Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Anim Nutr ; 15: 386-398, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38058564

ABSTRACT

Piglets are particularly susceptible to oxidative stress, which causes inferior growth performance and intestinal damage. Squalene (SQ), a natural bioactive substance enriched in shark liver oil, shows excellent antioxidant properties and can currently be obtained at a low cost from deodorizer distillate during the production of plant oil. This study aimed to evaluate the effects of plant-derived SQ supplementation on the growth performance of piglets and explore the beneficial roles of SQ against oxidative stress and intestinal injury in diquat-challenged piglets. Forty piglets were randomly divided into five groups and fed a basal diet supplemented with SQ at 0, 500, 1000, or 2000 mg/kg for 5 weeks. Acute oxidative stress was induced in the piglets with diquat (10 mg/kg BW) at the fourth week of the experiment, followed by a 7-d recovery period. Results showed that before the diquat challenge, SQ supplementation significantly improved growth performance (average daily gain and feed conversion ratio) and serum antioxidant status, and after the diquat challenge, SQ supplementation significantly mitigated diquat-induced growth arrest, intestinal villous atrophy, intestinal epithelial cell apoptosis, intestinal hyperpermeability, and deficiency of intestinal epithelial tight junction proteins (zonula occludens-1, occludin, and claudin-3). Under oxidative stress induced by diquat, SQ supplementation consistently improved the antioxidant status of the small intestine, liver, and muscle. In vitro, SQ was shown to alleviate hydrogen peroxide (H2O2)-induced increase of the levels of intracellular reactive oxygen species and apoptosis of porcine intestinal epithelial cells. Taken together, SQ supplementation improves growth performance and effectively alleviates acute oxidative stress-induced growth retardation and intestinal injury via improving antioxidant capacity in piglets. Our findings may provide an efficient strategy for alleviating oxidative stress-induced inferior growth performance and intestinal damage in piglets.

2.
Stem Cell Reports ; 18(7): 1451-1467, 2023 07 11.
Article in English | MEDLINE | ID: mdl-37327782

ABSTRACT

Early weaning usually causes small intestine epithelial development abnormality, increasing the risk of gastrointestinal diseases. Glutamine (Gln), enriching in plasma and milk, is widely reported to benefit intestinal health. However, whether Gln affects intestinal stem cell (ISC) activity in response to early weaning is unclear. Here, both the early weaning mice and intestinal organoids were used to study the role of Gln in regulating ISC activities. Results showed that Gln ameliorated early weaning-induced epithelial atrophy and augmented the ISC-mediated epithelial regeneration. Gln deprivation disabled ISC-mediated epithelial regeneration and crypt fission in vitro. Mechanistically, Gln augmented WNT signaling in a dose-dependent manner to regulate ISC activity, while WNT signaling blockage abolished the effects of Gln on ISCs. Together, Gln accelerates stem cell-mediated intestinal epithelial development associated with the augmentation of WNT signaling, which provides novel insights into the mechanism by which Gln promotes intestinal health.


Subject(s)
Glutamine , Wnt Signaling Pathway , Mice , Animals , Weaning , Intestine, Small , Stem Cells , Intestinal Mucosa/physiology , Cell Proliferation
3.
Nutrients ; 15(11)2023 May 26.
Article in English | MEDLINE | ID: mdl-37299439

ABSTRACT

Ornithine α-ketoglutarate (OKG), a nutritional compound, is an amino acid salt with anti-oxidative and anti-inflammatory effects on humans and animals. Ulcerative colitis (UC), as an inflammatory bowel disease (IBD), leads to chronic intestinal inflammatory dysfunction. This study evaluated the optimal dosage of OKG in healthy mice. Then, a mouse model of acute colitis was established using dextran sodium sulfate (DSS), and the preventive effect of OKG on DSS-induced colitis in mice was explored through analysis of serum inflammatory cytokines and fecal microbiota. Initially, the mice were randomly divided into a control group, a group given a low dose of OKG (LOKG: 0.5%), a group given a medium dose of OKG (MOKG: 1%), and a group given a high dose of OKG (HOKG: 1.5%); they remained in these groups for the entire 14-day experimental period. Our results demonstrated that 1% OKG supplementation increased body weight, serum growth hormone (GH), insulin (INS), alkaline phosphatase (ALP), Tyr, and His and decreased urea nitrogen (BUN), NH3L, and Ile. Then, a 2 × 2 factor design was used for a total of 40 mice, with diet (a standard diet or a 1% OKG diet) and challenge (4% DSS or not) as the main factors. During days 14 to 21, the DSS mice were administered 4% DSS to induce colitis. The results revealed that OKG alleviated weight loss and reversed the increases in colonic histological damage induced by DSS. OKG also increased serum IL-10 secretion. Moreover, OKG enhanced the abundance of Firmicutes and decreased that of Bacteriodetes at the phylum level and particularly enhanced the abundance of Alistipes and reduced that of Parabacterioides at the genus level. Our results indicated that OKG promotes growth performance and hormone secretion and regulates serum biochemical indicators and amino acid concentrations. Furthermore, 1% OKG supplementation prevents DSS-induced colitis in mice via altering microbial compositions and reducing the secretion of inflammatory cytokines in serum.


Subject(s)
Colitis, Ulcerative , Colitis , Gastrointestinal Microbiome , Humans , Mice , Animals , Cytokines/metabolism , Dextran Sulfate/adverse effects , Colitis/chemically induced , Colitis/drug therapy , Colitis/pathology , Inflammation/pathology , Colitis, Ulcerative/pathology , Colon/metabolism , Amino Acids , Mice, Inbred C57BL , Disease Models, Animal
4.
Stem Cell Res Ther ; 14(1): 65, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37020258

ABSTRACT

BACKGROUND: Early weaning and shorter breastfeeding duration are applied by a proportion of young mothers, especially in the social spheres of poverty-stricken areas. Early childhood is a critical period for intestinal development, which is driven by intestinal stem cells (ISCs). However, how early weaning practice affects the function of ISCs to mediate intestinal development remains unclear. METHODS: We established an excellent early weaning mice model that has significant intestinal atrophy and growth arrest symptoms to explore the responses of ISCs to early weaning. The primary and passaged intestinal organoids from the suckling or early weaning mice were cultured to explore the underlying mechanism of early weaning affecting the ISCs. RESULTS: Early weaning depressed the self-renewal of ISCs and attenuated the activity of ISCs-driven intestinal epithelial regeneration and crypt expansion in vivo and ex-vivo. Further results showed that early weaning retarded the differentiation of ISCs into transit-amplifying cells and Paneth cells, and accelerated the apoptosis of villous epithelial cells, jointly leading to intestinal epithelial atrophy. Mechanistically, early weaning inhibited Wnt signaling in ISCs, while an exogenous Wnt amplifier restored ISCs' function in ex-vivo. CONCLUSION: Our findings indicate that early weaning depresses the activity of ISCs via attenuating Wnt/ß-catenin signaling and triggers the proinflammatory cytokines TNF-α, IL-1ß, IL-6, and IL-17 in jejunum, thereby impeding ISCs-driven epithelial regeneration and intestinal growth, which may provide a basal theory for the development of infant nutrients targeting stem cells to alleviate early weaning-induced intestinal problems.


Subject(s)
Wnt Signaling Pathway , beta Catenin , Child, Preschool , Mice , Humans , Animals , beta Catenin/metabolism , Intestinal Mucosa/metabolism , Weaning , Cell Proliferation , Stem Cells/metabolism , Paneth Cells/metabolism
5.
Animals (Basel) ; 13(4)2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36830356

ABSTRACT

Phosphorus (P) pollution from modern swine production is a major environmental problem. Dietary interventions to promote bone growth can improve the utilization of dietary P, and thereby reduce its emission. Recent in vitro studies have shown that alpha-ketoglutarate (AKG) exerts a pro-osteogenic effect on osteoblast cells. This study aimed to evaluate the effects of AKG supplementation on bone growth, P and Ca digestion, and the gut microbial profile in piglets. Thirty-two piglets were randomly assigned into two dietary groups. The piglets were fed a basic diet containing 10 g/kg AKG or 10 g/kg maize starch (control) for 28 days. On days 21-28, titanium dioxide was used as an indicator to determine the apparent digestibility of P. AKG supplementation improved the bone mineral density, length, weight, and geometrical and strength properties of the femur and tibia. Furthermore, AKG supplementation increased apparent ileal and total tract digestibility of P. Colonic microbiota analysis results showed that AKG supplementation increased α-diversity and beneficial bacteria, including Lactobacillus and Clostridium butyricum, and decreased nitrogen fixation and chemoheterotrophy. Together, AKG supplementation improves bone growth, the utilization of dietary P, and the colonic microbial profile, which may provide a nutritional strategy for diminishing P pollution originating from the pig industry.

6.
Nutrients ; 15(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36771407

ABSTRACT

Age-related osteoporosis, a high-prevalence disease in the aged population, is generally attributed to the excessive activity of osteoclasts. Most approved drugs treat osteoporosis by inhibition of osteoclasts. Although in vivo studies have shown that alpha-ketoglutarate (AKG), an intermediate in the TCA cycle, can ameliorate age-related osteoporosis, the effects of AKG on osteoclastogenesis and the underlying mechanism of its action have not been studied yet. Here, we showed that the elevation of intracellular AKG levels by supplementing dimethyl AKG (DM-AKG, a cell-permeable derivative of AKG) inhibits the receptor activator of NF-κB ligand (RANKL)-induced osteoclasts differentiation from primary bone marrow-derived macrophages (BMMs) and RAW264.7 cells in vitro. We further found that DM-AKG treatment suppresses NF-κB signaling and oxidative phosphorylation (OXPHOS) during RANKL-induced osteoclastogenesis in RAW264.7 cells. Interestingly, dimethyl oxalylglycine (DMOG), an AKG competitive inhibitor of AKG-dependent prolyl hydroxylases (PHDs), antagonizes the suppression of the RANKL-activated NF-κB signaling pathway caused by DM-AKG treatment. Furthermore, blocked PHD1 expression (also known as EglN2), instead of PHD2 or PHD3, was confirmed to reverse the DM-AKG treatment-induced suppression of the RANKL-activated NF-κB signaling pathway. Accordingly, blocked PHD1 expression antagonized the inhibitory effects of DM-AKG on osteoclastogenesis. Together, our finding suggests that the elevation of intracellular AKG levels inhibits osteoclastogenesis by suppressing RANKL-activated NF-κB signaling in a PHD1-dependent manner, which may provide a novel nutritional strategy for osteoporosis treatment.


Subject(s)
Bone Resorption , Osteoporosis , Humans , Aged , NF-kappa B/metabolism , Osteogenesis , Ketoglutaric Acids/pharmacology , Ketoglutaric Acids/metabolism , Signal Transduction , Osteoclasts , Cell Differentiation , Osteoporosis/metabolism , RANK Ligand/pharmacology , RANK Ligand/metabolism , Bone Resorption/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases/pharmacology
7.
Front Nutr ; 9: 862498, 2022.
Article in English | MEDLINE | ID: mdl-35747266

ABSTRACT

Enterotoxigenic Escherichia coli (ETEC) is one of the main causes of diarrhea in weaned piglets, and ornithine α-ketoglutarate (OKG) as a food supplement has been shown to improve intestinal immune status in animals and humans. However, it remains unknown whether OKG alleviates inflammation through the regulation of gut microbiota and its metabolites on ETEC-infected piglets. This study was conducted to explore the impact of OKG on growth performance, immunity, and ileal mucosa microbiota and its metabolites in piglets infected with ETEC. On a total of 40 pigs, a 2 × 2 factor design was performed; the major factors were diet (basal diet or 1% OKG diet) and challenge (E. coli or LB Broth). The results showed that ETEC-infection inhibited growth performance, and OKG supplementation alleviated growth performance. Interestingly, ETEC-infection increased the serum TNF-α and IL-6, decreased the serum IL-10, downregulated the mRNA expression of IL-1ß, IL-6, MyD88, and improved the mRNA expression of IL-8, IL-18, and TLR4. OKG inhibited serum IL-6, suppressed the phosphorylation of downstream signals of NF-κB/JNK in the ileum, and enhanced serum IL-10 and ileum SIgA in ETEC-challenged piglets. OKG supplementation enhanced the mRNA expression of IL-1ß and IL-10 and reduced NF-κB and MyD88 in the ileum. Importantly, OKG reversed intestinal microbiota dysfunction, including the diversity of ileal microbiota, the relative abundances of Actinobacillus, Turicibacter, and [Acetivibrio]_ethanolgignens_group, which significantly affected arachidonic acid metabolism and primary bile acid biosynthesis. Collectively, our results suggest that OKG improves growth performance, regulates immunity, and ileal mucosa microbiota and its metabolites in ETEC-infected piglets.

8.
Arch Anim Nutr ; 74(1): 39-56, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31552757

ABSTRACT

The aim of the study was to investigate the effects of dietary alpha-ketoglutarate (AKG) on the faecal bacteria composition of suckling piglets after supplementation of AKG to the diet of lactating sows. After farrowing, the sows were assigned to either a normal lactation diet (control group, n = 12) or a diet supplemented with 0.25% AKG (AKG group, n = 12) based on body weight (BW) and parity. During the 21-d suckling period, BW and diarrhoea occurrences of piglets were recorded daily, while faeces were sampled weekly from sows and piglets. The levels of pH, ammonia, short-chain fatty acids (SCFA) and lactate in the faeces of piglets were determined. In particular, bacteria profiles in faeces of sows and their suckling piglets were examined by Illumina sequencing. The results showed that the AKG diet altered the faecal bacteria composition in sows during the 21-d lactation period, leading to increases (p < 0.05) in the abundances of genera Prevotella, Lactobacillus, Bacteroides and Methanobrevibacter, but decreases (p < 0.05) in the abundances of genera Oscillospira and Dorea. AKG supplement to the sows during lactation indirectly enhanced (p < 0.05) bacterial richness and SCFA levels (especially, acetate) in the faeces of piglets during the 21-d suckling period. It is suggested that maternal AKG supplementation alters the composition of faecal bacteria in the sows, and increases the faecal bacteria richness and acetate levels in the piglets, which might be associated with an enhanced growth performance of piglets.


Subject(s)
Feces/microbiology , Ketoglutaric Acids/metabolism , Sus scrofa/microbiology , Animal Feed/analysis , Animals , Animals, Newborn , Diet/veterinary , Dietary Supplements/analysis , Dose-Response Relationship, Drug , Female , Ketoglutaric Acids/administration & dosage , Lactation , Random Allocation , Sus scrofa/metabolism
9.
Food Funct ; 11(1): 472-482, 2020 Jan 29.
Article in English | MEDLINE | ID: mdl-31833510

ABSTRACT

The aim of this study was to evaluate the protective effects and underlying mechanisms of ornithine α-ketoglutarate (OKG) on d-galactose (d-gal)-induced chronic oxidative stress in a pig model. A total of 40 castrated young pigs were randomly separated into five groups, including a control group, a model group treated with 5 mg per kg body weight (BW) d-gal, and three d-gal + OKG groups in which the pigs received 0.5%, 1%, and 2% OKG (n = 8). The experiment lasted for 28 days. The growth performance, serum oxidative stress index, expression of relative intestinal genes, gut microbiota, and serum amino acid pool were determined. The results demonstrated that administration of d-gal significantly affected growth performance and superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) levels including related mRNA expression suppression, malondialdehyde (MDA) levels enhancement, gut microbiota dysfunction, and serum amino acid alteration in pigs. However, treatment with 0.5% OKG markedly ameliorated the reduction in the growth performance, as evidenced by the reversed final body weight, average feed intake, and average body weight. Also, 0.5% OKG enhanced the SOD and GSH-Px levels including relative mRNA expression in the intestine and inhibited lipid oxidation subsequent to MDA generation. The intestinal abundances of Firmicutes were increased and those of Proteobacteria, Fusobacteria, Bacteriodetes, and Euryarchaeota were decreased in the pigs supplemented with 0.5% OKG. Meanwhile, 0.5% OKG increased the glutamate, proline, aspartate, threonine, valine, isoleucine and leucine levels in the serum. Collectively, these results indicate that d-gal induced chronic oxidative stress and also proved the positive effects of 0.5% OKG on altering the pig gut microbe, restoring serum amino acid and alleviating the growth-suppression induced by d-gal chronic oxidative stress.


Subject(s)
Gastrointestinal Microbiome , Ornithine/analogs & derivatives , Oxidative Stress/drug effects , Amino Acids/blood , Animals , Galactose , Glutathione Peroxidase/metabolism , Malondialdehyde/metabolism , Ornithine/pharmacology , Superoxide Dismutase/metabolism , Swine/growth & development
10.
Arch Anim Nutr ; 73(6): 457-471, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31454268

ABSTRACT

The aim of the study was to investigate if dietary alpha-ketoglutarate (AKG) supplementation may improve the performance of lactating sows and their suckling piglets. After farrowing, 24 lactating sows (Large White × Landrace) with similar body weight (BW) were assigned to the control and AKG groups based on parity, and their lactation diets were supplemented with 0.00 or 0.25% AKG, respectively. It was found that supplementing the diet of lactating sows with 0.25% AKG enhanced growth performance of the suckling piglets from d 7 to d 21 of the lactation period, improved villus height of ileum and tended (p = 0.085) to increase mean volumetric bone mineral density of femur in the weanling piglets. In the lactating sows, dietary supplementation of AKG decreased plasma urea level on d 14 of lactation, decreased plasma calcium (Ca) concentrations from d 7 to d 21 of lactation and increased lactose and Ca levels in ordinary milk. Thus, it was proposed that AKG supplementation stimulates the capacity for lactose synthesis and Ca uptake in the mammary gland, thereby altering the composition of the ordinary milk which might be associated with the enhanced performance of piglets during the suckling period. These findings could lead to a better application of AKG in lactating nutrition, and therefore, promoting pork production.


Subject(s)
Amino Acids/metabolism , Animals, Suckling/growth & development , Ketoglutaric Acids/metabolism , Lactation/drug effects , Protein Biosynthesis , Sus scrofa/physiology , Amino Acids/drug effects , Animal Feed/analysis , Animals , Animals, Suckling/metabolism , Diet/veterinary , Dietary Supplements/analysis , Female , Ketoglutaric Acids/administration & dosage , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/physiology , Milk/chemistry , Nutritive Value/drug effects , Protein Biosynthesis/drug effects , Sus scrofa/growth & development
11.
J Agric Food Chem ; 67(21): 6019-6031, 2019 May 29.
Article in English | MEDLINE | ID: mdl-31067048

ABSTRACT

Intestinal endoplasmic reticulum stress (ERS) triggered by adverse factors disturbs the normal function of the intestine. Allicin has been reported to promote intestinal health and development. In the present study, we established in vivo (35-day-old weaned piglets, 4-week-old mice) and in vitro (IPEC-J2 cell line) ERS models to explore the possible mechanisms by which allicin may benefit intestinal health. This study revealed the following: (1) allicin supplementation improved intestinal morphological indices and ameliorated mild ERS in the jejunum of the weaned piglets; (2) allicin supplementation decreased cellular reactive oxygen species and upregulated the XBP-1s signaling pathways in IPEC-J2 cells; (3) allicin supplementation reduced the prolonged ERS-mediated apoptosis of IPEC-J2 cells and in the jejunal tissues of the KM mice; (4) allicin supplementation enhanced the intercellular junction protein levels of jejunal cells by alleviating the prolonged ERS. These novel findings suggest that eating garlic could alleviate some intestinal malfunctions associated with ERS.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Jejunum/drug effects , Plant Extracts/pharmacology , Sulfinic Acids/pharmacology , Unfolded Protein Response/drug effects , Animals , Apoptosis/drug effects , Cell Line , Disulfides , Female , Garlic/chemistry , Jejunum/metabolism , Jejunum/physiology , Male , Mice , Signal Transduction/drug effects , Swine
12.
Oncotarget ; 8(54): 91965-91978, 2017 Nov 03.
Article in English | MEDLINE | ID: mdl-29190890

ABSTRACT

Alpha-ketoglutarate (AKG) is an important cellular metabolite that participates in energy production and amino acid metabolism. However, the protective effects and mechanism of AKG on mucosal lesions have not been well understood. This study was conducted to investigate the effects of dietary AKG supplementation on epithelial restitution in early-weaning piglets under Escherichia coli lipopolysaccharide (LPS) induction. A total of 32 weaned piglets were used in a 2 × 2 factorial design; the major factors were dietary treatment (basal diet or AKG diet) and inflammatory challenge (LPS or saline). The results showed that AKG supplementation improved the growth performance and intestinal morphology in the LPS-induced early-weaning piglets. Compared with the basal diet, the AKG diet remarkably decreased the concentration and mRNA expression of intestinal inflammatory cytokines (IL-1ß, IL-6, and IL-12) in the LPS-induced piglets. Moreover, AKG administration upregulated the mRNA expression of nutrient-sensing transporters (GLUT-2, SGLT-1, PEPT-1, I-FABP2) in the small intestine of both saline- and LPS-treated piglets, and improved the distribution and expression of tight-junction genes andproteins (ZO-1, Occludin, Claudins, E-cadherin). Collectively, our findings indicate that AKG has the potential to alleviate intestinal inflammatory response and improve epithelial restitution and nutrient-sensing ability under stress injury in early-weaning piglets, and it also provides an experimental basis for enteral use of AKG in swine production and clinical application to prevent intestinal epithelial damage.

13.
Oncotarget ; 8(61): 102974-102988, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29262538

ABSTRACT

Alpha-ketoglutarate (AKG) is a critical nutritional factor in the maintenance of intestinal homeostasis. However, the relative mechanism of AKG has not been well understood. It was recently shown that the interaction between nuclear factor kappa B (NF-κB)-mediated inflammatory pathway and pregnane X receptor (PXR)-regulated detoxification pathway is a check and balance mechanism for keeping the homeostatic state of the intestine, preventing the onset of intestinal inflammation which may lead to cancer. In the current study we used lipopolysaccharide (LPS)-challenged piglet and intestinal porcine epithelial cells-J2 models to investigate the effects of dietary AKG supplementation on the intestinal immune system and PXR regulated target expression. We found that LPS induced significant activation of the NF-κB-mediated inflammatory pathway with concomitant impairment of intestinal nutrient absorption. AKG administration increased intracellular AKG and its metabolite concentrations and enhanced the mRNA expression of alpha-ketoglutarate dehydrogenase in vivo and in vitro. Thus dietary AKG supplementation reversed the adverse effects induced by LPS. We also found a strong inhibitory effects on the NF-κB-mediated inflammatory pathway, especially, in the AKG-treated intestinal tissues, LPS-induced NF-κB phosphorylation was inhibited and TNF-α was suppressed. Interestingly, AKG has potent effects in regulating the PXR and its downstream targets such as CYP3As and CYP2Bs in vivo and in vitro, although AKG is not a known PXR ligand. One potential mechanism for the up-regulation of the PXR pathway is through the down-regulation of NF-κB pathway which in turn de-represses the PXR-regulated target expression. Taken together, our results suggest that AKG improves intestinal immune system through modulating the interaction between PXR and NF-κB. Our findings have important implications for the prevention and treatment of intestinal inflammatory diseases in neonates.

14.
J Agric Food Chem ; 65(11): 2287-2298, 2017 Mar 22.
Article in English | MEDLINE | ID: mdl-28241728

ABSTRACT

Water and ion absorption via sensitive aquaporins (AQPs) and ion channels is of critical importance in intestinal health. However, whether α-ketoglutarate (AKG) could improve intestinal water and ion homeostasis in lipopolysaccharide (LPS)-challenged piglets and whether the AMP-activated protein kinase (AMPK) pathway is involved remains largely unknown. This study was conducted to investigate the effect of dietary AKG supplementation on the small intestinal water and ion homeostasis through modulating the AMPK pathway in a piglet diarrhea model. A total of 32 weaned piglets were used in a 2 × 2 factorial design; the major factors were diet (basal diet or 1% AKG diet) and challenge (Escherichia coli LPS or saline). The results showed that LPS challenge increased the diarrhea index and affected the concentrations of serum Na+, K+, Cl-, glucose, and AKG and its metabolites in piglets fed the basal or AKG diet. However, the addition of AKG attenuated diarrhea incidence and reversed these serum parameter concentrations. Most AQPs (e.g., AQP1, AQP3, AQP4, AQP5, AQP8, AQP10, and AQP11) and ion transporters (NHE3, ENaC, and DRA/PAT1) were widely distributed in the duodenum and jejunum of piglets. We also found that AKG up-regulated the expression of intestinal epithelial AQPs while inhibiting the expression of ion transporters. LPS challenge decreased (P < 0.05) the gene and protein expression of the AMPK pathway (AMPKα1, AMPKα2, SIRT1, PGC-1α, ACC, and TORC2) in the jejunum and ileum. Notably, AKG supplementation enhanced the abundance of these proteins in the LPS-challenged piglets. Collectively, AKG plays an important role in increasing water and ion homeostasis through modulating the AMPK pathway. Our novel finding has important implications for the prevention and treatment of gut dysfunction in neonates.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diarrhea/veterinary , Intestinal Mucosa/metabolism , Ketoglutaric Acids/metabolism , Swine Diseases/metabolism , Swine/metabolism , Water/metabolism , Animals , Biological Transport , Diarrhea/enzymology , Diarrhea/metabolism , Homeostasis , Intestines/enzymology , Ions/metabolism , Swine Diseases/enzymology
15.
Curr Protein Pept Sci ; 18(6): 562-570, 2017.
Article in English | MEDLINE | ID: mdl-27356941

ABSTRACT

AMP-activated protein kianse (AMPK) is a master sensor of cellular energy levels and a crucial regulator of nutrient metabolism such as the synthesis of fatty acids, glucose and protein as well as their oxidation to CO2 and water . Thus, AMPK signaling has important implications for fat deposition and glucose homeostasis in animals and humans. Much experimental and clinical evidence show that AMPK is a key therapeutic target for the prevention of diseases such as obesity, diabetes, cancer, inflammation and cardiac dysfunction. In this review we highlight recent advances on the upstream and downstream targets of AMPK, as well as the specific mechanisms whereby AMPK regulates digestive functions and chronic energy balance in animals and humans.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Glucose/metabolism , Lipid Metabolism , Proteins/metabolism , Signal Transduction , Animals , Digestion , Energy Metabolism , Humans
16.
J Agric Food Chem ; 64(13): 2668-73, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27018713

ABSTRACT

Alpha-ketoglutarate (AKG) plays a vital part in the tricarboxylic acid cycle and is a key intermediate in the oxidation of L-glutamine (Gln). The study was to evaluate effects of AKG on Gln metabolism in vivo and in vitro. A total of twenty-one piglets were weaned at 28 days with a mean body weight (BW) of 6.0 ± 0.2 kg, and randomly divided into 3 groups: corn soybean meal based diet (CON group); the basal diet with 1% alpha-ketoglutarate (AKG treatment group); and the basal diet with 1% L-glutamine (GLN treatment group). Intestinal porcine epithelial cells-1 (IPEC-1) were incubated to investigate effects of 0.5, 2, and 3 mM AKG addition on Gln metabolism. Our results showed that there were no differences (P > 0.05) among the 3 treatments in initial BW, final BW, and average daily feed intake. However, average daily gain (P = 0.013) and gain:feed (P = 0.041) of the AKG group were greater than those of the other two groups. In comparison with the CON group, the AKG and GLN groups exhibited an improvement in villus length, mucosal thickness, and crypt depth in the jejunum of piglets. Serum concentrations of Asp, Glu, Val, Ile, Tyr, Phe, Lys, and Arg in the piglets fed the 1% AKG or Gln diet were lower than those in the CON group. Compared with the CON group, the mRNA expression of jejunal and ileal amino acid (AA) transporters in the AKG and GLN groups were significantly increased (P < 0.05). Additionally, the in vitro study showed that the addition of 0.5, 2, and 3 mM AKG dose-dependently decreased (P < 0.05) the net utilization of Gln and formulation of ammonia, Glu, Ala, and Asp by IPEC-1. In conclusion, dietary AKG supplementation, as a replacement for Gln, could improve Gln metabolism in piglet enterocytes and enhance the utilization of AA.


Subject(s)
Enterocytes/metabolism , Glutamine/metabolism , Ketoglutaric Acids/metabolism , Amino Acids/metabolism , Animals , Arginine/blood , Aspartic Acid/blood , Body Weight , Citric Acid Cycle/physiology , Diet/veterinary , Dietary Supplements , Glutamine/blood , Ileum/metabolism , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Isoleucine/blood , Jejunum/metabolism , Lysine/blood , Phenylalanine/blood , RNA, Messenger/metabolism , Swine , Tyrosine/blood , Valine/blood , Weaning
SELECTION OF CITATIONS
SEARCH DETAIL
...