• Title/Summary/Keyword: bacterial microbiota

Search Result 157, Processing Time 0.03 seconds

Rumen bacteria influence milk protein yield of yak grazing on the Qinghai-Tibet plateau

  • Fan, Qingshan;Wanapat, Metha;Hou, Fujiang
    • Animal Bioscience
    • /
    • v.34 no.9
    • /
    • pp.1466-1478
    • /
    • 2021
  • Objective: Ruminants are completely dependent on their microbiota for rumen fermentation, feed digestion, and consequently, their metabolism for productivity. This study aimed to evaluate the rumen bacteria of lactating yaks with different milk protein yields, using high-throughput sequencing technology, in order to understand the influence of these bacteria on milk production. Methods: Yaks with similar high milk protein yield (high milk yield and high milk protein content, HH; n = 12) and low milk protein yield (low milk yield and low milk protein content, LL; n = 12) were randomly selected from 57 mid-lactation yaks. Ruminal contents were collected using an oral stomach tube from the 24 yaks selected. High-throughput sequencing of bacterial 16S rRNA gene was used. Results: Ruminal ammonia N, total volatile fatty acids, acetate, propionate, and isobutyrate concentrations were found to be higher in HH than LL yaks. Community richness (Chao 1 index) and diversity indices (Shannon index) of rumen microbiota were higher in LL than HH yaks. Relative abundances of the Bacteroidetes and Tenericutes phyla in the rumen fluid were significantly increased in HH than LL yaks, but significantly decreased for Firmicutes. Relative abundances of the Succiniclasticum, Butyrivibrio 2, Prevotella 1, and Prevotellaceae UCG-001 genera in the rumen fluid of HH yaks was significantly increased, but significantly decreased for Christensenellaceae R-7 group and Coprococcus 1. Principal coordinates analysis on unweighted UniFrac distances revealed that the bacterial community structure of rumen differed between yaks with high and low milk protein yields. Furthermore, rumen microbiota were functionally enriched in relation to transporters, ABC transporters, ribosome, and urine metabolism, and also significantly altered in HH and LL yaks. Conclusion: We observed significant differences in the composition, diversity, fermentation product concentrations, and function of ruminal microorganisms between yaks with high and low milk protein yields, suggesting the potential influence of rumen microbiota on milk protein yield in yaks. A deeper understanding of this process may allow future modulation of the rumen microbiome for improved agricultural yield through bacterial community design.

Effects of nasopharyngeal microbiota in respiratory infections and allergies

  • Kang, Hyun Mi;Kang, Jin Han
    • Clinical and Experimental Pediatrics
    • /
    • v.64 no.11
    • /
    • pp.543-551
    • /
    • 2021
  • The human microbiome, which consists of a collective cluster of commensal, symbiotic, and pathogenic microorganisms living in the human body, plays a key role in host health and immunity. The human nasal cavity harbors commensal bacteria that suppress the colonization of opportunistic pathogens. However, dysbiosis of the nasal microbial community is associated with many diseases, such as acute respiratory infections including otitis media, sinusitis and bronchitis and allergic respiratory diseases including asthma. The nasopharyngeal acquisition of pneumococcus, which exists as a pathobiont in the nasal cavity, is the initial step in virtually all pneumococcal diseases. Although the factors influencing nasal colonization and elimination are not fully understood, the adhesion of opportunistic pathogens to nasopharyngeal mucosa receptors and the eliciting of immune responses in the host are implicated in addition to bacterial microbiota properties and colonization resistance dynamics. Probiotics or synbiotic interventions may show promising and effective roles in the adjunctive treatment of dysbiosis; however, more studies are needed to characterize how these interventions can be applied in clinical practice in the future.

Babeisa duncani infection alters gut microbiota profile in hamsters

  • Shangdi Zhang;Jinming Wang;Xiaoyun Li;Yanbo Wang;Yueli Nian;Chongge You;Dekui Zhang;Guiquan Guan
    • Parasites, Hosts and Diseases
    • /
    • v.61 no.1
    • /
    • pp.42-52
    • /
    • 2023
  • The genus Babesia includes parasites that can induce human and animal babesiosis, which are common in tropical and subtropical regions of the world. The gut microbiota has not been examined in hamsters infected by Babesia duncani. Red blood cells infected with B. duncani were injected into hamsters through intraperitoneal route. To evaluate the changes in gut microbiota, DNAs were extracted from small intestinal contents, acquired from hamsters during disease development. Then, the V4 region of the 16S rRNA gene of bacteria was sequenced using the Illumina sequencing platform. Gut microbiota alternation and composition were assessed according to the sequencing data, which were clustered with >97.0% sequence similarity to create amplicon sequence variants (ASVs). Bacteroidetes and Firmicutes were made up of the major components of the gut microbiota in all samples. The abundance of Bacteroidetes elevated after B. duncani infection than the B. duncani-free group, while Firmicutes and Desulfobacterota declined. Alpha diversity analysis demonstrated that the shown ASVs were substantially decreased in the highest parasitemia group than B. duncani-free and lower parasitemia groups. Potential biomarkers were discovered by Linear discriminant analysis Effect Size (LEfSe) analysis, which demonstrated that several bacterial families (including Muribaculaceae, Desulfovibrionaceae, Oscillospiraceae, Helicobacteraceae, Clostridia UGG014, Desulfovibrionaceae, and Lachnospiraceae) were potential biomarkers in B. duncani-infected hamsters. This research demonstrated that B. duncani infectious can modify the gut microbiota of hamsters.

Intestine Bacterial Microbiota of Asian Hornet (Vespa Velutina Nigrithorax) and Honey Bee (등검은말벌과 꿀벌의 장내 세균 군집 비교)

  • Kim, Euyeon;Seo, Jeongwon;Yang, So Hee;Kim, In-Seon;Koo, Yeonjong
    • Korean Journal of Environmental Agriculture
    • /
    • v.37 no.2
    • /
    • pp.135-140
    • /
    • 2018
  • BACKGROUND: The Asian hornet (Vespa velutina nigrithorax), a wasp species, has attacked honey bee populations and affected the beekeeping industry in Korea over the past 15 years. However, little research has been done with this invasive species. In this study, we investigated the intestine bacterial microbiota of Asian hornets and honey bees to design an attractive trap for Asian hornets. METHODS AND RESULTS: Genomic DNAs isolated from the intestine microorganisms of Asian hornets and honey bees were utilized to amplify bacterial 16S rDNA for the comparative sequence analysis. The next generation sequencing analysis identified that the orders Flavobacteriales as the most abundant intestinal microorganisms in Asian hornets, showing a clear difference compared to honey bees in which Aeromonadales are dominant. We also report five newly identified 16S rDNA sequences of Asian hornet intestinal bacteria. According to the sequence blast search, these five bacteria belong to the genera Thalassomonas, Caedobacter, Vampirovibrio, Alkaliphilus and Calothrix. CONCLUSION: While Asian hornets and honey bees show similar intestine bacterial diversity, the relative ratio of bacterial populations is different. providing useful information to design pest control agents specifically targeting Asian hornets.

PD-1 deficiency protects experimental colitis via alteration of gut microbiota

  • Park, Seong Jeong;Kim, Ji-Hae;Song, Mi-Young;Sung, Young Chul;Lee, Seung-Woo;Park, Yunji
    • BMB Reports
    • /
    • v.50 no.11
    • /
    • pp.578-583
    • /
    • 2017
  • Programmed cell death-1 (PD-1) is a coinhibitory molecule and plays a pivotal role in immune regulation. Here, we demonstrate a role for PD-1 in pathogenesis of inflammatory bowel disease (IBD). Wild-type (WT) mice had severe wasting disease during experimentally induced colitis, while mice deficient for PD-1 ($PD-1^{-/-}$) did not develop colon inflammation. Interestingly, $PD-1^{-/-}$ mice cohoused with WT mice became susceptible to colitis, suggesting that resistance of $PD-1^{-/-}$ mice to colitis is dependent on their gut microbiota. 16S rRNA gene-pyrosequencing analysis showed that $PD-1^{-/-}$ mice had altered composition of gut microbiota with significant reduction in Rikenellaceae family. These altered colon bacteria of $PD-1^{-/-}$ mice induced less amount of inflammatory mediators from colon epithelial cells, including interleukin (IL)-6, and inflammatory chemokines. Taken together, our study indicates that PD-1 expression is involved in the resistance to experimental colitis through altered bacterial communities of colon.

The role of rumen microbiota in enteric methane mitigation for sustainable ruminant production

  • Takumi Shinkai;Shuhei Takizawa;Miho Fujimori;Makoto Mitsumori
    • Animal Bioscience
    • /
    • v.37 no.2_spc
    • /
    • pp.360-369
    • /
    • 2024
  • Ruminal methane production functions as the main sink for metabolic hydrogen generated through rumen fermentation and is recognized as a considerable source of greenhouse gas emissions. Methane production is a complex trait affected by dry matter intake, feed composition, rumen microbiota and their fermentation, lactation stage, host genetics, and environmental factors. Various mitigation approaches have been proposed. Because individual ruminants exhibit different methane conversion efficiencies, the microbial characteristics of low-methane-emitting animals can be essential for successful rumen manipulation and environment-friendly methane mitigation. Several bacterial species, including Sharpea, uncharacterized Succinivibrionaceae, and certain Prevotella phylotypes have been listed as key players in low-methane-emitting sheep and cows. The functional characteristics of the unclassified bacteria remain unclear, as they are yet to be cultured. Here, we review ruminal methane production and mitigation strategies, focusing on rumen fermentation and the functional role of rumen microbiota, and describe the phylogenetic and physiological characteristics of a novel Prevotella species recently isolated from low methane-emitting and high propionate-producing cows. This review may help to provide a better understanding of the ruminal digestion process and rumen function to identify holistic and environmentally friendly methane mitigation approaches for sustainable ruminant production.

Effects of fermented coffee on human gut microbiota (발효커피가 사람장내미생물에 미치는 영향)

  • Ko, Gwangpyo;Kim, Jin-Kyeong;Jo, Seong-Wha;Jeong, Do-Youn;Unno, Tatsuya
    • Journal of Applied Biological Chemistry
    • /
    • v.63 no.1
    • /
    • pp.83-87
    • /
    • 2020
  • Fermented foods have been recognized as functional foods that provide health benefits, including the modulation of intestinal microbiota. Therefore, the aim of the present study was to examine the effects of coffee beans fermented with Lactobacillus plantarum and Bacillus amyloliquefaciens on healthy human gut microbiota. Fermentation increased the content of beneficial substances (i.e., flavonoids and polyphenols). The consumption of fermented coffee increased the occurrence of beneficial microorganisms such as fiber degraders and short-chain fatty acid producers, although no significant microbiota shifts were observed after the coffee consumption. The analysis of metabolic activities also showed no difference after the coffee consumption. Our study demonstrates that the consumption of the fermented coffee may increase some beneficial bacterial while remaining the gut microbiota and its activities.

Changes in gut microbiota with mushroom consumption (버섯 섭취와 장내 미생물 균총의 변화)

  • Kim, Eui-Jin;Shin, Hyun-Jae
    • Journal of Mushroom
    • /
    • v.19 no.3
    • /
    • pp.115-125
    • /
    • 2021
  • Mushroom consumption causes changes in the immune system and gut microbiota via the actions of mushroom probiotic components. β-Glucan structure-related substances suppress secretion of inflammatory mediators, and induce macrophage activation, enhancing immunity and immune function. Substances other than directly useful components can be metabolized into short-chain fatty acids by gut microbiota. These short-chain fatty acids can then induce immunity, alleviating various diseases. Substances used to stimulate growth of health-promoting gut bacteria, thereby changing the gut microbiota community are defined to be probiotics. Probiotic altered intestinal microflora can prevent various types of bacterial infection from external sources, and can help to maintain immune system balance, thus preventing diseases. Research into beneficial components of Pleurotus eryngii, Lentinula edodes, Pleurotus ostreatus, Flammulina velutipes, Auricularia auricula-judae, and Agaricus bisporus, which are frequently consumed in Korea, changes in microbiota, changes in short-chain fatty acids, and correlations between consumption and health contribute to our understanding of the effects of dietary mushrooms on disease prevention and mitigation.

Enhanced pig production: potential use of insect gut microbiota for pig production

  • Shin, Jiwon;Kim, Bo-Ra;Guevarra, Robin B.;Lee, Jun Hyung;Lee, Sun Hee;Kim, Young Hwa;Wattanaphansak, Suphot;Kang, Bit Na;Kim, Hyeun Bum
    • Korean Journal of Agricultural Science
    • /
    • v.45 no.4
    • /
    • pp.655-663
    • /
    • 2018
  • The insect gut microbiome is known to have important roles in host growth, development, digestion, and resistance against pathogens. In addition, the genetic diversity of the insect gut microbiota has recently been recognized as potential genetic resources for industrial bioprocessing. However, there is limited information regarding the insect gut microbiota to better help us understand their potential benefits for enhanced pig production. With the development of next-generation sequencing methods, whole genome sequence analysis has become possible beyond traditional culture-independent methods. This improvement makes it possible to identify and characterize bacteria that are not cultured and located in various environments including the gastrointestinal tract. Insect intestinal microorganisms are known to have an important role in host growth, digestion, and immunity. These gut microbiota have recently been recognized as potential genetic resources for livestock farming which is using the functions of living organisms to integrate them into animal science. The purpose of this literature review is to emphasize the necessity of research on insect gut microbiota and their applicability to pig production or bioindustry. In conclusion, bacterial metabolism of feed in the gut is often significant for the nutrition intake of animals, and the insect gut microbiome has potential to be used as feed additives for enhanced pig performance. The exploration of the structure and function of the insect gut microbiota needs further investigation for their potential use in the swine industry particularly for the improvement of growth performance and overall health status of pigs.

Prebiotics enhance the biotransformation and bioavailability of ginsenosides in rats by modulating gut microbiota

  • Zhang, Xiaoyan;Chen, Sha;Duan, Feipeng;Liu, An;Li, Shaojing;Zhong, Wen;Sheng, Wei;Chen, Jun;Xu, Jiang;Xiao, Shuiming
    • Journal of Ginseng Research
    • /
    • v.45 no.2
    • /
    • pp.334-343
    • /
    • 2021
  • Background: Gut microbiota mainly function in the biotransformation of primary ginsenosides into bioactive metabolites. Herein, we investigated the effects of three prebiotic fibers by targeting gut microbiota on the metabolism of ginsenoside Rb1 in vivo. Methods: Sprague Dawley rats were administered with ginsenoside Rb1 after a two-week prebiotic intervention of fructooligosaccharide, galactooligosaccharide, and fibersol-2, respectively. Pharmacokinetic analysis of ginsenoside Rb1 and its metabolites was performed, whilst the microbial composition and metabolic function of gut microbiota were examined by 16S rRNA gene amplicon and metagenomic shotgun sequencing. Results: The results showed that peak plasma concentration and area under concentration time curve of ginsenoside Rb1 and its intermediate metabolites, ginsenoside Rd, F2, and compound K (CK), in the prebiotic intervention groups were increased at various degrees compared with those in the control group. Gut microbiota dramatically responded to the prebiotic treatment at both taxonomical and functional levels. The abundance of Prevotella, which possesses potential function to hydrolyze ginsenoside Rb1 into CK, was significantly elevated in the three prebiotic groups (P < 0.05). The gut metagenomic analysis also revealed the functional gene enrichment for terpenoid/polyketide metabolism, glycolysis, gluconeogenesis, propanoate metabolism, etc. Conclusion: These findings imply that prebiotics may selectively promote the proliferation of certain bacterial stains with glycoside hydrolysis capacity, thereby, subsequently improving the biotransformation and bioavailability of primary ginsenosides in vivo.