Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Sci Food Agric ; 103(11): 5569-5577, 2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37071083

RESUMO

BACKGROUND: Overuse of antibiotics contributes to bacterial resistance in animals. Therefore, it is necessary to find a new way to ensure animal health and promote animal growth. This experiment was conducted to investigate the effect of mannan oligosaccharide (MOS)/vitamin E (VE)/attapulgite (APT) nanocomposites (SLK1, SLK3, SLK5) on growth performance and intestinal health in weaned piglets. Each 1 kg of SLK1, SLK3 or SLK5 contains 50 g of vitamin E, and each had a different MOS concentration [SLK1 (50 g kg -1 MOS), SLK3 (100 g kg -1 MOS), SLK5 (150 g kg -1 MOS)]. In total, 135 piglets were randomly divided into five groups (normal control group, traditional antibiotic substitutes group, SLK1 group, SLK3 group and SLK5 group), and growth performance, diarrhea index, intestinal epithelial barrier function and intestinal microbial composition were measured. RESULTS: SLK1 and SLK5 significantly decreased diarrhea frequency in weaned piglets (p < 0.05). Furthermore, SLK5 significantly increased survival rate of weaned piglets compared to the traditional antibiotic substitutes group (p < 0.05). SLK5 also increased villus height of ileum, and increased goblet number of the jejunum (p < 0.05). 16S rRNA sequencing showed that SLK5 significantly regulated intestinal colonic microbiota composition (p < 0.05). Specifically, SLK5 significantly increased the abundance of Phascolarctobacterium succinatutens in the cecum and increased the abundance of Lactobacillus and Bifidobacterium in the colon (p < 0.05). In addition, dietary supplementation with 1 kg T-1 SLK5 also significantly increased the propionate content in the colon, which is significantly correlated with Phascolarctobacterium (p < 0.05). CONCLUSION: Dietary supplementation with 1 kg T-1 SLK5 improved intestinal epithelial barrier function, and regulated intestinal microbiota composition to prevent diarrhea in weaned piglets. © 2023 Society of Chemical Industry.


Assuntos
Microbioma Gastrointestinal , Nanocompostos , Animais , Antibacterianos/farmacologia , Diarreia/prevenção & controle , Diarreia/veterinária , Suplementos Nutricionais/análise , Mananas , Oligossacarídeos/farmacologia , RNA Ribossômico 16S , Suínos , Vitamina E/farmacologia , Desmame
2.
Crit Rev Food Sci Nutr ; 62(18): 4867-4892, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33523720

RESUMO

Growth retardation (GR), which commonly occurs in childhood, is a major health concern globally. However, the specific mechanism remains unclear. It has been increasingly recognized that changes in the gut microbiota may lead to GR through affecting the microbiota-gut-brain axis. Microbiota interacts with multiple factors such as birth to affect the growth of individuals. Microbiota communicates with the nerve system through chemical signaling (direct entry into the circulation system or stimulation of enteroendocrine cells) and nervous signaling (interaction with enteric nerve system and vagus nerve), which modulates appetite and immune response. Besides, they may also influence the function of enteric glial cells or lymphocytes and levels of systemic inflammatory cytokines. Environmental stress may cause leaky gut through perturbing the hypothalamic-pituitary-adrenal axis to further result in GR. Nutritional therapies involving probiotics and pre-/postbiotics are being investigated for helping the patients to overcome GR. In this review, we summarize the role of microbiota in GR with human and animal models. Then, existing and potential regulatory mechanisms are reviewed, especially the effect of microbiota-gut-brain axis. Finally, we propose nutritional therapeutic strategies for GR by the intervention of microbiota-gut-brain axis, which may provide novel perspectives for the treatment of GR in humans and animals.


Assuntos
Sistema Hipotálamo-Hipofisário , Microbiota , Animais , Encéfalo/fisiologia , Eixo Encéfalo-Intestino , Transtornos do Crescimento , Sistema Hipófise-Suprarrenal
3.
Int Immunopharmacol ; 117: 109742, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36822096

RESUMO

Attapulgite is a kind of natural clay mineral. Its unique pore structure makes it an ideal adsorption material and carrier material. However, the beneficial effect of modified attapulgites (SLK) in livestock is still unknown. The study was aimed to investigate the beneficial effect of modified attapulgites on diarrhea. 135 piglets were randomly divided into 5 groups and fed with control diet, traditional antibiotic substitute (TAS) supplementation diet, 0.5 mg/kg SLK supplementation diet, 1 mg/kg SLK supplementation diet, and 1.5 mg/kg SLK supplementation diet. This experiment lased two weeks. According to our result, 1.5 mg/kg SLK supplementation diet significantly decreased diarrhea score and diarrhea frequency, and effectively increased survival rate (P < 0.05). Dietary supplementation with 1.5 mg/kg SLK significantly increased high density lipoprotein cholesterol (HDLC), and choline esterase (CHE) concentration in serum (P < 0.05). AS compared with TAS group, 1.5 mg/kg SLK supplementation diet significantly decreased villus height and increased goblet number in jejunum, and increased villus height and decreased goblet number in ileum (P < 0.05). 1.5 mg/kg SLK supplementation diet also significantly changed cecal microbial community composition, including increased Limosilactobacillus abundance (P < 0.05). 1.5 mg/kg SLK supplementation diet significantly increased colonic microbial community composition, including decreased Escherichia-shigella abundance and increased Limosilactobacillus abundance (P < 0.05). Moreover, 1.5 mg/kg SLK supplementation diet significantly increased valerate, propionate, butyrate, and total short chain fatty acid contents in colon (P < 0.05). Valerate, propionate, butyrate, and total short chain fatty acid significantly associated with Lactobacillus. Fourerenilla and Fourerenilla.unclassfied significantly associated with acetate contents in colon (P < 0.05). In conclusion, dietary supplementation with modified apptapulgites significantly regulate intestinal microbial community composition and alleviate intestinal epithelial barrier to prevent diarrhea in piglets.


Assuntos
Suplementos Nutricionais , Microbioma Gastrointestinal , Animais , Butiratos/farmacologia , Diarreia/prevenção & controle , Suplementos Nutricionais/análise , Propionatos/farmacologia , Suínos , Valeratos/farmacologia , Desmame
4.
Front Vet Sci ; 10: 1202369, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37576837

RESUMO

Aspartate (asp), glutamate (glu), and glutamine (gln) are the major energy fuels for the small intestine, and it had been indicated in our previous study that the mix of these three amino acid supplementations could maintain intestinal energy homeostasis. This study aimed to further investigate whether the treatment of gln, glu, and asp in low energy diet affects the intestinal barrier integrity and amino acid pool in weaning piglets. A total of 198 weaned piglets were assigned to 3 treatments: control (basal diet + 1.59% L-Ala); T1 (basal diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp); and T2 (low energy diet + 1% L-Gln + 0.5% L-Glu + 0.1% L-Asp). The blood, jejunum, and ileum were obtained on day 5 or on day 21 post-weaning, respectively. Our results showed that T1 and T2 treatments increased the abundances of occludin, claudin-1, and claudin-3 in the small intestine while decreasing the serum diamine oxidase (DAO) and D-lactate levels in weaning piglets. Meanwhile, T1 and T2 treatments significantly increased the positive rate of proliferating cell nuclear antigen (PCNA) of the small intestine, promoting intestinal cell proliferation. We also found that supplementation with glu, gln, and asp improved the serum amino acid pool and promoted ileal amino acid transporter gene expression of slc3a2, slc6a14, and slc7a11 in weaned piglets. Additionally, on day 21 post-weaning, T1 and T2 treatments stimulated the phosphorylation of the mTOR-S6K1-4EBP1 signaling pathway in the small intestine, which may implicate the enhanced protein synthesis rate. In summary, dietary supplementation of gln, glu, and asp was beneficial to the intestinal barrier function and amino acid pool regulation, while the benefits of gln, glu, and asp treatment might be diminished by the low-energy diet. The results demonstrated that the supplementation of gln, glu, and asp under low energy levels was preferentially supplied as the energy fuel to restore the gut barrier function in piglets on day 5 post-weaning. With the increase in age and intestinal maturation (on day 21 post-weaning), gln, glu, and asp supplementation could also show an effect on the regulation of the amino acid pool and protein synthesis.

5.
Microbiome ; 11(1): 264, 2023 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-38007451

RESUMO

BACKGROUND: Compared to that of bacteria, the role of gut fungi in obesity development remains unknown. RESULTS: Here, alterations in gut fungal biodiversity and composition were confirmed in obese pig models and high-fat diet (HFD)-fed mice. Antifungal drugs improved diet-induced obesity, while fungal reconstruction by cohousing or fecal microbiota transplantation maintained the obese phenotype in HFD-fed mice. Fungal profiling identified 5 fungal species associated with obesity. Specifically, Ascomycota_sp. and Microascaceae_sp. were reduced in obese mice and negatively correlated with fat content. Oral supplementation with fungi was sufficient to prevent and treat diet-induced obesity. Clec7a, which is involved in fungal recognition, was highly expressed in HFD-fed mice. The Clec7a agonist accelerated diet-induced obesity, while Clec7a deficieny in mice resulted in resistance to diet-induced obesity and blocked the anti-obese effect of antifungal drugs and fungi. CONCLUSIONS: Taken together, these results indicate that gut fungi/Clec7a signaling is involved in diet-induced obesity and may have therapeutic implications as a biomarker for metabolic dysregulation in humans. Video Abstract.


Assuntos
Antifúngicos , Obesidade , Animais , Humanos , Camundongos , Dieta Hiperlipídica/efeitos adversos , Fungos , Lipídeos , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Suínos
6.
Front Vet Sci ; 10: 1294095, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38026634

RESUMO

Abnormal hepatic energy metabolism limits the growth and development of piglets. We hypothesized that ß-hydroxybutyrate (BHB) might improve the growth performance of piglets by maintaining hepatic caloric homeostasis. A total of 30 litters of newborn piglets were tracked, and 30 postnatal growth retardation (PGR) piglets and 40 healthy piglets were selected to treat with normal saline with or without BHB (25 mg/kg/days) at 7-d-old. At the age of 42 days, 8 piglets in each group were sacrificed, and serum and liver were collected. Compared with the healthy-control group piglets, PGR piglets showed lower body weight (BW) and liver weight (p < 0.05), and exhibited liver injury and higher inflammatory response. The contents of serum and hepatic BHB were lower (p < 0.05), and gene expression related to hepatic ketone body production were down-regulated in PGR piglets (p < 0.05). While BHB treatment increased BW and serum BHB levels, but decreased hepatic BHB levels in PGR piglets (p < 0.05). BHB alleviated the liver injury by inhibiting the apoptosis and inflammation in liver of PGR piglets (p < 0.05). Compared with the healthy-control group piglets, liver glycogen content and serum triglyceride level of PGR piglets were increased (p < 0.05), liver gluconeogenesis gene and lipogenesis gene expression were increased (p < 0.05), and liver NAD+ level was decreased (p < 0.05). BHB supplementation increased the ATP levels in serum and liver (p < 0.05), whereas decreased the serum glucose, cholesterol, triglyceride and high-density lipoprotein cholesterol levels and glucose and lipid metabolism in liver of PGR piglets (p < 0.05). Therefore, BHB treatment might alleviate the liver injury and inflammation, and improve hepatic energy metabolism by regulating glucose and lipid metabolism, thereby improving the growth performance of PGR piglets.

7.
Front Nutr ; 9: 881371, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35634396

RESUMO

Aims: Small peptides are more energy-saving and efficiently absorbed compared to amino acids. Our study aimed to evaluate the effect of the Lys-Lys dipeptide on the improvement of growth performance, amino acid metabolism, and gut development in suckling piglets. Methods and Results: Twenty-eight newborn suckling piglets were orally administrated with 0.1%, 1%, and 5% Lys-Lys dipeptide for 21 days. Our results showed that the Lys-Lys dipeptide has no significant effect on growth performance and intestinal morphology compared with the control group. We also found that the 1% Lys-Lys dipeptide significantly increased the concentrations of serum Lys, Thr, Phe, and Pro while decreasing Cys compared to the control group. Similarly, the 5% Lys-Lys dipeptide markedly increased the concentrations of serum Lys, Iso, Thr, Asp, Glu, and Pro compared to the control group. Moreover, the Lys-Lys dipeptide downregulated the expression of jejunal Slc7a1, Slc7a2, and Slc15a1 and ileal Slc7a2. Additionally, the Lys-Lys dipeptide decreased the microbiota richness indices and relative abundance of Bacteroidales. Conclusion: In this study, we found that the Lys-Lys dipeptide contributes to the metabolism of amino acids but failed to affect the growth performance of piglets. Additionally, the Lys-Lys dipeptide decreased the relative abundance of Bacteroidales. These results provide a theoretical for the future application and research of Lys-Lys dipeptide in intestinal development of suckling piglets.

8.
Antioxidants (Basel) ; 11(2)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35204135

RESUMO

The gastrointestinal tract is a key source of superoxide so as to be one of the most vulnerable to oxidative stress damage. Ellagic acid (EA), a polyphenol displays widely biological activities owing to its strong antioxidant properties. Here, we investigated the protective benefits of EA on oxidative stress and intestinal barrier injury in paraquet (PQ)-challenged piglets. A total of 40 weaned piglets were randomly divided into five groups: Control, PQ, 0.005% EA-PQ, 0.01% EA-PQ, and 0.02% EA-PQ. Piglets were intraperitoneally injected with 4 mg/kg (BW) PQ or saline on d-18, and sacrificed on d-21 of experiment. EA treatments eliminated growth-check induced by PQ and increased serum superoxide dismutase (SOD) activity but decreased serum malondialdehyde (MDA) level as compared to PQ group. EA supplementation promoted Nrf2 nuclear translocation and enhanced heme oxygenase-1 (HO-1) and quinone oxidoreductase 1 (NQO1) protein abundances of small intestinal mucosa. Additionally, EA improved PQ-induced crypt deepening, goblet cells loss, and villi morphological damage. Consistently, EA increased tight junction protein expression as was evident from the decreased serum diamine oxidase (DAO) levels. EA could ameliorate the PQ-induced oxidative stress and intestinal damage through mediating Nrf2 signaling pathway. Intake of EA-rich food might prevent oxidative stress-mediated gut diseases.

9.
Environ Pollut ; 293: 118572, 2022 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-34838710

RESUMO

Paraquat, a widely used herbicide, causes environmental pollution, and liver injury in humans and animals. As a natural compound in fruits, ellagic acid (EA) shows anti-inflammatory and antioxidant effects. This study examines the beneficial effects of dietary EA against the paraquat-induced hepatic injury and further explores the underlying molecular mechanisms using a piglet model. Post-weaning piglets are fed basal diet supplemented with 50 mg/kg, 100 mg/kg, or 200 mg/kg EA for 3 weeks. At week 2, hepatic injury is induced by 4 mg/kg paraquat followed by 7 days recovery. EA supplementation significantly mitigates paraquat-induced hepatic fibrosis, steatosis, and high apoptotic rate. In agreement, EA supplementation reduces serum pro-inflammatory levels, ameliorates inflammatory cells infiltration into hepatic tissue, which are associated with suppressed NF-κB signaling during paraquat exposure. In addition, EA supplementation significantly improves activities of antioxidative enzymes which were correlated with activated Nrf2/Keap 1 signaling during paraquat exposure. Furthermore, EA supplementation restores cecal microbial community during paraquat exposure. The protective effect of EA is strongly linked with increased relative abundance of Lactobacillus reuteri and Lactobacillus amylovorus. Taken together, EA supplementation effectively reduced the occurrence of hepatic oxidative damage and inflammation induced by paraquat through modulating cecal microbial communities, which provides a novel nutritional therapeutic strategy for hepatic injury.


Assuntos
Doença Hepática Crônica Induzida por Substâncias e Drogas , Microbioma Gastrointestinal , Animais , Ácido Elágico/farmacologia , Humanos , Paraquat/toxicidade , Suínos
10.
J Cachexia Sarcopenia Muscle ; 13(1): 677-695, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34811946

RESUMO

BACKGROUND: Gut health plays a vital role in the overall health and disease control of human and animals. Intestinal oxidative stress is a critical player in the induction and progression of cachexia which is conventionally diagnosed and classified by weight loss. Therefore, reduction of intestinal oxidative injury is a common and highly effective strategy for the maintenance of human and animal health. Here we identify intestinal myeloid differentiation primary response gene 88 (MyD88) as a novel target for intestinal oxidative stress using canonical oxidative stress model induced by paraquat (PQ) in vitro and in vivo. METHODS: Intestinal oxidative stress was induced by administration of PQ in intestinal epithelial cells (IECs) and mouse model. Cell proliferation, apoptosis, DNA damage, mitochondrial function, oxidative status, and autophagy process were measured in wild-type and MyD88-deficient IECs during PQ exposure. Autophagy inhibitor (3-methyladenine) and activator (rapamycin) were employed to assess the role of autophagy in MyD88-deficient IECs during PQ exposure. MyD88 specific inhibitor, ST2825, was used to verify function of MyD88 during PQ exposure in mouse model. RESULTS: MyD88 protein levels and apoptotic rate of IECs are increased in response to PQ exposure (P < 0.001). Intestinal deletion of MyD88 blocks PQ-induced apoptosis (~42% reduction) and DNA damage (~86% reduction), and improves mitochondrial fission (~37% reduction) and function including mitochondrial membrane potential (~23% increment) and respiratory metabolism capacity (~26% increment) (P < 0.01). Notably, there is a marked decrease in reactive oxygen species in MyD88-deficient IECs during PQ exposure (~70% reduction), which are consistent with high activity of antioxidative enzymes (~83% increment) (P < 0.001). Intestinal ablation of MyD88 inhibits mTOR signalling, and further phosphorylates p53 proteins during PQ exposure, which eventually promotes intestinal autophagy (~74% increment) (P < 0.01). Activation of autophagy (rapamycin) promotes IECs growth as compared with 3-methyladenine-treatment during PQ exposure (~173% increment), while inhibition of autophagy (3-methyladenine) exacerbates oxidative stress in MyD88-deficient IECs (P < 0.001). In mouse model, inhibition of MyD88 using specific inhibitor ST2825 followed by PQ treatment effectively ameliorates weight loss (~4% increment), decreased food intake (~92% increment), gastrocnemius and soleus loss (~24% and ~20% increment, respectively), and intestinal oxidative stress in an autophagy dependent manner (P < 0.01). CONCLUSIONS: MyD88 modulates intestinal oxidative stress in an autophagy-dependent mechanism, which suggests that reducing MyD88 level may constitute a putative therapeutic target for intestinal oxidative injury-induced weight loss.


Assuntos
Autofagia , Estresse Oxidativo , Animais , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Paraquat/farmacologia , Doenças da Imunodeficiência Primária , Redução de Peso
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA