantly promoted the fibrotic phenotype of bMECs. On the other hand, in vivo results showed that fibrotic mammary tissue had a significantly stronger mitochondrial damage phenotype and significantly higher ROS than the control group. In vitro, the results also found that TGF-ß1 induced a significant increase in the mitochondrial damage phenotype of bMECs, accompanied by a large amount of ROS production. Furthermore, in a TGF-ß1-induced bMEC model, inhibiting the accumulation of ROS effectively alleviated the elevated fibrotic phenotype of TGF-ß1-induced bMECs. In conclusion, the fibrotic phenotype of mammary gland tissue in dairy cows with mastitis was significantly increased, and mastitis disease was positively correlated with mammary fibrotic lesions. In an in vitro and in vivo model of cow mammary fibrosis, bMECs have impaired mitochondrial structure and dysfunction. Inhibiting the accumulation of ROS effectively alleviates the elevated fibrotic phenotype, which may be a potential therapeutic approach to alleviate mammary fibrosis.

15.
J Agric Food Chem ; 70(46): 14718-14731, 2022 Nov 23.
Article in English | MEDLINE | ID: mdl-36375817

ABSTRACT

Ulcerative colitis (UC), one of the foremost common forms of inflammatory bowel disease, poses a serious threat to human health. Currently, safe and effective treatments are not available. This study investigated the protective effect of ginkgolide C (GC), a terpene lactone extracted from Ginkgo biloba leaves, on UC and its underlying mechanism. The results showed that GC remarkably mitigated the severity of DSS-induced colitis in mice, as demonstrated by decreased body weight loss, reduced disease activity index, mitigated tissue damage, and increased colon length. Furthermore, GC inhibited DSS-induced hyperactivation of inflammation-related signaling pathways (NF-κB and MAPK) to reduce the production of inflammatory mediators, thereby mitigating the inflammatory response in mice. GC administration also restored gut barrier function by elevating the number of goblet cells and boosting the levels of tight junction-related proteins (claudin-3, occludin, and ZO-1). In addition, GC rebalanced the intestinal flora of DSS-treated mice by increasing the diversity of the flora, elevating the abundance of beneficial bacteria, such as Lactobacillus and Allobaculum, and decreasing the abundance of harmful bacteria, such as Bacteroides, Oscillospira, Ruminococcus, and Turicibacter. Taken together, these results suggest that GC administration effectively alleviates DSS-induced colitis by inhibiting the inflammatory response, maintaining mucosal barrier integrity, and regulating intestinal flora. This study may provide a scientific basis for the rational use of GC in preventing colitis and other related diseases.


Subject(s)
Colitis, Ulcerative , Colitis , Gastrointestinal Microbiome , Mice , Humans , Animals , Dextran Sulfate/metabolism , Disease Models, Animal , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Lactones/metabolism , Colitis, Ulcerative/metabolism , Colon/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Tight Junction Proteins/genetics , Tight Junction Proteins/metabolism , Mice, Inbred C57BL
16.
Virulence ; 13(1): 1684-1696, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36128739

ABSTRACT

Staphylococcus aureus can survive within phagocytes. Indeed, we confirm in this study that approximately 10% of population persists in macrophages during S. aureus infection, while the rest are eliminated due to bacteriolysis, which is of particular interest to us. Herein, we observe that the bacteriolysis is an early event accompanied by macrophage death during S. aureus infection. Furthermore, the cell death is significantly accelerated following increased intracellular bacteriolysis, indicating that intracellular bacteriolysis induces cell death. Subsequently, we establish that the cell death is not apoptosis or pyroptosis, but AIM2-mediated necroptosis, accompanied by AIM2 inflammasome activation. This finding challenges the classical model that the cell death that accompanies inflammasome activation is always pyroptosis. In addition, we observe that the apoptosis-associated genes are highly inhibited during S. aureus infection. Finally, we establish in vivo that increased bacteriolysis significantly enhances S. aureus pathogenicity by promoting its dissemination to kidney and leading to an inflammatory cytokine storm in AIM2-mediated manner. Collectively, our data demonstrate that bacteriolysis is detrimental when triggered in excess and its side effect is mediated by AIM2. Meanwhile, we propose a potential immune manipulation strategy by which S. aureus sacrifices the minority to trigger a limited necroptosis, thereby releasing signals from dead cells to inhibit apoptosis and other anti-inflammatory cascades of live cells, eventually surviving within host cells and establishing infection.


Subject(s)
Inflammasomes , Staphylococcal Infections , Bacteriolysis , DNA-Binding Proteins/genetics , Humans , Inflammasomes/genetics , Inflammation , Necroptosis , Staphylococcus aureus/genetics , Staphylococcus aureus/metabolism , Virulence
17.
J Anim Physiol Anim Nutr (Berl) ; 106(6): 1420-1430, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35923149

ABSTRACT

Lysine is one of the essential amino acids. The effect of lysine on milk protein and milk fat anabolism has been reported, but the effect on mammary glands development has not been studied in detail. The normal development of the mammary glands at puberty is crucial to lactation of mammals. In this study, to explore the effect of lysine on mammary glands development, we fed different concentrations of lysine (0.025%, 0.05%, 0.1%) to pubertal mice and found that the addition of 0.1% lysine to drinking water significantly promoted mammary glands development. Furthermore, we treated mMECs (mouse mammary epithelial cells) with different concentrations of lysine (0, 0.2, 0.4, 0.6, 0.8 and 1 mM) to explore the underlying mechanism, and found that lysine promoted the proliferation of mMECs and development of mammary glands through PI3K/AKT/mTOR signalling pathway in pubertal mice. Overall, the results of this study revealed that lysine activated the PI3K/AKT/mTOR signal axis, elevated protein concentrations of cell proliferation markers, such as PCNA, Cyclin D1 and D3, and enhanced the proliferation of mMECs, finally promoted the murine mammary glands development at puberty.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Female , Mice , Animals , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Lysine/pharmacology , Mammary Glands, Animal , Sexual Maturation , TOR Serine-Threonine Kinases/metabolism , Epithelial Cells , Mammals/metabolism
18.
J Agric Food Chem ; 70(36): 11401-11411, 2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36040330

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) induced by obesity is a grave threat to human health. Phytic acid (PA) is a natural compound found in high-fiber diets, such as soybeans. This study investigated the effects and mechanisms of PA on obesity, hepatic lipid metabolism, and gut-liver axis homeostasis in high-fat diet (HFD)-fed mice. PA was observed to significantly inhibit obesity and alleviate liver steatosis in mice. PA improved HFD-induced liver inflammation, oxidative stress and fibrosis. Moreover, PA improved HFD-induced colonic inflammation, gut barrier damage and systemic inflammation in mice. Furthermore, PA effectively ameliorated the decreased diversity and gut microbiota composition in HFD-fed mice. Additionally, PA decreased the abundance of harmful bacteria Proteobacteria and Desulfovibrionaceae and increased the abundance of probiotic bacteria Muribaculaceae and Lachnospiraceae. Thus, PA is effective in restoring the homeostasis of the gut-liver axis. It further provides a theoretical basis for the prevention and treatment of NAFLD in patients with obesity by the rational intake of foods containing PA.


Subject(s)
Diet, High-Fat , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat/adverse effects , Humans , Inflammation/drug therapy , Inflammation/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/drug therapy , Obesity/etiology , Obesity/metabolism , Oxidative Stress , Phytic Acid/metabolism
19.
J Agric Food Chem ; 70(36): 11324-11335, 2022 Sep 14.
Article in English | MEDLINE | ID: mdl-36040348

ABSTRACT

Cell-death-inducing DNA fragmentation factor-α-like effector A (CIDEA) is a lipid-droplet-associated protein that helps to promote lipid metabolism in adipocytes of mice and humans. However, studies on the regulatory mechanism of CIDEA on lipid metabolism in the mammary glands of dairy cows are rare. Therefore, the role of CIDEA in bovine mammary epithelial cells (bMECs) was investigated in this study. The CIDEA expression levels in the mammary glands of high-fat-milk-producing cows were significantly higher compared to those in low-fat-milk-producing cows. Results of in vitro studies in bMECs showed that the inhibition of CIDEA inhibited the expression of fatty acid synthesis-related genes and triglyceride (TAG) synthesis-related genes. Conversely, the overexpression of CIDEA leads to an increase in the content of TAG and fatty acid. The results of mechanistic studies indicated that the overexpression of CIDEA inhibits AMP-activated protein kinase (AMPK) activity, which enhances the expression of peroxisome proliferator-activated receptor-γ (PPARγ) and consequently increases the TAG content. Furthermore, the overexpression of CIDEA promoted the nuclear translocation of sterol regulatory element-binding protein 1 (SREBP1). Therefore, a theoretical framework is provided by this study for the regulation of lipid metabolism in dairy cows by means of nutrition and the hormone targeting of CIDEA.


Subject(s)
Mammary Glands, Animal , PPAR gamma , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cattle , Epithelial Cells/metabolism , Fatty Acids/metabolism , Female , Humans , Mammary Glands, Animal/metabolism , PPAR gamma/genetics , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism
20.
J Anim Physiol Anim Nutr (Berl) ; 106(6): 1383-1395, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35616019

ABSTRACT

Amino acids have been shown to affect the development of mammary gland (MG). However, it is unclear whether L-arginine promotes the development of pubertal MG. Therefore, our study aims to explore the effect of L-arginine on the development of MG in pubertal mice. To investigate its internal mechanism of action, we will use mouse mammary epithelial cells (mMECs) line. Whole-mount staining showed that L-arginine can promote the extension of MG duct. In vitro, 0.4 mM L-arginine could activate the G protein-coupled receptor family C, group 6, subtype A (GPRC6A)/phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signalling pathway and increase the phosphorylation of eukaryotic initiation factor 4E binding protein 1 (4EBP1) to promote the synthesis of cell cycle regulatory protein D1 (Cyclin D1), leading to the dissociation of the retinoblastoma tumour suppressor protein (Rb)-E2F1 transcription factor (E2F1) complex in mMECs and releasing E2F1 to promote cell proliferation. Furthermore, GPRC6A was knocked down or inhibition of the PI3K/AKT/mTOR signalling pathway with corresponding inhibitors completely abolished the arginine-induced promotion of mMECs proliferation. In vivo, it was further confirmed that 0.1% L-arginine can activate the PI3K/AKT/mTOR signalling pathway in the MG of pubertal mice. These results were able to indicate that L-arginine stimulates the development of MG in pubertal mice through the GPRC6A/PI3K/AKT/mTOR signalling pathway.


Subject(s)
Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Animals , Mice , Arginine/pharmacology , Cell Cycle Proteins , Cell Proliferation , Epithelial Cells/metabolism , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, G-Protein-Coupled/metabolism , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL