Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Biol Macromol ; 269(Pt 2): 132025, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38704076

ABSTRACT

The intestine defends against pathogenic microbial invasion via the secretion of host defense peptides (HDPs). Nutritional immunomodulation can stimulate the expression of endogenous HDPs and enhance the body's immune defense, representing a novel non-antibiotic strategy for disease prevention. The project aims to explore the regulatory mechanism of protegrin-1 (PG-1) expression using sodium phenylbutyrate (PBA) by omics sequencing technology and further investigate the role of key regulatory genes on intestinal health. The results showed that PBA promoted PG-1 expression in intestinal epithelial cells based on cell density through epidermal growth factor receptor (EGFR) and G protein-coupled receptor (GPR43). Transcriptome sequencing and microRNA sequencing revealed that C-X-C motif chemokine receptor 2 (CXCR2) exhibited interactions with PG-1. Pre-treatment cells with a CXCR2 inhibitor (SB225002) effectively suppressed the induction of PG-1 by PBA. Furthermore, SB225002 significantly suppressed the gene expression of HDPs in the jejunum of mice without influencing on the morphology, number of goblet cells, and proliferation of the intestine. CXCR2 inhibition significantly reduced the expression of HDPs during E. coli infection, and resulted in the edema of jejunal epithelial cells. The 16S rDNA analysis of cecal contents showed that the E. coli and SB225002 treatments changed gut microbiota diversity and composition at different taxonomic levels. Correlation analysis suggested a potential regulatory relationship between gut microbiota and HDPs. To that end, a gene involved in the HDP expression, CXCR2, has been identified in the study, which contributes to improving intestinal immune function. PBA may be used as a functional additive to regulate intestinal mucosal function, thereby enhancing the health of the intestinal and host.


Subject(s)
Homeostasis , Intestinal Mucosa , Receptors, Interleukin-8B , Animals , Humans , Male , Mice , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , ErbB Receptors/metabolism , ErbB Receptors/genetics , Escherichia coli Infections/genetics , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Homeostasis/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Receptors, G-Protein-Coupled , Receptors, Interleukin-8B/genetics , Receptors, Interleukin-8B/metabolism
2.
J Agric Food Chem ; 2023 Feb 08.
Article in English | MEDLINE | ID: mdl-36753427

ABSTRACT

Host defense peptides (HDPs) are primary components of the innate immune system with diverse biological functions, such as antibacterial ability and immunomodulatory function. HDPs are produced and released by immune and epithelial cells against microbial invasion, which are widely distributed in humans, animals, plants, and microbes. Notably, there are great differences in endogenous HDP distribution and expression in humans and animals. Moreover, HDP expression could be regulated by exogenous substances, such as nutrients, and different physiological statuses in health and disease. In this review, we systematically assessed the regulation of expression and mechanism of endogenous HDPs from nutrition and disease perspectives, providing a basis to identify the specificity and regularity of HDP expression. Furthermore, the regulation mechanism of HDP expression was summarized systematically, and the differences in the regulation between nutrients and diseases were explored. From this review, we provide novel ideas targeted the immune regulation of HDPs for protecting host health in nutrition and practical and effective new ideas using the immune regulation theory for further research on protecting host health from pathogenic infection and excessive immunity diseases under the global challenge of the antibiotic-abuse-induced series of problems, including food security and microbial resistance.

3.
J Agric Food Chem ; 70(51): 16080-16095, 2022 Dec 28.
Article in English | MEDLINE | ID: mdl-36521060

ABSTRACT

Tryptophan has drawn wide attention due to its involvement in improving intestinal immune defense directly and indirectly by regulating metabolic pathways. The study aims to elucidate the potential modulating roles of tryptophan to protect against intestinal inflammation and elucidate the underlying molecular mechanisms. The protective effects of tryptophan against intestinal inflammation are examined in the lipopolysaccharide (LPS)-induced inflammatory model. We first found that tryptophan markedly (p < 0.01) inhibited proinflammatory cytokines production and nuclear factor κB (NF-κB) pathway activation upon LPS challenge. Next, we demonstrated that tryptophan (p < 0.05) attenuated LPS-caused intestinal mucosal barrier damage by increasing the number of goblet cells, mucins, and antimicrobial peptides (AMPs) in the ileum of mice. In addition, tryptophan (p < 0.05) inhibited LPS-induced autophagic flux through the AMP-activated protein kinase (AMPK)-sirtuin 1 (SIRT1) pathway in the intestinal systems to maintain autophagy homeostasis. Meanwhile, tryptophan also reshaped the gut microbiota composition in LPS-challenge mice by increasing the abundance of short-chain fatty acid (SCFA)-producing bacteria such as Acetivibrio (0.053 ± 0.017 to 0.21 ± 0.0041%). Notably, dietary tryptophan resulted in the activation of metabolic pathways during the inflammatory response. Furthermore, exogenous treatment of tryptophan metabolites kynurenine (Kyn) and 5-HT in porcine intestinal epithelial cells (IPEC-J2 cells) reproduced similar protective effects as tryptophan to attenuate LPS-induced intestinal inflammation through regulating the AMPK-SIRT1-autophagy. Taken together, the present study indicates that tryptophan exhibits intestinal protective and immunoregulatory effects resulting from the activation of metabolic pathways, maintenance of gut mucosal barrier integrity, microbiota composition, and AMPK-SIRT1-autophagy level.


Subject(s)
Sirtuin 1 , Tryptophan , Swine , Mice , Animals , Sirtuin 1/genetics , Sirtuin 1/metabolism , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Lipopolysaccharides , Autophagy , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Dietary Supplements
4.
Nutrients ; 14(14)2022 Jul 08.
Article in English | MEDLINE | ID: mdl-35889766

ABSTRACT

Thymol is a natural antibacterial agent found in the essential oil extracted from thyme, which has been proven to be beneficial in food and medicine. Meanwhile, the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome and autophagy have been reported to play key roles in the progression of liver injury. However, the effects of thymol on the NLRP3 inflammasome and autophagy in protecting the liver remain unclear. The present study used a mouse model with liver injury induced by lipopolysaccharides (LPS) to investigate the regulatory mechanisms of thymol. We found that thymol alleviated LPS-induced liver structural damage, as judged by reduced inflammatory cell infiltration and improved structure. In addition, elevated levels of the liver damage indicators (alanine transaminase (ALT), aspartate transaminase (AST), and total bilirubin (TBIL)) dropped after thymol administration. The mRNA and protein expression of inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6, and IL-22), apoptosis-related genes (caspase3 and caspase9), and the activity of apoptosis-related genes (caspase3 and caspase9) were increased in LPS-treated livers, whereas the changes were alleviated after thymol administration. Thymol inhibited LPS-induced increment in lactate dehydrogenase (LDH) activity in primary hepatocytes of the mouse. In addition, thymol protected mice from liver injury by inhibiting NLRP3 inflammasome activation induced by LPS. Mechanistically, the present study indicates that thymol has liver protective activity resulting from the modulation of the AMP-activated protein kinase-mammalian target of rapamycin (AMPK-mTOR) to regulate the autophagy pathway, hence curbing inflammation.


Subject(s)
Hepatitis , Thymol , AMP-Activated Protein Kinases , Animals , Apoptosis , Autophagy , Hepatitis/drug therapy , Inflammasomes/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Lipopolysaccharides/toxicity , Mice , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , TOR Serine-Threonine Kinases , Thymol/pharmacology , Tumor Necrosis Factor-alpha/metabolism
5.
J Food Biochem ; 46(7): e14126, 2022 07.
Article in English | MEDLINE | ID: mdl-35322444

ABSTRACT

Defensins represent an integral part of the innate immune system to ward off potential pathogens. The study used a rat model to investigate mechanisms by which sodium butyrate (NaB) regulates ß-defensin to inhibit lipopolysaccharide (LPS)-induced nephrotoxicity. We found that NaB alleviated LPS-induced renal structural damage, as judged by reduced renal lesions and improved glomerular vascular structure. In addition, elevated levels of indicators of kidney damage creatinine and blood urine nitrogen, inflammatory mediators TNF-α, and IL-6 dropped after NaB administration. Rat ß-defensin 2 (rBD2), as estimated by mRNA level, was significantly higher in LPS-treated kidneys, whereas the changes of rBD2 reduced in NaB-treated kidneys. In addition, NaB alleviated LPS-induced increase in TLRs mRNA expression. Mechanistically, the present study indicates that NaB has nephroprotective activity resulting from modulation of TLR2/4 to regulate rBD2 expression hence curbing inflammation. PRACTICAL APPLICATIONS: In practice, adding NaB to diet can improve animal performance. Our results suggest that dietary supplementation of NaB increases animal feed intake and improves the body's defense ability to relieve inflammation caused by bacteria. Especially in the age of resistance prohibition, sodium butyrate can partially replace antibiotics to induce the expression of body defensin. It may become a health care product to enhance the body's immunity.


Subject(s)
Butyric Acid , Kidney , Toll-Like Receptor 2 , Toll-Like Receptor 4 , beta-Defensins , Animals , Butyric Acid/pharmacology , Inflammation/metabolism , Kidney/metabolism , Kidney/physiopathology , Lipopolysaccharides/adverse effects , RNA, Messenger , Rats , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , beta-Defensins/genetics
6.
Food Funct ; 13(3): 1360-1369, 2022 Feb 07.
Article in English | MEDLINE | ID: mdl-35044411

ABSTRACT

Bacterial endotoxin invasion reduces intestinal barrier functions, such as intestinal bacterial translocation and enteric infection. In this study, we investigated whether sodium butyrate (NaB) alleviates lipopolysaccharide (LPS)-induced inflammation by reducing intestinal damage and regulating the microflora. Rats were divided into four groups for the intraperitoneal injection of LPSs and intragastric gavage with NaB: Con, LPS, LPS + NaB, and NaB. The results showed that NaB alleviated intestinal villus injury and inflammatory infiltration caused by LPS. NaB supplementation decreased the mRNA levels of toll-like receptor (TLR)-4, tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6), and the trend was most pronounced in the jejunum. The morphology of the intestinal nucleus and mitochondria was further observed by transmission electron microscopy. The results showed that NaB supplementation alleviated LPS-induced nuclear atrophy, apoptosis, mitochondrial damage, and rupture. Moreover, NaB improved the LPS-induced inflammatory response by regulating the intestinal barrier. Furthermore, 16S rRNA sequencing showed that the LPS increased the abundance of the harmful bacterium Bacteroides, while the abundance of beneficial bacteria decreased. In the LPS + NaB group, the intestinal microbiota destroyed by the LPS was rebalanced, including a decrease in Bacteroides and an increase in Bifidobacterium and Odoribacter. In conclusion, NaB alleviates LPS-induced enteritis by regulating inflammatory cytokines, maintaining the mucosal barrier, and restoring the microbiota changes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Butyric Acid/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Butyric Acid/administration & dosage , Dietary Supplements , Disease Models, Animal , Gastrointestinal Microbiome/drug effects , Intestinal Diseases/chemically induced , Intestinal Diseases/prevention & control , Lipopolysaccharides , Male , Rats , Rats, Sprague-Dawley
7.
Front Nutr ; 8: 763700, 2021.
Article in English | MEDLINE | ID: mdl-34859032

ABSTRACT

In China, the use of antibiotics growth promoters as feed additives has been banned. The goal of raising dairy heifers is to gain a relatively high body weight on a high-fiber diet at first mating or calving, thus increasing economic benefits. The objective of this experiment was to explore the effects of supplemental Clostridium butyricum (C. butyricum) on growth performance, rumen fermentation and microbiota, and blood parameters in Holstein heifers. Twenty Holstein heifers [mean ± standard deviation (SD); age = 182 ± 4.20 d, body weight = 197.53 ± 5.94 kg, dry matter intake (DMI) = 6.10 ± 0.38 kg] were randomly assigned to one of two diets group for a 42-day feeding period: (1) basal diet (an untreated control group, i.e., the CON group) or (2) basal diet plus daily 2 × 108 (colony-forming unit, CFU) of C. butyricum per kg of DMI per heifer (the CB group). The results demonstrated that C. butyricum supplementation increased the average daily gain from d 21 to 42 and DMI compared to the control group. Supplementation with C. butyricum significantly decreased the molar proportion of acetate and the acetate to propionate ratio but increased the molar proportion of butyrate and propionate. Compared with the control group, the relative abundance of Butyrivibrio fibrisolvens, Ruminococcus albus, Ruminobacter amylophilus, Ruminococcus flavefaciens, and Streptococcus bovis increased during the trial period in the CB group. However, C. butyricum had no significant effect on the blood parameters in Holstein heifers. In conclusion, these results show that feeding C. butyricum can improve growth performance and rumen fermentation without any negative impact on blood parameters in Holstein heifers.

8.
J Agric Food Chem ; 69(45): 13460-13473, 2021 Nov 17.
Article in English | MEDLINE | ID: mdl-34748328

ABSTRACT

The gastrointestinal tract forms a robust line of defense against invading pathogens through the production of endogenous antimicrobial peptides (AMPs), which are crucial molecules of the innate defense system. Tryptophan could modulate intestinal immunity through promoting the expression of AMPs. However, the precise mechanism needs to be further clarified. In this study, we show that treatment with tryptophan for 24 h triggers (p < 0.05) the expression of porcine ß-defensin (pBD) 1 (62.67 ± 3.10 pg/mL) and pBD2 (74.41 ± 1.33 pg/mL) in the porcine intestinal epithelial cells (IPEC-J2) though calcium-sensing receptor (CaSR)-tryptophan metabolic pathways. Meanwhile, tryptophan alleviates (p < 0.05) intestinal inflammation induced by lipopolysaccharide (LPS) through induction of the defensins and activation of the CaSR-AMP-activated protein kinase (AMPK) pathways in vitro and in vivo. Moreover, the activation of CaSR induces the expression of defensins and decreases the levels of IL-1ß (75.26 ± 2.74 pg/mL) and TNF-α (449.8 ± 23.31 pg/mL) induced by LPS (p < 0.05). Importantly, tryptophan maintains kynurenine homeostasis through the activation of CaSR during the inflammatory response. To that end, the work identifies a regulatory circuit between CaSR signaling and tryptophan metabolic pathways involved in the tryptophan-trigged AMP expression, which contributes to improving intestinal immune defense.


Subject(s)
Receptors, Calcium-Sensing , Tryptophan , Animals , Epithelial Cells/metabolism , Intestines , Metabolic Networks and Pathways , Receptors, Calcium-Sensing/genetics , Receptors, Calcium-Sensing/metabolism , Swine
9.
Front Microbiol ; 12: 738334, 2021.
Article in English | MEDLINE | ID: mdl-34803954

ABSTRACT

The purpose of this experiment was to investigate the changes of carbohydrate composition in fermented total mixed diet and its effects on rumen fermentation, methane production, and rumen microbiome in vitro. The concentrate-to-forage ratio of the total mixed ration (TMR) was 4:6, and TMR was ensiled with lactic acid bacteria and fibrolytic enzymes. The results showed that different TMRs had different carbohydrate compositions and subfractions, fermentation characteristics, and bacterial community diversity. After fermentation, the fermented total mixed ration (FTMR) group had lower contents of neutral detergent fiber, acid detergent fiber, starch, non-fibrous carbohydrates, and carbohydrates. In addition, lactic acid content and relative abundance of Lactobacillus in the FTMR group were higher. Compared with the TMR group, the in vitro ammonia nitrogen and total volatile fatty acid concentrations and the molar proportion of propionate and butyrate were increased in the FTMR group. However, the ruminal pH, molar proportion of acetate, and methane production were significantly decreased in the FTMR group. Notably, we found that the relative abundance of ruminal bacteria was higher in FTMR than in TMR samples, including Prevotella, Coprococcus, and Oscillospira. At the same time, we found that the diversity of methanogens in the FTMR group was lower than that in the TMR group. The relative abundance of Methanobrevibacter significantly decreased, while the relative abundances of Methanoplanus and vadinCA11 increased. The relative abundances of Entodinium and Pichia significantly decreased in the FTMR group compared with the TMR group. These results suggest that FTMR can be used as an environmentally cleaner technology in animal farming due to its ability to improve ruminal fermentation, modulate the rumen microbiome, and reduce methane emissions.

10.
Food Funct ; 12(13): 5821-5836, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-34047325

ABSTRACT

The use of antimicrobial peptide (AMP), found in all forms of life and playing a pivotal role in the innate immune system, has been developed as a new strategy for maintaining intestinal health and reducing antibiotic usage due to its ability to resist pathogens and commensal microbes. The current study investigated the effects of l-threonine on ß-defensin expression, the intestinal mucosal barrier and inflammatory cytokine expression in porcine intestinal epithelial cell lines (IPEC-J2). The results revealed that in IPEC-J2 cells, l-threonine significantly increased the expression of ß-defensin (including pBD-1, pBD-2, and pBD-3) in a dose- and time-dependent manner (P < 0.05). By using different concentrations and treatment times of l-threonine, the results showed that the expression of ß-defensin was upregulated to the greatest extent in IPEC-J2 cells cultured with 1 mM l-threonine for 24 h. Although the mRNA expression levels of ß-defensins were markedly increased (P < 0.05), there was relatively little inducible pBD-1, pBD-2 and pBD-3 mRNA expression at the sub-confluent and confluent densities in comparison with post-confluent densities. Furthermore, we found that l-threonine enhanced the ß-defensin expression by suppressing the expression of SIRT1, which increased acetylated p65 expression, and activating the NF-κB signaling pathway, which induced the translocation of p65 from the cytoplasm to the nucleus. In addition, l-threonine significantly prevented LPS-induced intestinal mucosal barrier damage by attenuating the decreasing tendency of the mRNA expression of Mucin1 and Mucin2 (P < 0.05). Simultaneously, l-threonine enhanced the expression of ß-defensins upon LPS challenge in IPEC-J2 cells (P < 0.05). l-Threonine obviously decreased the mRNA expression of inflammatory cytokines compared to that in untreated cells (P < 0.05). In conclusion, l-threonine can upregulate ß-defensin expression and reduce inflammatory cytokine expression in IPEC-J2 cells; meanwhile, l-threonine alleviates LPS-induced intestinal mucosal barrier damage in porcine intestinal epithelial cells. The l-threonine-mediated modulation of endogenous defensin expression may be a promising approach to reduce antibiotic use, enhance disease resistance and intestinal health in animals.


Subject(s)
Epithelial Cells/drug effects , Intestines/drug effects , NF-kappa B/metabolism , Signal Transduction/drug effects , Sirtuin 1/metabolism , Threonine/pharmacology , Up-Regulation/drug effects , beta-Defensins/metabolism , Animals , Cell Line , Cytokines/genetics , Cytokines/metabolism , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , NF-kappa B/genetics , Neoplasm Proteins , Nucleocytoplasmic Transport Proteins , RNA, Messenger/metabolism , Sirtuin 1/genetics , Swine , Threonine/metabolism , beta-Defensins/genetics
11.
Food Nutr Res ; 642020.
Article in English | MEDLINE | ID: mdl-33281536

ABSTRACT

BACKGROUND: As a major disease affecting dairy cow production worldwide, bovine mastitis is caused by a variety of pathogenic microorganisms that eventually cause mammary gland inflammation. Acremonium terricola culture (ATC) is a new type of affordable feed additive produced by the solid fermentation of A. terricola isolated from Cordyceps gunnii and exerted its anti-inflammatory effect. OBJECTIVES: To evaluate the protective effects of ATC on mastitis and investigate its active mechanism, a lipopolysaccharide (LPS)-induced rat mastitis model was used in two experiments. DESIGN: In Experiment 1, a total of 40 female Sprague-Dawley rats were used to determine the optimal supplementary dose of ATC via gavage trial. In Experiment 2, we examined the effects of an optimal dose of ATC on LPS-induced mastitis in rats. RESULTS: The results of Experiment 1 showed that administration of ATC improved growth performance and antioxidant functions in the serum and the liver, as well as immunoglobulin A, G, and M levels in rat serum, and it decreased the content of alanine aminotransferase, aspartate aminotransferase, triglyceride, cholesterol, low-density lipoprotein, and serum urea nitrogen in rat serum; a dosage of 250-1,250 mg/kg/day was shown to be high enough to be effective. The results of Experiment 2 indicated that ATC can relieve the inflammatory reaction of mammary glands in rats, and the LPS-induced expression of tumor necrosis factor-α, interleukin-1ß, interleukin-6, and inducible nitric oxide synthase significantly decreased after ATC treatment. Moreover, our results demonstrated that ATC markedly enhanced the activity of antioxidase in this rat mastitis model. The results of Western blot analysis revealed that ATC could suppress the expression of toll-like receptor 4, phosphorylation of extracellular signal-regulated kinase, and activity of c-Jun N-terminal kinase in the LPS-stimulated mastitis model. CONCLUSION: Taken together, ATC was shown to exert its anti-inflammatory effect by blocking mitogen-activated protein kinase signaling pathways. These results demonstrate that ATC exerts anti-inflammatory and antioxidant effects in mastitis prevention.

12.
Animals (Basel) ; 10(7)2020 Jul 07.
Article in English | MEDLINE | ID: mdl-32645998

ABSTRACT

Inflammatory bowel disease (IBD) develops as a result of complicated interactions between genetic susceptibility, excessive innate immunity, and environmental factors, which are mainly related to the gut microbiota. The present study aimed to elucidate the protective effects and underlying mechanisms of a short-chain fatty acid salt, sodium butyrate, on colonic inflammation induced by dextran sulfate sodium (DSS) in mice. Pretreatment with sodium butyrate attenuated colitis, as demonstrated by the decreased disease activity index (DAI), colon length shortening, spleen tumidness, and histopathology scores, while maintaining intestinal barrier integrity, as observed by H&E staining and electron microscopy. 16S rRNA sequence analysis revealed that sodium butyrate caused a remarkable alteration of the gut microbiota. Bacteroides, Lachnospiraceae, the Lachnospiraceae NK4A136 group, and Ruminiclostridium 6 presented dramatic differences after sodium butyrate supplementation. This work verifies that sodium butyrate can improve mouse colitis via microbe-host interactions by regulating the microbial community. Taken together, the findings demonstrate that sodium butyrate shows great potential as a probiotic agent for ameliorating colitis.

13.
J Agric Food Chem ; 68(9): 2648-2663, 2020 Mar 04.
Article in English | MEDLINE | ID: mdl-32064872

ABSTRACT

Nutritional regulation of endogenous antimicrobial peptide (AMP) expression is considered a promising nonantibiotic approach to suppressing intestinal infection of pathogen. The current study investigated the effects of l-arginine on LPS-induced intestinal inflammation and barrier dysfunction in vivo and in vitro. The results revealed that l-arginine attenuated LPS-induced inflammatory response, inhibited the downregulation of tight junction proteins (TJP) (p < 0.05) by LPS, and maintained intestinal integrity. In porcine intestinal epithelial cells (IPEC-J2), l-arginine obviously suppressed (p < 0.05) the levels of IL-6 (220.63 ± 2.82), IL-8 (333.95 ± 3.75), IL-1ß (693.08 ± 2.38), and TNF-α (258.04 ± 4.14) induced by LPS. Furthermore, l-arginine diminished the LPS-induced expression of Toll-like receptor 4 (TLR4) and inhibited activation of TLR4-mediated nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways. Importantly, we proposed a new mechanism that l-arginine had the ability to stimulate the expression of porcine epithelial ß-defensins through activating the mammalian target of the rapamycin (mTOR) pathway, which exerts anti-inflammatory influence. Moreover, pBD-1 gene overexpression decreased (p < 0.05) the TNF-α level stimulated by LPS in IPEC-J2 cells (4.22 ± 1.64). The present study indicated that l-arginine enhanced disease resistance through inhibiting the TLR4/NF-κB and MAPK pathways and partially, possibly through increasing the intestinal ß-defensin expression.


Subject(s)
Arginine/administration & dosage , Intestines/immunology , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/immunology , Toll-Like Receptor 4/immunology , beta-Defensins/genetics , Animals , Epithelial Cells/drug effects , Epithelial Cells/immunology , Intestines/drug effects , Lipopolysaccharides/adverse effects , Male , Mice , Mice, Inbred BALB C , Mitogen-Activated Protein Kinases/genetics , NF-kappa B/genetics , Swine , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology , beta-Defensins/immunology
14.
J Agric Food Chem ; 68(2): 512-522, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31870150

ABSTRACT

Host defense peptides (HDPs) are vital mucosal defense effectors of the innate immune response. The expression of HDPs is inducible in epithelial cells by potent endogenous inducers. Herein, our results demonstrate that sodium butyrate (NaB) induces the expression of porcine ß-defensin-3 (pBD3) and porcine epididymis protein 2 splicing variant C (pEP2C) in a dose- and time-dependent manner, without modifying the production of proinflammatory cytokines, in porcine intestinal epithelial cells (IPEC J2). Moreover, NaB promotes toll-like receptor 2 (TLR2) expression. TLR2 silencing inhibits the pBD3 and pEP2C expression induced by NaB but does not abolish the histone deacetylase (HDAC) inhibitory activity of NaB. We found that NaB activated the nuclear factor-κB (NF-κB) pathway. Importantly, the degree of cell confluence governs the regulatory responses but does not affect the HDAC activity of NaB. Furthermore, epidermal growth factor receptor (EGFR), but not the mitogen-activated protein kinase (MAPK) pathway, is vital during the NaB-induced pBD3 and pEP2C regulation process. We also demonstrated that pBD3 overexpression increases interleukin-18 levels. This study showed that NaB simultaneously induces pBD3 and pEP2C via TLR2 and EGFR in IPEC J2 cells without increasing the risk of a harmful inflammatory response.


Subject(s)
Butyric Acid/pharmacology , ErbB Receptors/metabolism , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylases/metabolism , Toll-Like Receptor 2/metabolism , beta-Defensins/metabolism , Animals , Epithelial Cells/drug effects , Epithelial Cells/metabolism , ErbB Receptors/genetics , Gene Expression Regulation/drug effects , Histone Deacetylases/genetics , Interleukin-18/genetics , Interleukin-18/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Swine , Toll-Like Receptor 2/genetics , beta-Defensins/genetics
16.
Int J Mol Sci ; 20(6)2019 Mar 20.
Article in English | MEDLINE | ID: mdl-30897850

ABSTRACT

Antimicrobial peptides (AMPs) have emerged as a promising class of antimicrobial agents that could potentially address the global antibiotic resistance. Generating mirror-like peptides by minimizing dermaseptin family sequences is an effective strategy for designing AMPs. However, the previous research still had some limitations such as lower effectiveness and a narrow spectrum of antibacterial activity. To further expand and hone this strategy, we designed a series of AMPs consisting of the WXMXW-NH2 motif (X represents V, I, F, and W; M represents KAAAKAAAK). The peptides formed α-helices and displayed broad-spectrum antimicrobial activities against eleven types of clinical bacteria including both Gram-negative and Gram-positive bacteria. The optimized peptide WW exhibited high physical rupture by inducing membrane shrinkage, disruption, and lysis. Moreover, WW effectively neutralized endotoxins and inhibited the inflammatory response while having the highest therapeutic index. In conclusion, these results indicated that the peptide WW has potential as a broad-spectrum antimicrobial agent or preservative for overcoming the risk of multidrug resistance in localized or external therapeutic applications.


Subject(s)
Amphibian Proteins/chemistry , Amphibian Proteins/pharmacology , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/pharmacology , Anti-Infective Agents/chemistry , Anti-Infective Agents/pharmacology , Endotoxins/chemistry , Endotoxins/pharmacology , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Peptides/chemistry , Peptides/pharmacology
18.
J Med Chem ; 62(5): 2286-2304, 2019 03 14.
Article in English | MEDLINE | ID: mdl-30742437

ABSTRACT

Poor proteolytic resistance is an urgent problem to be solved in the clinical application of antimicrobial peptides (AMPs), yet common solutions, such as complicated chemical modifications and utilization of d-amino acids, greatly increase the difficulty and cost of producing AMPs. In this work, a set of novel peptides was synthesized based on an antitrypsin/antichymotrypsin hydrolytic peptide structure unit (XYPX) n (X represents I, L, and V; Y represents R and K), which was designed using a systematic natural amino acid arrangement. Of these peptides, 16 with seven repeat units had the highest average selectivity index (GMSI = 99.07) for all of the Gram-negative bacteria tested and remained highly effective in combating Escherichia coli infection in vivo. Importantly, 16 also had dramatic resistance to a high concentration of trypsin/chymotrypsin hydrolysis and exerted bactericidal activity through a membrane-disruptive mechanism. Overall, these findings provide new approaches for the development of antiprotease hydrolytic peptides that target Gram-negative bacteria.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Gram-Negative Bacteria/drug effects , Animals , Antimicrobial Cationic Peptides/chemistry , Biocompatible Materials , Gram-Negative Bacteria/ultrastructure , HEK293 Cells , Humans , Mice , Microbial Sensitivity Tests , Microscopy, Electron, Scanning , Proteolysis , RAW 264.7 Cells , Spectrometry, Fluorescence
19.
Med Res Rev ; 39(3): 831-859, 2019 05.
Article in English | MEDLINE | ID: mdl-30353555

ABSTRACT

Antimicrobial peptides (AMPs), critical components of the innate immune system, are widely distributed throughout the animal and plant kingdoms. They can protect against a broad array of infection-causing agents, such as bacteria, fungi, parasites, viruses, and tumor cells, and also exhibit immunomodulatory activity. AMPs exert antimicrobial activities primarily through mechanisms involving membrane disruption, so they have a lower likelihood of inducing drug resistance. Extensive studies on the structure-activity relationship have revealed that net charge, hydrophobicity, and amphipathicity are the most important physicochemical and structural determinants endowing AMPs with antimicrobial potency and cell selectivity. This review summarizes the recent advances in AMPs development with respect to characteristics, structure-activity relationships, functions, antimicrobial mechanisms, expression regulation, and applications in food, medicine, and animals.


Subject(s)
Anti-Bacterial Agents/pharmacology , Antimicrobial Cationic Peptides/pharmacology , Amino Acid Sequence , Animals , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/classification , Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/classification , Drug Therapy, Combination , Humans , Immunomodulation , Structure-Activity Relationship
20.
Food Nutr Res ; 622018.
Article in English | MEDLINE | ID: mdl-30574051

ABSTRACT

BACKGROUND: Host defense peptides (HDPs) possess direct antibacterial, antineoplastic, and immunomodulatory abilities, playing a vital role in innate immunity. Dietary-regulated HDP holds immense potential as a novel pathway for preventing infection. OBJECTIVE: In this study, we examined the regulation mechanism of HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression by sodium phenylbutyrate (PBA). DESIGN: The effects of PBA on HDP induction and the mechanism involved were studied in porcine intestinal epithelial cell lines (IPEC J2). RESULTS: In this study, the results showed that HDPs (pEP2C, pBD-1, and pBD-3) and cytokines (IL-8 and IL-18) expression was increased significantly upon stimulation with PBA in IPEC J2 cells. Furthermore, toll-like receptor 2 (TLR2) and TLR4 were required for the PBA-mediated upregulation of the HDPs. This process occurred and further activated the NF-κB pathway via the phosphorylation of p65 and an IκB α synthesis delay. Meanwhile, histone deacetylase (HDAC) inhibition and an increased phosphorylation of histone H3 on serine S10 also occurred in PBA-induced HDP expression independently with TLR2 and TLR4. Furthermore, p38-MAPK suppressed PBA-induced pEP2C, pBD-1 pBD-3, IL-8, and IL-18 expression, but ERK1/2 failed to abolish the regulation of pBD-3, IL-8, and IL-18. Moreover, epidermal growth factor receptor (EGFR) is involved in PBA-mediated HDP regulation. CONCLUSIONS: We concluded that PBA induced HDP and cytokine increases but did not cause an excessive pro-inflammatory response, which proceeded through the TLR2 and TLR4-NF-κB pathway and histone modification in IPEC J2 cells.

SELECTION OF CITATIONS
SEARCH DETAIL
...