Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Redox Biol ; 72: 103160, 2024 06.
Article in English | MEDLINE | ID: mdl-38631120

ABSTRACT

Iron overload can lead to oxidative stress and intestinal damage and happens frequently during blood transfusions and iron supplementation. However, how iron overload influences intestinal mucosa remains unknown. Here, the aim of current study was to investigate the effects of iron overload on the proliferation and differentiation of intestinal stem cells (ISCs). An iron overload mouse model was established by intraperitoneal injection of 120 mg/kg body weight iron dextran once a fortnight for a duration of 12 weeks, and an iron overload enteroid model was produced by treatment with 3 mM or 10 mM of ferric ammonium citrate for 24 h. We found that iron overload caused damage to intestinal morphology with a 64 % reduction in villus height/crypt depth ratio, and microvilli injury in the duodenum. Iron overload mediated epithelial function by inhibiting the expression of nutrient transporters and enhancing the expression of secretory factors in the duodenum. Meanwhile, iron overload inhibited the proliferation of ISCs and regulated their differentiation into secretory mature cells, such as goblet cells, through inhibiting Notch signaling pathway both in mice and enteroid. Furthermore, iron overload caused oxidative stress and ferroptosis in intestinal epithelial cells. In addition, ferroptosis could also inhibit Notch signaling pathway, and affected the proliferation and differentiation of ISCs. These findings reveal the regulatory role of iron overload on the proliferation and differentiation of ISCs, providing a new insight into the internal mechanism of iron overload affecting intestinal health, and offering important theoretical basis for the scientific application of iron nutrition regulation.


Subject(s)
Cell Differentiation , Ferroptosis , Goblet Cells , Iron Overload , Oxidative Stress , Receptors, Notch , Signal Transduction , Stem Cells , Animals , Ferroptosis/drug effects , Mice , Goblet Cells/metabolism , Iron Overload/metabolism , Signal Transduction/drug effects , Stem Cells/metabolism , Stem Cells/cytology , Cell Differentiation/drug effects , Receptors, Notch/metabolism , Oxidative Stress/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Male
2.
Animals (Basel) ; 13(19)2023 Oct 03.
Article in English | MEDLINE | ID: mdl-37835698

ABSTRACT

Maternal folic acid intake has important effects on offspring growth and development. The mechanism involved in the renewal of intestinal epithelial cells remains unclear. Thus, this study aimed to investigate the potential effect of maternal folic acid supplementation during gestation and lactation on the structural and functional development of the small intestine in piglet offspring. Twenty-four Duroc sows were assigned to a control group (CON) and a folic-acid-supplemented group (CON + FA, supplemented with 15 mg/kg of folic acid). The results showed that maternal folic acid supplementation throughout gestation and lactation significantly increased the body weight, serum folate level, and intestinal folate metabolism in piglets. It also improved the villus length, villus height-to-crypt depth ratio, and transcript levels of nutrient transporters (GLUT4, SNAT2, FABP2, and SLC7A5) in piglets' duodenum and jejunum. In addition, maternal folic acid supplementation increased Ki67-positive cells and the expression of proliferation-related marker genes (C-Myc, CyclinD1, and PCNA) in piglets' intestinal stem cells. It also boosted the expression of genes associated with mature secreted cells (ChrA, Muc2, Lyz, Vil1), indicating enhanced proliferation and differentiation of intestinal stem cells. These findings demonstrate that maternal folic acid supplementation enhances growth performance and gut health in piglet offspring by promoting epithelial cell renewal equilibrium.

3.
Biomed Pharmacother ; 144: 112253, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607106

ABSTRACT

Iron supplementation is necessary for the treatment of anemia, one of the most frequent complications in inflammatory bowel disease (IBD). However, oral iron supplementation leads to an exacerbation of intestinal inflammation. Gut barrier plays a key role in the pathogenesis of IBD. The aim of this study was to characterize the interrelationship between systemic iron, intestinal barrier and the development of intestinal inflammation in a dextran sulfate sodium (DSS) induced experimental colitis mice model. We found that DSS-treated mice developed severe inflammation of colon, but became much healthy when intraperitoneal injection with iron. Iron supplementation alleviated colonic and systemic inflammation by lower histological scores, restorative morphology of colonic villi, and reduced expression of pro-inflammatory cytokines. Moreover, intraperitoneal supplementation of iron enhanced intestinal barrier function by upregulating the colonic expressions of tight junction proteins, restoring intestinal immune homeostasis by regulating immune cell infiltration and T lymphocyte subsets, and increasing mucous secretion of goblet cells in the colon. High-throughput sequencing of fecal 16 S rRNA showed that iron injection significantly increased the relative abundance of Bacteroidetes, which was suppressed in the gut microbiota of DSS-induced colitis mice. These results provided evidences supporting the protective effects of systemic iron repletion by intraperitoneal injection of iron on intestinal barrier functions. The finding highlights a novel approach for the treatment of IBD with iron injection therapy.


Subject(s)
Colitis/drug therapy , Colon/drug effects , Dietary Supplements , Goblet Cells/drug effects , Intestinal Mucosa/drug effects , Iron-Dextran Complex/administration & dosage , Tight Junction Proteins/metabolism , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Colon/metabolism , Colon/microbiology , Dextran Sulfate , Disease Models, Animal , Dysbiosis , Gastrointestinal Microbiome/drug effects , Goblet Cells/metabolism , Goblet Cells/microbiology , Injections, Intraperitoneal , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice, Inbred C57BL , Permeability , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/microbiology , Up-Regulation
4.
Foods ; 10(8)2021 Aug 01.
Article in English | MEDLINE | ID: mdl-34441564

ABSTRACT

Dysregulation in iron metabolism is associated with obesity, type 2 diabetes, and other metabolic diseases, whereas the underlying mechanisms of imbalanced glycolipid metabolism are still obscure. Here, we demonstrated that iron overload protected mice from obesity both with normal diets (ND) or high-fat diets (HFD). In iron-overload mice, the body fat was significantly decreased, especially when fed with HFD, excessive iron mice gained 15% less weight than those without iron supplements. Moreover, glucose tolerance and insulin sensitivity were all significantly reduced, and hepatic steatosis was prevented. Furthermore, these mice show a considerable decrease in lipogenesis and lipidoses of the liver. Compared with control groups, iron treated groups showed a 79% decrease in the protein level of Perilipin-2 (PLIN2), a protein marker for lipid droplets. These results were consistent with their substantial decrease in adiposity. RNA-seq and signaling pathway analyses showed that iron overload caused ferroptosis in the liver of mice with a decrease in GPX4 expression and an increase in Ptgs2 expression, resulting in a high level of lipid peroxidation. Overall, this study reveals the protective function of iron overload in obesity by triggering the imbalance of glucolipid metabolism in the liver and highlights the crucial role of ferroptosis in regulating lipid accumulation.

5.
J Anim Physiol Anim Nutr (Berl) ; 104(2): 597-605, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31891214

ABSTRACT

In this study, we aimed to determine the effects of dietary supplementation with chitosan nanoparticles (CNP) on growth performance, immune status, gut microbiota and immune responses after lipopolysaccharide challenge in weaned pigs. A total of 144 piglets were assigned to four groups receiving different dietary treatments, including basal diets supplemented with 0, 100, 200 and 400 mg/kg CNP fed for 28 days. Each treatment group included six pens (six piglets per pen). The increase in supplemental CNP concentration improved the average daily gain (ADG) and decreased the feed and gain (F/G) and diarrhoea rate (p < .05). However, significant differences in the average daily feed intake (ADFI) among different CNP concentrations were not observed. CNP also increased plasma immunoglobulin (Ig)A and IgG, and C3 and C4 concentrations in piglets in a dose-dependent manner on day 28, whereas IgM concentration was not affected by CNP. A total of 24 piglets in the control diet and control diet with 400 mg/kg CNP supplementation groups were randomly selected for the experiment of immunological stress. Half of the pigs in each group (n = 6) were injected i.p. with Escherichia coli lipopolysaccharide (LPS) at a concentration of 100 µg/kg. The other pigs in each group were injected with sterile saline solution at the same volume. Plasma concentrations of cortisol, prostaglandin E2 (PEG2), interleukin (IL)-6, tumour necrosis factor (TNF)-α and IL-1ß dramatically increased after LPS challenge. However, CNP inhibited the increase in cortisol, PEG2, IL-6 and IL-1ß levels in plasma, whereas TNF-α level slightly increased. Moreover, the effects of CNP on the gut microbiota were also evaluated. Our results showed that dietary supplementation with CNP modified the composition of colonic microbiota, where it increased the amounts of some presumably beneficial intestinal bacteria and suppressed the growth of potential bacterial pathogens. These findings suggested CNP supplementation improved the growth performance and immune status, alleviated immunological stress and regulated intestinal ecology in weaned piglets. Based on these beneficial effects, CNP could be applied as a functional feed additives supplemented in piglets diet.


Subject(s)
Chitosan/pharmacology , Gastrointestinal Microbiome/drug effects , Lipopolysaccharides/toxicity , Nanoparticles/chemistry , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Chitosan/chemistry , Diet/veterinary , Dietary Supplements , Hydrocortisone/blood , Immunity, Humoral , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/veterinary , Swine
6.
Biomed Pharmacother ; 120: 109479, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31557572

ABSTRACT

Iron deficiency is the most common nutritional deficiency disorder in early postnatal period, which often manifesting into clinical complications. Therefore, iron supplementation is necessary to avoid iron deficiency anemia in the neonatal period. However, how to supplement iron effectively is a big problem. Thus, using newborn piglets as a model for iron deficiency, we compared the effects of routinely used protocol by intramuscular injection of high amount of iron dextran and a modified strategy by split iron supplementation with reduced amounts of iron. The results showed that split iron supplementation efficiently improved hematological status of piglets and attenuated the induction of hepcidin expression, which resulted in the recovery of piglets from iron deficiency and the increase of iron utilization. Compared with piglets received large amounts of iron dextran, low dose supplementation of iron improved the growth performance and duodenum development by increasing the villus height and crypt depth and enhancing microvilli morphology. Furthermore, split iron supplementation minimized the potential toxicity of the administered iron due to the oxidative stress and hepatocyte autophagy. Overall, the present study demonstrated that split supplementation with reduced amount of iron dextran not only protected newborn piglets from iron deficiency but also eliminated potential toxicity. It suggested that besides combating anemia, possible negative effects of excessive iron on oxidative stress, which is especially important for infant development, should be considered.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Iron/administration & dosage , Anemia, Iron-Deficiency/metabolism , Animals , Animals, Newborn , Dietary Supplements , Disease Models, Animal , Duodenum/metabolism , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepcidins/metabolism , Intestinal Mucosa/metabolism , Oxidative Stress/drug effects , Swine
7.
Biol Trace Elem Res ; 190(2): 457-465, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30456562

ABSTRACT

Iron plays an important role both in bacterial pathogenicity and in host defense mechanisms, which has frequently been underestimated. The primary purpose of this study was to investigate the influence of iron supplementation on the progression of bacterial infection. We used mice as an experimental model to supplement iron after Escherichia coli (E. coli) O157:H7 infection and found that iron supplementation exacerbated clinical symptoms of bacterial infection by increasing mortality and reducing body weight. Iron supplementation promoted the colonization of bacteria and enhanced inflammatory responses by increasing C-reaction protein level and the phagocytic capacity of PBMCs, as well as upregulating the expression of TNF-α and IL-1ß in E. coli O157:H7-challenged mice. In vitro cell experiment confirmed that an excess of iron would enhance the growth of E. coli O157:H7 and worsen the outcome of bacterial infection. Therefore, it is certainly plausible that iron supplementation in bacterial infection may worsen rather than improve host outcome.


Subject(s)
Escherichia coli Infections/metabolism , Escherichia coli O157/metabolism , Iron/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Disease Models, Animal , Escherichia coli Infections/drug therapy , Escherichia coli O157/growth & development , Escherichia coli O157/isolation & purification , Iron/analysis , Male , Mice , Mice, Inbred C57BL , Streptomycin/administration & dosage , Streptomycin/therapeutic use , Vancomycin/administration & dosage , Vancomycin/therapeutic use
8.
Nutrients ; 10(6)2018 Jun 05.
Article in English | MEDLINE | ID: mdl-29874829

ABSTRACT

Early nutrition is key to promoting gut growth and education of the immune system. Although iron deficiency anemia has long been recognized as a serious iron disorder, the effects of iron supplementation on gut development are less clear. Therefore, using suckling piglets as the model for iron deficiency, we assessed the impacts of iron supplementation on hematological status, gut development, and immunity improvement. Piglets were parenterally supplied with iron dextran (FeDex, 60 mg Fe/kg) by intramuscular administration on the third day after birth and slaughtered at the age of two days, five days, 10 days, and 20 days. It was expected that iron supplementation with FeDex improved the iron status with higher levels of serum iron, ferritin, transferrin, and iron loading in the liver by regulating the interaction of hepcidin and ferroportin (FPN). FeDex supplementation increased villus length and crypt depth, attenuated the pathological status of the duodenum, and was beneficial to intestinal mucosa. FeDex also influenced the intestinal immune development by stimulating the cytokines' production of the intestine and enhancing the phagocytotic capacity of monocytes. Overall, the present study suggested that iron supplementation helped promote the development of the intestine by improving its morphology, which maintains its mucosal integrity and enhances the expression of immuno-associated factors.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Duodenum/drug effects , Intestinal Mucosa/drug effects , Iron-Dextran Complex/administration & dosage , Anemia, Iron-Deficiency/blood , Anemia, Iron-Deficiency/immunology , Anemia, Iron-Deficiency/physiopathology , Animal Nutritional Physiological Phenomena , Animals , Animals, Newborn , Cation Transport Proteins/metabolism , Cytokines/immunology , Dietary Supplements , Disease Models, Animal , Duodenum/growth & development , Duodenum/immunology , Duodenum/pathology , Ferritins/blood , Hepcidins/metabolism , Injections, Intramuscular , Intestinal Mucosa/growth & development , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Liver/drug effects , Liver/metabolism , Monocytes/drug effects , Monocytes/immunology , Nutritional Status , Phagocytosis/drug effects , Sus scrofa , Time Factors , Transferrin/metabolism
9.
Sci Rep ; 6: 27070, 2016 05 27.
Article in English | MEDLINE | ID: mdl-27230284

ABSTRACT

Butyrate has been used to treat different inflammatory disease with positive outcomes, the mechanisms by which butyrate exerts its anti-inflammatory effects remain largely undefined. Here we proposed a new mechanism that butyrate manipulate endogenous host defense peptides (HDPs) which contributes to the elimination of Escherichia coli O157:H7, and thus affects the alleviation of inflammation. An experiment in piglets treated with butyrate (0.2% of diets) 2 days before E. coli O157:H7 challenge was designed to investigate porcine HDP expression, inflammation and E. coli O157:H7 load in feces. The mechanisms underlying butyrate-induced HDP gene expression and the antibacterial activity and bacterial clearance of macrophage 3D4/2 cells in vitro were examined. Butyrate treatment (i) alleviated the clinical symptoms of E. coli O157:H7-induced hemolytic uremic syndrome (HUS) and the severity of intestinal inflammation; (ii) reduced the E. coli O157:H7 load in feces; (iii) significantly upregulated multiple, but not all, HDPs in vitro and in vivo via histone deacetylase (HDAC) inhibition; and (iv) enhanced the antibacterial activity and bacterial clearance of 3D4/2 cells. Our findings indicate that butyrate enhances disease resistance, promotes the clearance of E. coli O157:H7, and alleviates the clinical symptoms of HUS and inflammation, partially, by affecting HDP expression via HDAC inhibition.


Subject(s)
Butyric Acid/pharmacology , Defensins/genetics , Escherichia coli Infections/immunology , Escherichia coli O157/immunology , Hemolytic-Uremic Syndrome/immunology , Histone Deacetylase Inhibitors/pharmacology , Animals , Butyric Acid/therapeutic use , Cell Line , Colitis/blood , Colitis/drug therapy , Colitis/immunology , Colitis/microbiology , Colon/immunology , Colon/metabolism , Colon/pathology , Cytokines/blood , Defensins/metabolism , Drug Evaluation, Preclinical , Escherichia coli Infections/blood , Escherichia coli Infections/drug therapy , Escherichia coli Infections/microbiology , Feces/microbiology , Gene Expression , Hemolytic-Uremic Syndrome/blood , Hemolytic-Uremic Syndrome/drug therapy , Hemolytic-Uremic Syndrome/microbiology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/metabolism , Sus scrofa , Transcriptional Activation , Up-Regulation/drug effects
10.
Biol Trace Elem Res ; 167(1): 28-35, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25774043

ABSTRACT

Iron deficiency is common throughout the world and has been linked to immunity impairments. Using piglets to model human infants, we assessed the impact of systemic iron homeostasis on proinflammatory status. Artificially reared piglets were parenterally supplied with iron dextran by intramuscular administration at the age of 3 days. Relative to no iron supplementation (control), iron dextran-treated (FeDex) piglets increased hematological parameters as well as iron levels in serum and tissues from days 21 to 49. High expression of hepcidin was observed in FeDex-treated piglets, which correlated with suppressed expression of ferroportin in duodenum. Lower levels of proinflammatory cytokine (IL-6, TNF-α, IFN-γ, and IL-1ß) transcripts were detected in ileum of FeDex-treated piglets, which indicated that iron supplementation could attenuate the increase of inflammatory cytokines caused by iron deficiency. Histopathological analysis of liver and duodenum proved the less inflammatory responses after iron supplementation. Hepcidin was highly stimulated by FeDex supplementation and attenuated the inflammation of anemia, which implied that hepcidin might had antiinflammatory function and is a candidate regulator of the cross-talk between iron regulation and inflammation.


Subject(s)
Anemia, Iron-Deficiency/prevention & control , Hepcidins/metabolism , Inflammation/prevention & control , Iron-Dextran Complex/pharmacology , Administration, Intranasal , Anemia, Iron-Deficiency/genetics , Anemia, Iron-Deficiency/metabolism , Animals , Animals, Newborn , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Duodenum/drug effects , Duodenum/metabolism , Gene Expression Regulation/drug effects , Hematinics/administration & dosage , Hematinics/pharmacology , Hepcidins/genetics , Humans , Ileum/drug effects , Ileum/metabolism , Immunohistochemistry , Infant , Inflammation/genetics , Inflammation/metabolism , Inflammation Mediators/metabolism , Iron-Dextran Complex/administration & dosage , Liver/drug effects , Liver/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Swine , Time Factors
11.
Biol Trace Elem Res ; 136(1): 71-8, 2010 Jul.
Article in English | MEDLINE | ID: mdl-19789846

ABSTRACT

In order to investigate the effects of montmorillonite (MMT) on reducing dietary lead (Pb) toxicity to tilapia (Oreochromis niloticus), 240 fish were randomly divided into four treatments denominated as follows: control treatment (fed with a basal diet), MMT treatment (fed with a basal diet added with 0.5% MMT), Pb treatment (fed with a basal diet added with 100 mg Pb per kilogram dry weight (dw)), and Pb + MMT treatment (fed with a basal diet added with 100 mg Pb per kilogram dw and 0.5% MMT). Changes in Pb accumulation, oxidative stress, and DNA damage in tilapia were measured after 60 days. DNA damage was assessed using comet assay. The results showed that MMT supplemented in diet significantly reduced Pb accumulation in kidney and blood of tilapia exposed to dietary Pb (P < 0.05). Malondialdehyde level decreased insignificantly while levels of total antioxidant capacity and glutathione (GSH), activities of glutathione peroxidase, and superoxide dismutase increased insignificantly in kidney of tilapia in Pb + MMT treatment as compared to Pb treatment (P > 0.05). Significant decreases in tail length, tail DNA, tail moment, and Olive tail moment of peripheral blood cells in Pb + MMT treatment were observed when compared with Pb treatment (P < 0.05). The results indicated that dietary MMT supplementation could alleviate dietary Pb toxicity to tilapia effectively.


Subject(s)
Bentonite/pharmacology , DNA Damage/drug effects , Lead/toxicity , Oxidative Stress/drug effects , Animals , Antioxidants/pharmacology , Dietary Supplements , Kidney/metabolism , Lead/blood , Lead/metabolism , Tilapia
SELECTION OF CITATIONS
SEARCH DETAIL