• 제목/요약/키워드: Microbiota Modulation

검색결과 47건 처리시간 0.028초

Fenchone Ameliorates Constipation-Predominant Irritable Bowel Syndrome via Modulation of SCF/c-Kit Pathway and Gut Microbiota

  • Li Cui;Bin Zhang;Shuting Zou;Jing Liu;Pingrong Wang;Hui Li;Zhenhai Zhang
    • Journal of Microbiology and Biotechnology
    • /
    • 제34권2호
    • /
    • pp.367-378
    • /
    • 2024
  • In this study we sought to elucidate the therapeutic effects of fenchone on constipation-predominant irritable bowel syndrome (IBS-C) and the underlying mechanisms. An IBS-C model was established in rats by administration of ice water by gavage for 14 days. Fenchone increased the reduced body weight, number of fecal pellets, fecal moisture, and intestinal transit rate, and decreased the enhanced visceral hypersensitivity in the rat model of IBS-C. In addition, fenchone increased the serum content of excitatory neurotransmitters and decreased the serum content of inhibitory neurotransmitters in the IBS-C rat model. Meanwhile, western blot and immunofluorescence experiments indicated that fenchone increased the expressions of SCF and c-Kit. Furthermore, compared with the IBS-C model group, fenchone increased the relative abundance of Lactobacillus, Blautia, Allobaculum, Subdoligranulum, and Ruminococcaceae_UCG-008, and reduced the relative abundance of Bacteroides, Enterococcus, Alistipes, and Escherichia-Shigella on the genus level. Overall, fenchone ameliorates IBS-C via modulation of the SCF/c-Kit pathway and gut microbiota, and could therefore serve as a novel drug candidate against IBS-C.

한국 남부 지역별 돼지 장내 미생물생태 비교분석 (Differences in swine gut microbiota in southern region of Republic of Korea)

  • 김정만;;;운노타쯔야
    • 미생물학회지
    • /
    • 제51권1호
    • /
    • pp.81-85
    • /
    • 2015
  • 성장촉진제로 항생제 사용이 금지가 된 이후, 가축들의 사망률이 증가되어 항생제 대체제를 개발해야 하는 것이 시급하다. 그러한 대체제 개발에 새로운 접근 중 하나는 숙주의 신체적 기능에 영향을 준다고 알려진 장내미생물생태를 조절하는 것이다. 하지만 가축의 장내미생물에 대한 이해가 인간과 비교하여 볼 때 많이 부족한 실정이다. 본 연구에서는 돼지장내미생물생태가 지역적 차이가 있음에 대한 기본적인 정보를 제공한다. 돼지 분변샘플은 제주(n=40), 광주(n=28), 해남(n=30) 농가로부터 채취하였으며, MiSeq을 이용하여 16S rRNA V4 지역을 시퀀싱하였다. 또한 Mothur 파이프라인을 이용하여 MiSeq으로부터 얻은 데이터를 처리하였다. 총 5,642,125 reads를 얻었으며, 에러시퀀스들을 제거한 후 최종적으로 3,868,143 reads가 남았다. Phylum 수준의 taxonomic composition 분석에서는 제주 돼지들이 Firmicutes를 가장 많이 포함하고 있었으며, Operational Taxonomic Units 분포분석에서 또한 지역적 차이에 따라 돼지장내미생물생태가 다르다는 것을 확인하였다. Non-metric multidimensional scaling과 Phyla의 풍부함 사이의 상관관계분석에서는 Actinobacter, Verrucomicrobia, Firmicutes, Fibrobacteres이 세 개의 지역에 있는 돼지들의 장내미생물생태 차이를 나타나게 하는 장내 미생물 요소라는 것을 확인하였다. 그러한 가축의 장내미생물생태는 농장에서 사용하는 사료와 사양관리에 의해 많은 영향을 미치는 것으로 생각된다. 본 연구결과는 돼지장내미생물생태가 지역적으로 많은 차이가 있다는 것을 나타내며, 추후에 가축의 장내미생물생태에 관한 연구는 지역적 차이가 있다는 것을 고려하여 설계해야 될 것이다.

Effect of Probiotic-Fortified Infant Formula on Infant Gut Health and Microbiota Modulation

  • Ju Young Eor;Chul Sang Lee;Sung Ho Moon;Ju Young Cheon;Duleepa Pathiraja;Byeonghyeok Park;Min Jae Shin;Jae-Young Kim;Sangjong Kim;Youngbae Noh;Yunhan Kim;In-Geol Choi;Sae Hun Kim
    • 한국축산식품학회지
    • /
    • 제43권4호
    • /
    • pp.659-673
    • /
    • 2023
  • Compared to infant formula, breast milk is the best source of nutrition for infants; it not only improves the neonatal intestinal function, but also regulates the immune system and gut microbiota composition. However, probiotic-fortified infant formula may further enhance the infant gut environment by overcoming the limitations of traditional infant formula. We investigated the probiotic formula administration for one month by comparing 118 Korean infants into the following three groups: infants in each group fed with breast milk (50), probiotic formula (35), or placebo formula-fed group (33). Probiotic formula improved stool consistency and defecation frequency compared to placebo formula-fed group. The probiotic formula helped maintaining the level of secretory immunoglobulin A (sIgA), which had remarkably decreased over time in placebo formula-fed infants (compared to weeks 0 and 4). Moreover, probiotic formula decreased the acidity of stool and considerably increased the butyrate concentration. Furthermore, the fecal microbiota of each group was evaluated at weeks 0 and 4. The microbial composition was distinct between each groups, and the abundance of health-promoting bacteria increased in the probiotic formula compared to the placebo formula-fed group. In summary, supplementation of probiotic infant formula can help optimize the infant gut environment, microbial composition, and metabolic activity of the microbiota, mimicking those of breast milk.

Gut microbiota-generated metabolites: missing puzzles to hosts' health, diseases, and aging

  • Yan Zhang;Shibo Wei;Hang Zhang;Yunju Jo;Jong-Sun Kang;Ki-Tae Ha;Jongkil Joo;Hyun Joo Lee;Dongryeol Ryu
    • BMB Reports
    • /
    • 제57권5호
    • /
    • pp.207-215
    • /
    • 2024
  • The gut microbiota, an intricate community of bacteria residing in the gastrointestinal system, assumes a pivotal role in various physiological processes. Beyond its function in food breakdown and nutrient absorption, gut microbiota exerts a profound influence on immune and metabolic modulation by producing diverse gut microbiota-generated metabolites (GMGMs). These small molecules hold potential to impact host health via multiple pathways, which exhibit remarkable diversity, and have gained increasing attention in recent studies. Here, we elucidate the intricate implications and significant impacts of four specific metabolites, Urolithin A (UA), equol, Trimethylamine N-oxide (TMAO), and imidazole propionate, in shaping human health. Meanwhile, we also look into the advanced research on GMGMs, which demonstrate promising curative effects and hold great potential for further clinical therapies. Notably, the emergence of positive outcomes from clinical trials involving GMGMs, typified by UA, emphasizes their promising prospects in the pursuit of improved health and longevity. Collectively, the multifaceted impacts of GMGMs present intriguing avenues for future research and therapeutic interventions.

Clostridium difficile colonization and/or infection during infancy and the risk of childhood allergic diseases

  • Lee, Sun Hwa;Gong, Yun Na;Ryoo, Eell
    • Clinical and Experimental Pediatrics
    • /
    • 제60권5호
    • /
    • pp.145-150
    • /
    • 2017
  • Purpose: The gut microbiota can influence several diseases through immune modulation; however, the exact role of microbes such as Clostridium difficile and the relationship between microbiota colonization and allergic diseases are not well known. This study aimed to determine the relationship between C. difficile colonization and/or infection (CDCI) during infancy and allergic diseases during early childhood. Methods: Infants 1-12 months of age presenting changes in bowel habits for more than 2 weeks were enrolled in this study. After dividing them into 2 groups according to the presence and absence of C. difficile, the risk of allergic disease development during childhood was identified and compared. Results: Sixty-five patients were included in this study; 22 (33.8%) were diagnosed with CDCI. No significant differences were observed in baseline characteristics between the C. difficile-positive and-negative groups except for antibiotic exposure (22.7% vs. 60.5%, P=0.004). Compared to the C. difficile-negative group, the risk of developing at least one allergic disease was higher in the C. difficile-positive group after adjusting other variables (adjusted odds ratios, 5.61; 95% confidence interval, 1.52-20.74; P=0.007). Furthermore, food allergies were more prevalent in the C. difficile-positive group (P=0.03). Conclusion: CDCI during infancy were associated with a higher risk of developing allergic diseases during early childhood. These results suggest that CDCI during infancy might reflect the reduced diversity of the intestinal microbiota, which is associated with an increased risk of allergic sensitization. To identify the underlying mechanism, further investigation and a larger cohort study will be needed.

Bacillus amyloliquefaciens and Saccharomyces cerevisiae feed supplements improve growth performance and gut mucosal architecture with modulations on cecal microbiota in red-feathered native chickens

  • Lee, Tzu-Tai;Chou, Chung-Hsi;Wang, Chinling;Lu, Hsuan-Ying;Yang, Wen-Yuan
    • Animal Bioscience
    • /
    • 제35권6호
    • /
    • pp.869-883
    • /
    • 2022
  • Objective: The aim of study was to investigate the effects of in-feed supplementation of Bacillus amyloliquefaciens (BA) and Saccharomyces cerevisiae (SC) on growth performance, gut integrity, and microbiota modulations in red-feathered native chickens (RFCs). Methods: A total of 18,000 RFCs in a commercial farm were evenly assigned into two dietary treatments (control diet; 0.05% BA and 0.05% SC) by randomization and raised for 11 weeks in two separate houses. Fifty RFCs in each group were randomly selected and raised in the original house with the partition for performance evaluations at the age of 9 and 11 weeks. Six non-partitioned RFCs per group were randomly selected for analyses of intestinal architecture and 16S rRNA metagenomics. Results: Feeding BA and SC increased the body weight and body weight gain, significantly at the age of 11 weeks (p<0.05). The villus height/crypt ratio in the small intestines and Firmicutes to Bacteroidetes ratio were also notably increased (p<0.05). The supplementation did not disturb the microbial community structure but promote the featured microbial shifts characterized by the significant increments of Bernesiella, Prevotellaceae_NK3B31_group, and Butyrucimonas, following remarkable decrements of Bacteroides, Rikenellaceae_RC9_gut_group, and Succinatimonas in RFCs with growth benefits. Besides, functional pathways of peptidoglycan biosynthesis, nucleotide excision repair, glycolysis/gluconeogenesis, and aminoacyl transfer ribonucleic acid (tRNA) biosynthesis were significantly promoted (p<0.05). Conclusion: In-feed supplementation of BA and SC enhanced the growth performance, improved mucosal architectures in small intestines, and modulated the cecal microbiota and metabolic pathways in RFCs.

Effect of Korea red ginseng on nonalcoholic fatty liver disease: an association of gut microbiota with liver function

  • Hong, Ji Taek;Lee, Min-Jung;Yoon, Sang Jun;Shin, Seok Pyo;Bang, Chang Seok;Baik, Gwang Ho;Kim, Dong Joon;Youn, Gi Soo;Shin, Min Jea;Ham, Young Lim;Suk, Ki Tae;Kim, Bong-Soo
    • Journal of Ginseng Research
    • /
    • 제45권2호
    • /
    • pp.316-324
    • /
    • 2021
  • Background: Korea Red Ginseng (KRG) has been used as remedies with hepato-protective effects in liver-related condition. Microbiota related gut-liver axis plays key roles in the pathogenesis of chronic liver disease. We evaluated the effect of KRG on gut-liver axis in patients with nonalcoholic statohepatitis by the modulation of gut-microbiota. Methods: A total of 94 patients (KRG: 45 and placebo: 49) were prospectively randomized to receive KRG (2,000 mg/day, ginsenoside Rg1+Rb1+Rg3 4.5mg/g) or placebo during 30 days. Liver function test, cytokeraton 18, and fatigue score were measured. Gut microbiota was analyzed by MiSeq systems based on 16S rRNA genes. Results: In KRG group, the mean levels (before vs. after) of aspartate aminotransferase (53 ± 19 vs. 45 ± 23 IU/L), alanine aminotransferase (75 ± 40 vs. 64 ± 39 IU/L) and fatigue score (33 ± 13 vs. 26 ± 13) were improved (p < 0.05). In placebo group, only fatigue score (34 ± 13 vs. 31 ± 15) was ameliorated (p < 0.05). The changes of phyla were not statistically significant on both groups. In KRG group, increased abundance of Lactobacillus was related with improved alanine aminotransferase level and increased abundance of Clostridium and Intestinibacter was associated with no improvement after KRG supplementation. In placebo group, increased abundance of Lachnospiraceae could be related with aggravation of liver enzyme (p < 0.05). Conclusion: KRG effectively improved liver enzymes and fatigue score by modulating gut-microbiota in patients with fatty liver disease. Further studies are needed to understand the mechanism of improvement of nonalcoholic steatohepatitis. ClnicalTrials.gov: NCT03945123 (www.ClinicalTrials.gov).

Effects of Rubus coreanus byproducts on intestinal microbiota and the immune modulation

  • Yu, Da Yoon;Kim, Sung Hak;Kim, Jeong A;Kim, In Sung;Moon, Yang Soo;Lee, Sang Suk;Park, Hwa Chun;Jung, Jong Hyun;Chung, Yi Hyung;Shin, Dae Keun;Nam, Ki Chang;Choi, In Soon;Cho, Kwang Keun
    • Asian-Australasian Journal of Animal Sciences
    • /
    • 제31권3호
    • /
    • pp.429-438
    • /
    • 2018
  • Objective: Although the efficacy of Rubus coreanus (RC) byproducts as a feed additive has been recognized, its effects on intestinal microorganisms and the immune system are still unknown. Methods: Six-week-old male rats were treated with 0.5% RC (T1), 1.0% RC (T2), and 1.5% RC (T3) for 4 weeks. Results: We found that treatment with RC byproducts significantly increased the daily gain of body weight and feed intake. Treg-cell differentiation was enhanced in the mesenteric lymph nodes and spleen from the rats fed with RC byproducts. Illumina sequencing showed that bacteria in the phylum Firmicutes decreased and while those in the phylum Bacteroidetes increased in RC-treated groups. Particularly, the pathogenic microorganisms in the family Peptococcaceae decreased, and the non-pathogenic families Lachnospiraceae and S24-7 increased. Quantitative polymerase chain reaction analysis showed that the RC byproducts increased the lactic acid bacteria Bifidobacterium spp., Oscillospira spp., Leuconostoc citreum, and Weissella cibaria in a concentration-dependent manner. Conclusion: RC byproducts may be effective in immunomodulation by affecting intestinal microorganisms.

The Role of Meat Protein in Generation of Oxidative Stress and Pathophysiology of Metabolic Syndromes

  • Ahmad, Muhammad Ijaz;Ijaz, Muhammad Umair;Haq, Ijaz ul;Li, Chunbao
    • 한국축산식품학회지
    • /
    • 제40권1호
    • /
    • pp.1-10
    • /
    • 2020
  • Various processing methods have a great impact on the physiochemical and nutritional properties of meat that are of health concern. Hence, the postmortem processing of meat by different methods is likely to intensify the potential effects on protein oxidation. The influence of meat protein oxidation on the modulation of the systemic redox status and underlying mechanism is well known. However, the effects of processed meat proteins isolated from different sources on gut microbiota, oxidative stress biomarkers, and metabolomic markers associated with metabolic syndromes are of growing interest. The application of advanced methodological approaches based on OMICS, and mass spectrometric technologies has enabled to better understand the molecular basis of the effect of processed meat oxidation on human health and the aging process. Animal studies indicate the involvement of dietary proteins isolated from different sources on health disorders, which emphasizes the impact of processed meat protein on the richness of bacterial taxa such as (Mucispirillum, Oscillibacter), accompanied by increased expression of lipogenic genes. This review explores the most recent evidences on meat processing techniques, meat protein oxidation, underlying mechanisms, and their potential effects on nutritional value, gut microbiota composition and possible implications on human health.

Pathophysiology and protective approaches of gut injury in critical illness

  • Jung, Chang Yeon;Bae, Jung Min
    • Journal of Yeungnam Medical Science
    • /
    • 제38권1호
    • /
    • pp.27-33
    • /
    • 2021
  • The gut is a complex organ that has played an important role in digestion, absorption, endocrine functions, and immunity. The gut mucosal barriers consist of the immunologic barrier and nonimmunologic barrier. During critical illnesses, the gut is susceptible to injury due to the induction of intestinal hyperpermeability. Gut hyperpermeability and barrier dysfunction may lead to systemic inflammatory response syndrome. Additionally, gut microbiota are altered during critical illnesses. The etiology of such microbiome alterations in critical illnesses is multifactorial. The interaction or systemic host defense modulation between distant organs and the gut microbiome is increasingly studied in disease research. No treatment modality exists to significantly enhance the gut epithelial integrity, permeability, or mucus layer in critically ill patients. However, multiple helpful approaches including clinical and preclinical strategies exist. Enteral nutrition is associated with an increased mucosal barrier in animal and human studies. The trophic effects of enteral nutrition might help to maintain the intestinal physiology, prevent atrophy of gut villi, reduce intestinal permeability, and protect against ischemia-reperfusion injury. The microbiome approach such as the use of probiotics, fecal microbial transplantation, and selective decontamination of the digestive tract has been suggested. However, its evidence does not have a high quality. To promote rapid hypertrophy of the small bowel, various factors have been reported, including the epidermal growth factor, membrane permeant inhibitor of myosin light chain kinase, mucus surrogate, pharmacologic vagus nerve agonist, immune-enhancing diet, and glucagon-like peptide-2 as preclinical strategies. However, the evidence remains unclear.