• Title/Summary/Keyword: Intestinal microbial communities

Search Result 21, Processing Time 0.023 seconds

Inhibition of Escherichia coli O157:H7 Attachment by Interactions Between Lactic Acid Bacteria and Intestinal Epithelial Cells

  • Kim, Young-Hoon;Kim, Sae-Hun;Whang, Kwang-Youn;Kim, Young-Jun;Oh, Se-Jong
    • Journal of Microbiology and Biotechnology
    • /
    • v.18 no.7
    • /
    • pp.1278-1285
    • /
    • 2008
  • The intestinal epithelial cell (IEC) layer of the intestinal tract makes direct contact with a number of microbiota communities, including bacteria known to have deleterious health effects. IECs possess innate protective strategies against pathogenic challenge, which primarily involve the formation of a physicochemical barrier. Intestinal tract mucins are principal components of the mucus layer on epithelial surfaces, and perform a protective function against microbial damage. However, little is currently known regarding the interactions between probiotics/pathogens and epithelial cell mucins. The principal objective of this study was to determine the effects of Lactobacillus on the upregulation of MUC2 mucin and the subsequent inhibition of E. coli O157:H7 attachment to epithelial cells. In the current study, the attachment of E. coli O157:H7 to HT-29 intestinal epithelial cells was inhibited significantly by L. acidophilus A4 and its cell extracts. It is also important to note that the expression of MUC2 mucin was increased as the result of the addition of L. acidophilus A4 cell extracts (10.0 mg/ml), which also induced a significant reduction in the degree to which E. coli O157:H7 attached to epithelial cells. In addition, the mRNA levels of IL-8, IL-1$\beta$, and TNF-$\alpha$ in HT-29 cells were significantly induced by treatment with L. acidophilus A4 extracts. These results indicate that MUC2 mucin and cytokines are important regulatory factors in the immune systems of the gut, and that selected lactobacilli may be able to induce the upregulation of MUC2 mucin and specific cytokines, thereby inhibiting the attachment of E. coli O157:H7.

L-arginine and N-carbamoylglutamic acid supplementation enhance young rabbit growth and immunity by regulating intestinal microbial community

  • Sun, Xiaoming;Shen, Jinglin;Liu, Chang;Li, Sheng;Peng, Yanxia;Chen, Chengzhen;Yuan, Bao;Gao, Yan;Meng, Xianmei;Jiang, Hao;Zhang, Jiabao
    • Asian-Australasian Journal of Animal Sciences
    • /
    • v.33 no.1
    • /
    • pp.166-176
    • /
    • 2020
  • Objective: An experiment was conducted to determine the effects of L-arginine (L-Arg) and N-carbamoylglutamic acid (NCG) on the growth, metabolism, immunity and community of cecal bacterial flora of weanling and young rabbits. Methods: Eighteen normal-grade male weanling Japanese White rabbits (JWR) were selected and randomly divided into 6 groups with or without L-Arg and NCG supplementation. The whole feeding process was divided into weanling stage (day 37 to 65) and young stage (day 66 to 85). The effects of L-Arg and NCG on the growth, metabolism, immunity and development of the ileum and jejunum were compared via nutrient metabolism experiments and histological assessment. The different communities of cecal bacterial flora affected by L-Arg and NCG were assessed using high-throughput sequencing technology and bioinformatics analysis. Results: The addition of L-Arg and NCG enhanced the growth of weanling and young rabbit by increasing the nitrogen metabolism, protein efficiency ratio, and biological value, as well as feed intake and daily weight gain. Both L-Arg and NCG increased the concentration of immunoglobulin A (IgA), IgM, and IgG. NCG was superior to L-Arg in promoting intestinal villus development by increasing villus height, villus height/crypt depth index, and reducing the crypt depth. The effects of L-Arg and NCG on the cecal bacterial flora were mainly concentrated in different genera, including Parabacteroides, Roseburia, dgA-11_gut_group, Alistipes, Bacteroides, and Ruminococcaceae_UCG-005. These bacteria function mainly in amino acid transport and metabolism, energy production and conversion, lipid transport and metabolism, recombination and repair, cell cycle control, cell division, and cell motility. Conclusion: L-Arg and NCG can promote the growth and immunity of weanling and young JWR, as well as effecting the jejunum and ileum villi. L-Arg and NCG have different effects in the promotion of nutrient utilization, relieving inflammation and enhancing adaptability through regulating microbial community.

High-throughput sequencing-based metagenomic and transcriptomic analysis of intestine in piglets infected with salmonella

  • KyeongHye, Won;Dohyun, Kim;Donghyun, Shin;Jin, Hur;Hak-Kyo, Lee;Jaeyoung, Heo;Jae-Don, Oh
    • Journal of Animal Science and Technology
    • /
    • v.64 no.6
    • /
    • pp.1144-1172
    • /
    • 2022
  • Salmonella enterica serovar Typhimurium isolate HJL777 is a virulent bacterial strain in pigs. The high rate of salmonella infection are at high risk of non-typhoidal salmonella gastroenteritis development. Salmonellosis is most common in young pigs. We investigated changes in gut microbiota and biological function in piglets infected with salmonella via analysis of rectal fecal metagenome and intestinal transcriptome using 16S rRNA and RNA sequencing. We identified a decrease in Bacteroides and increase in harmful bacteria such as Spirochaetes and Proteobacteria by microbial community analysis. We predicted that reduction of Bacteroides by salmonella infection causes proliferation of salmonella and harmful bacteria that can cause an intestinal inflammatory response. Functional profiling of microbial communities in piglets with salmonella infection showed increasing lipid metabolism associated with proliferation of harmful bacteria and inflammatory responses. Transcriptome analysis identified 31 differentially expressed genes. Using gene ontology and Innate Immune Database analysis, we identified that BGN, DCN, ZFPM2 and BPI genes were involved in extracellular and immune mechanisms, specifically salmonella adhesion to host cells and inflammatory responses during infection. We confirmed alterations in gut microbiota and biological function during salmonella infection in piglets. Our findings will help prevent disease and improve productivity in the swine industry.

Research on the Diversity of Intestinal Microbial Communities of Red tilefish (Branchiostegus japonicus) by 16S rDNA Sequence Analysis (16S rDNA 염기서열 분석에 의한 옥돔(Branchiostegus japonicus)의 장내미생물 군집의 다양성 조사)

  • Kim, Min-Seon;Lee, Seung-Jong;Heo, Moon-Soo
    • Journal of Life Science
    • /
    • v.28 no.3
    • /
    • pp.361-368
    • /
    • 2018
  • This study investigated the diversity of communities of intestinal microorganisms, separated from the intestinal organs of Red tilefish (Branchiostegus japonicas), collected on the Jeju Coast. First, in the isolation of 1.5% BHIA, MA, TSA and R2A Agar on the medium, there were most colonies in 1.5% BHIA. The results of aerobic culture and anaerobic culture were $1.7{\times}10^6CFU/g^{-1}$ and $1.1{\times}10^5cfu/g^{-1}$, respectively, on average, and 147 pure colonies were separated in total. In 16S rDNA sequencing, there were 58 genera and 74 species, showing 95-100% similarity with the basic strain. They were divided broadly into 5 phyla, and as the main phyletic group, Proteobacteria phylum comprised 50% with 9 families, 35 genera and 35 species of Moraxellaceae, Rhodobacteraceae, Shewanellae, Halomondaceae, Enterobacteriaceae, Vibrionaceae, Hahellaceae, Pseudomonadaceae, and Erythrobacteraceae, with the highest index of dominance. Actinobacteria phylum comprised 24% with 8 families, 11 genera and 17 species of Microbacteriaceae, Intrasporangiaceae, Dietziaceae, Dermabacteraceae, Dermacoccaceae, Nocardiodaceae, Brevibacteriaceae and Propionobacteriacea; Firmicutes phylum, 16% with 6 families, 8 genera and 17 species of Bacillaceae, Staphylcoccaceae, Planococcaceae, Streptococcaceae, Paenibacillaceae and Clostridiaceae; Bacteroidetes phylum, 6% with 2 families, 3 genera and 4 species of Cyclobacteriaceae and Flavobacteriaceae; and Deinococcus-Thermus phylum, 4% with 1 family, 1 genus and 1 species of Deinococcaceae.

Microbial community analysis of an eco-friendly recirculating aquaculture system for olive flounder (Paralichthys olivaceus) using complex microbial probiotics (복합미생물 프로바이오틱을 이용한 환경친화적 넙치 순환여과양식시스템에서의 미생물군집 분석)

  • Rhee, Chaeyoung;Kim, Haham;Emmanuel, S. Aalfin;Kim, Hong-Gi;Won, Seonghun;Bae, Jinho;Bai, Sungchul C.;Koh, Sung-Cheol
    • Korean Journal of Microbiology
    • /
    • v.54 no.4
    • /
    • pp.369-378
    • /
    • 2018
  • This study was conducted to evaluate effects of dietary microbial probiotics on the growth and disease resistance of olive flounder (Paralichthys olivaceus) in a recirculating aquaculture system (RAS), and the effects of the probiotic bioaugmentation on the microbial community structure and water quality. For the analysis, 80 juvenile fish (average weight, $25.7{\pm}7.6g$; average length, $15.2{\pm}1.7cm$) were fed a basal diet containing a commercial microbial product CES-AQ1 (CES; $1{\times}10^9\;CFU/kg$ diet) in an RAS for 8 weeks. Weight gain, the specific growth rate, feed efficiency, and protein efficiency ratio of the fish fed the CES diet in the RAS were 1.5~2.5 times higher than those of fish fed the basal diet alone, or the basal diet containing oxytetracycline (OTC), yeast plus bacterium, or Bacillus subtilis in a still water system. There was no significant difference in the pathogen challenge test between fish fed the OTC diet and fish fed the CES diet in the RAS, suggesting the CES-AQ1 probiotic used in the RAS as a potential replacement for antibiotics. The RAS biofilter maintained the highest microbial diversity and appeared to harbor microbial communities with ammonium oxidation, denitrification, and fish pathogen suppression functions. Ammonia, which is hazardous to fish, was significantly decreased to < 0.5 mg/L in 19 days, indicating the effectiveness of probiotic supplementation to maintain good water quality in RAS. These results suggest that the intestinal microbial communities of fish are stabilized by a probiotic-containing diet (CES) and that bioaugmentation with probiotics may be an eco-friendly and economical supplement for aquaculture of olive flounder, promoting both good water quality and fish health in an RAS.

Components of human breast milk: from macronutrient to microbiome and microRNA

  • Kim, Su Yeong;Yi, Dae Yong
    • Clinical and Experimental Pediatrics
    • /
    • v.63 no.8
    • /
    • pp.301-309
    • /
    • 2020
  • Human breast milk (HBM) is essential for the infant's growth and development right after birth and is an irreplaceable source of nutrition for early human survival. Various infant formulas have many similarities to HBM in many components, but there is no perfect substitute for HBM. Recently, various breast milk components and their roles have been studied according to the development of various analysis techniques. As is already well known, HBM contains about 87%-88% water, and 124-g/L solid components as macronutrients, including about 7% (60-70 g/L) carbohydrates, 1% (8-10 g/L) protein, and 3.8% (35-40 g/L) fat. The composition may vary depending on the environmental factors, including maternal diet. Colostrum is low in fat but high in protein and relatively rich in immuneprotective components. Although HBM contains enough vitamins to ensure normal growth of the infant, vitamins D and K may be insufficient, and the infant may require their supplementation. Growth factors in HBM also serve as various bioactive proteins and peptides on the intestinal tract, vasculature, nervous system, and endocrine system. In the past, HBM of a healthy mother was thought to be sterile. However, several subsequent studies have confirmed the presence of rich and diverse microbial communities in HBM. Some studies suggested that the genera Staphylococcus and Streptococcus may be universally predominant in HBM, but the origin of microbiota still remains controversial. Lastly, milk is the one of most abundant body fluid of microRNAs, which are known to play a role in various functions, such as immunoprotection and developmental programming, through delivering from HBM and absorption by intestinal epithelial cells. In conclusion, HBM is the most important source of nutrition for infants and includes microbiomes and miRNAs for growth, development, and immunity.

Gut-Brain Connection: Microbiome, Gut Barrier, and Environmental Sensors

  • Min-Gyu Gwak;Sun-Young Chang
    • IMMUNE NETWORK
    • /
    • v.21 no.3
    • /
    • pp.20.1-20.18
    • /
    • 2021
  • The gut is an important organ with digestive and immune regulatory function which consistently harbors microbiome ecosystem. The gut microbiome cooperates with the host to regulate the development and function of the immune, metabolic, and nervous systems. It can influence disease processes in the gut as well as extra-intestinal organs, including the brain. The gut closely connects with the central nervous system through dynamic bidirectional communication along the gut-brain axis. The connection between gut environment and brain may affect host mood and behaviors. Disruptions in microbial communities have been implicated in several neurological disorders. A link between the gut microbiota and the brain has long been described, but recent studies have started to reveal the underlying mechanism of the impact of the gut microbiota and gut barrier integrity on the brain and behavior. Here, we summarized the gut barrier environment and the 4 main gut-brain axis pathways. We focused on the important function of gut barrier on neurological diseases such as stress responses and ischemic stroke. Finally, we described the impact of representative environmental sensors generated by gut bacteria on acute neurological disease via the gut-brain axis.

The Role of Gut Microbiota in Obesity and Utilization of Fermented Herbal Extracts (비만에서 장내 미생물 균총의 역할과 발효 한양의 활용)

  • Park, Jung-Hyun;Kim, Ho-Jun;Lee, Myeong-Jong
    • Journal of Korean Medicine for Obesity Research
    • /
    • v.9 no.1
    • /
    • pp.1-14
    • /
    • 2009
  • Complex microbial communities play an important role in the human health and co-evolved with human in the form of symbiosis. Many literatures provide new evidences that the increased prevalence of obesity cannot be attributed solely to changes in the human genome, nutritional habits, or reduction of physical activity in our daily lives. The intestinal flora was recently proposed as an environmental factor responsible for the control of body weight and energy metabolism. A number of studies suggest that the modulation of gut microbiota affects host metabolism and has an impact on energy storage and demonstrated a role for the gut microbiota in weight gain, fat increase, and insulin resistance. Variations in microbiota composition are found in obese humans and mice and the microbiota from an obese mouse confers an obese phenotype when transferred to an axenic mouse. As well, the gut microbial flora plays a role in converting nutrients into calories. Specific strategies for modifying gut microbiota may be a useful means to treat or prevent obesity. Dietary modulations of gut microbiota with a view to increasing bifidobacteria have demonstrated to reduce endotoxemia and improve metabolic diseases such as obesity. The fermentation of medicinal herbs is intended to exert a favorable influence on digestability, bioavailability and pharmacological activity of herbal extract. Therefore we also expect that the fermented herbal extracts may open up a new area to treat obesity through modulating gut microbiota.

  • PDF

Comparison of the gut microbiota profile in breast-fed and formula-fed Korean infants using pyrosequencing

  • Lee, Sang A;Lim, Ji Ye;Kim, Bong-Soo;Cho, Su Jin;Kim, Nak Yon;Kim, Ok Bin;Kim, Yuri
    • Nutrition Research and Practice
    • /
    • v.9 no.3
    • /
    • pp.242-248
    • /
    • 2015
  • BACKGROUND/OBJECTIVES: Feeding in infancy is the most significant determinant of the intestinal microbiota in early life. The aim of this study was to determine the gut microbiota of Korean infants and compare the microbiota obtained between breast-fed and formula-fed Korean infants. SUBJECTS/METHODS: We analyzed the microbial communities in fecal samples collected from twenty 4-week old Korean (ten samples in each breast-fed or formula-fed) infants using pyrosequencing. RESULTS: The fecal microbiota of the 4-week-old Korean infants consisted of the three phyla Actinobacteria, Firmicutes, and Proteobacteria. In addition, five species, including Bifidocbacterium longum, Streptococcus salivarius, Strepotococcus lactarius, Streptococcus pseudopneumoniae, and Lactobacillus gasseri were common commensal intestinal microbiota in all infants. The predominant intestinal microbiota in the breast-fed infants (BFI) included the phylum Actinobacteria (average 70.55%), family Bifidobacteriacea (70.12%), genus Bifidobacterium (70.03%) and species Bifidobacterium longum (69.96%). In the microbiota from the formula-fed infants (FFI), the proportion of the phylum Actinobacteria (40.68%) was less, whereas the proportions of Firmicutes (45.38%) and Proteobacteria (13.85%) as well as the diversity of each taxonomic level were greater, compared to those of the BFI. The probiotic species found in the 4-week-old Korean infants were Bifidobacterium longum, Streptococcus salivarius, and Lactobacillus gasseri. These probiotic species accounted for 93.81% of the microbiota from the BFI, while only 63.80% of the microbiota from the FFI. In particular, B. longum was more abundant in BFI (69.96%) than in FFI (34.17%). CONCLUSIONS: Breast milk supports the growth of B. longum and inhibits others. To the best of our knowledge, this study was the first attempt to analyze the gut microbiota of healthy Korean infants according to the feeding type using pyrosequencing. Our data can be used as a basis for further studies to investigate the development of intestinal microbiota with aging and disease status.

Comparison of gut microbial diversity of breast-fed and formula-fed infants (모유수유와 분유수유에 따른 영아 장내 미생물 군집의 특징)

  • Kim, Kyeong Soon;Shin, Jung;Sim, JiSoo;Yeon, SuJi;Lee, Pyeong An;Chung, Moon Gyu
    • Korean Journal of Microbiology
    • /
    • v.55 no.3
    • /
    • pp.268-273
    • /
    • 2019
  • The intestinal microbiomes vary according to the factors such environment, age and diet. The purpose of this study was to compare the gut microbial diversity between Korean infants receiving breast-fed milk and formula-fed milk. We analyzed microbial communities in stool samples collected from 80 Korean infants using next generation sequencing. Phylum level analysis revealed that microbial communities in both breast-fed infants group (BIG) was dominated by Actinobacteria ($74.22{\pm}3.48%$). Interestingly, the phylum Actinobacteria was dominant in formula-fed infants group A (FIG-A) at $73.46{\pm}4.12%$, but the proportions of phylum Actinobacteria were lower in formulafed infants group B and C (FIG-B and FIG-C) at $66.52{\pm}5.80%$ and $68.88{\pm}4.33%$. The most abundant genus in the BIG, FIG-A, FIG-B, and FIG-C was Bifidobacterium, comprising $73.09{\pm}2.31%$, $72.25{\pm}4.93%$, $63.81{\pm}6.05%$, and $67.42{\pm}5.36%$ of the total bacteria. Furthermore, the dominant bifidobacterial species detected in BIG and FIG-A was Bifidobacterium longum at $68.77{\pm}6.07%$ and $66.85{\pm}4.99%$ of the total bacteria. In contrast, the proportions of B. longum of FIG-B and FIG-C were $58.94{\pm}6.20%$ and $61.86{\pm}5.31%$ of the total bacteria. FIG-A showed a community similar to BIG, which may be due to the inclusion of galactooligosaccharide, galactosyllactose, synergy-oligosaccharide, bifidooligo and improvement material of gut microbiota contained in formula-milk. We conclude that 5-Bifidus factor contained in milk powder promotes the growth of Bifidobacterium genus in the intestines.