• Title/Summary/Keyword: gut dysbiosis

Search Result 42, Processing Time 0.034 seconds

Alteration of Lung and Gut Microbiota in IL-13-Transgenic Mice Simulating Chronic Asthma

  • Sohn, Kyoung-Hee;Baek, Min-gyung;Choi, Sung-Mi;Bae, Boram;Kim, Ruth Yuldam;Kim, Young-Chan;Kim, Hye-Young;Yi, Hana;Kang, Hye-Ryun
    • Journal of Microbiology and Biotechnology
    • /
    • 제30권12호
    • /
    • pp.1819-1826
    • /
    • 2020
  • Increasing evidence suggests a potential role of microbial colonization in the inception of chronic airway diseases. However, it is not clear whether the lung and gut microbiome dysbiosis is coincidental or a result of mutual interaction. In this study, we investigated the airway microbiome in interleukin 13 (IL-13)-rich lung environment and related alterations of the gut microbiome. IL-13-overexpressing transgenic (TG) mice presented enhanced eosinophilic inflammatory responses and mucus production, together with airway hyperresponsiveness and subepithelial fibrosis. While bronchoalveolar lavage fluid and cecum samples obtained from 10-week-old IL-13 TG mice and their C57BL/6 wild-type (WT) littermates showed no significant differences in alpha diversity of lung and gut microbiome, they presented altered beta diversity in both lung and gut microbiota in the IL-13 TG mice compared to the WT mice. Lung-specific IL-13 overexpression also altered the composition of the gut as well as the lung microbiome. In particular, IL-13 TG mice showed an increased proportion of Proteobacteria and Cyanobacteria and a decreased amount of Bacteroidetes in the lungs, and depletion of Firmicutes and Proteobacteria in the gut. The patterns of polymicrobial interaction within the lung microbiota were different between WT and IL-13 TG mice. For instance, in IL-13 TG mice, lung Mesorhizobium significantly affected the alpha diversity of both lung and gut microbiomes. In summary, chronic asthma-like pathologic changes can alter the lung microbiota and affect the gut microbiome. These findings suggest that the lung-gut microbial axis might actually work in asthma.

반려견 장내미생물의 조성, 다양성, 관련 질환 및 치료에 대한 이해: 친구인가 적인가? (The Comprehension of Composition, Diversity, Related Diseases, and Treatment of the Gut Microbiome in Companion Dogs: Friend or Foe?)

  • 최정현;홍용근
    • 생명과학회지
    • /
    • 제30권11호
    • /
    • pp.1021-1032
    • /
    • 2020
  • 전 세계적으로 반려동물과 반려동물 양육 인구는 증가하고 있으며 이와 관련된 사업은 고급화, 전문화되고 있다. 특히, 개는 사람과 수 천년 동안 공생하고 있으며 이들은 탄수화물 식이와 더불어 사람과 비슷한 생활 방식을 영위하고 있다. 주거와 음식을 공유하며 형성된 강력한 유대관계를 통해 사람은 반려견에 대한 책임감 및 정서적 안정을 느끼며 반려견은 사람을 의지하는 행동을 보인다. 하지만 사람과 반려견 사이에는 식이와 관련된 스트레스가 존재한다. 반려견은 본질적으로 육식성이므로, 반려견에게 영양학적으로 균형 잡힌 식이와 기능성 물질들을 제공하는 것은 그들의 건강을 위한 장내 미생물 군집 유지에 매우 중요하다고 할 수 있다. 장내미생물은 병원균과 같은 외부 유해인자로부터 반려견을 보호하고 면역계를 조절, 생리학적 항상성에도 관여하기 때문에 최근 높은 관심을 받고 있다. 비정상적인 장내미생물 군집 형성은 장질환, 대사질환, 정신질환, 신경계 질환의 병리적 과정과 증상에 영향을 미칠 뿐만 아니라 반려견의 질병은 사람에게 기회감염을 통해 영향을 미칠 수 있다. 따라서 본 논문에서 반려견의 장내미생물의 조성, 다양성뿐만 아니라 장내미생물과 질병 사이의 상관성에 대하여 논의하고자 한다.

저식이섬유 및 고지방 사료 급여 마우스의 장내 미생물 생태 변화 (Comparison of gut microbiome between low fiber and high fat diet fed mice)

  • 황낙원;엄태길;운노타쯔야
    • Journal of Applied Biological Chemistry
    • /
    • 제61권2호
    • /
    • pp.165-172
    • /
    • 2018
  • 경제발전으로 인해 한국인의 식습관이 점차 서구화됨에 따라 웰빙(Well-being)의 문제가 야기되고 있다. 웰빙은 장내 미생물 군집과 밀접하게 연관되어 있으며, 이는 섭취한 음식에 따라 가변적이다. 이에 본 연구에서는 장내 미생물의 16S rRNA 유전자를 기반으로 하여 MiSeq을 진행하였고, 고지방 식이(HFD) 및 저식이섬유 식이(LFD)로 인한 장내의 미생물 생태 비교 및 분석하고자 수행되었다. 일반 대조군(CTL) 그룹과 비교하여 각각 LFD 그룹과 HFD 그룹은 species richness가 유의적으로 감소하였고, species evenness에서는 차이가 나타나지 않았다. phylum 수준에서는 Proteobacteria는 두 처리군에서 유의적으로 증가하였고(p<0.05), 그 중 Sutterella genus가 유의적으로 가장 많이 증가하였다. Bacteroidetes는 HFD 그룹에서 유의적으로 감소하였고, S24-7 family가 가장 큰 비율로 감소하였다. 한편 Firmicutes는 HFD:LFD 그룹에서 차이를 보였고, LFD 그룹에서 Lachnospiraceae family가 유의적으로 낮은 비율로 나타난 것이 확인되었다(p<0.05). PICUSt 기반 신진대사 분석에서 LFD 그룹은 아미노산 대사 및 탄수화물 대사에 관여하는 미생물 수가 유의적으로 감소하는 양상을 보였고(p<0.05), 에너지 대사에서는 메탄 대사에 관여하는 미생물이 유의적으로 감소하였다(p<0.01). 한편 HFD 그룹에서는 아미노산 대사에 관여하는 미생물 수가 유의적으로 증가하였다(p<0.05). 글리칸 생합성 및 대사에 관여하는 미생물은 LFD 그룹과 HFD 그룹에서 유의적으로 증가하는 것으로 나타났다(p<0.01). 이상의 결과를 통해 지속적으로 불균형한 식단을 섭취하는 것은 장내 환경을 dysbiosis시켜, 대사성 질환 및 장 기능 저하를 유발할 것으로 예상된다.

Interplays between human microbiota and microRNAs in COVID-19 pathogenesis: a literature review

  • Hong, Bok Sil;Kim, Myoung-Ryu
    • 운동영양학회지
    • /
    • 제25권2호
    • /
    • pp.1-7
    • /
    • 2021
  • [Purpose] Recent studies have shown that COVID-19 is often associated with altered gut microbiota composition and reflects disease severity. Furthermore, various reports suggest that the interaction between COVID-19 and host-microbiota homeostasis is mediated through the modulation of microRNAs (miRNAs). Thus, in this review, we aim to summarize the association between human microbiota and miRNAs in COVID-19 pathogenesis. [Methods] We searched for the existing literature using the keywords such "COVID-19 or microbiota," "microbiota or microRNA," and "COVID-19 or probiotics" in PubMed until March 31, 2021. Subsequently, we thoroughly reviewed the articles related to microbiota and miRNAs in COVID-19 to generate a comprehensive picture depicting the association between human microbiota and microRNAs in the pathogenesis of COVID-19. [Results] There exists strong experimental evidence suggesting that the composition and diversity of human microbiota are altered in COVID-19 patients, implicating a bidirectional association between the respiratory and gastrointestinal tracts. In addition, SARS-CoV-2 encoded miRNAs and host cellular microRNAs modulated by human microbiota can interfere with viral replication and regulate host gene expression involved in the initiation and progression of COVID-19. These findings suggest that the manipulation of human microbiota with probiotics may play a significant role against SARS-CoV-2 infection by enhancing the host immune system and lowering the inflammatory status. [Conclusion] The human microbiota-miRNA axis can be used as a therapeutic approach for COVID-19. Hence, further studies are needed to investigate the exact molecular mechanisms underlying the regulation of miRNA expression in human microbiota and how these miRNA profiles mediate viral infection through host-microbe interactions.

Lactic Acid Bacteria Isolated from Human Breast Milk Improve Colitis Induced by 2,4,6-Trinitrobenzene Sulfonic Acid by Inhibiting NF-κB Signaling in Mice

  • Kyung-Joo Kim;Suhyun Kyung;Hui Jin;Minju Im;Jae-won Kim;Hyun Su Kim;Se-Eun Jang
    • Journal of Microbiology and Biotechnology
    • /
    • 제33권8호
    • /
    • pp.1057-1065
    • /
    • 2023
  • Inflammatory bowel disease (IBD), a chronic inflammatory disease, results from dysregulation of the immune responses. Some lactic acid bacteria (LAB), including Lactobacillus, alleviate IBD through immunomodulation. In this study, the anti-colitis effect of LAB isolated from human breast milk was investigated in a mouse model induced acute colitis with 2,4,6-trinitrobenzene sulfonic acid (TNBS). TNBS remarkably increased weight loss, colon shortening, and colonic mucosal proliferation, as well as the expression levels of inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin (IL)-1β. Oral administration of LAB isolated from human breast milk resulted in a reduction in TNBS-induced colon shortening, as well as induced cyclooxygenase (COX)-2, nitric oxide synthase (iNOS), nuclear factor-kappa B (NF-κB). In addition, LAB suppressed inflammatory cytokines such as TNF-α, IL-6, and IL-1β, and thus showed an effect of suppressing the level of inflammation induced by TNBS. Furthermore, LAB alleviated gut microbiota dysbiosis, and inhibited intestinal permeability by increasing the expression of intestinal tight junction protein including ZO-1. Collectively, these results suggest that LAB isolated from human breast milk can be used as a functional food for colitis treatment by regulating NF-κB signaling, gut microbiota and increasing expression of intestinal tight junction protein.

In Vitro Evaluation of Probiotic Properties of Two Novel Probiotic Mixtures, Consti-Biome and Sensi-Biome

  • You Jin Jang;Bonggyu Min;Jong Hyun Lim;Byung-Yong Kim
    • Journal of Microbiology and Biotechnology
    • /
    • 제33권9호
    • /
    • pp.1149-1161
    • /
    • 2023
  • Changes in the gut microbiome cause recolonization by pathogens and inflammatory responses, leading to the development of intestinal disorders. Probiotics administration has been proposed for many years to reverse the intestinal dysbiosis and to enhance intestinal health. This study aimed to evaluate the inhibitory effects of two newly designed probiotic mixtures, Consti-Biome and Sensi-Biome, on two enteric pathogens Staphylococcus aureus and Escherichia coli that may cause intestinal disorders. Additionally, the study was designed to evaluate whether Consti-Biome and Sensi-Biome could modulate the immune response, produce short-chain fatty acids (SCFAs), and reduce gas production. Consti-Biome and Sensi-Biome showed superior adhesion ratios to HT-29 cells and competitively suppressed pathogen adhesion. Moreover, the probiotic mixtures decreased the levels of pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-6 and IL-1β. Cell-free supernatants (CFSs) were used to investigate the inhibitory effects of metabolites on growth and biofilms of pathogens. Consti-Biome and Sensi-Biome CFSs exhibited antimicrobial and anti-biofilm activity, where microscopic analysis confirmed an increase in the number of dead cells and the structural disruption of pathogens. Gas chromatographic analysis of the CFSs revealed their ability to produce SCFAs, including acetic, propionic, and butyric acid. SCFA secretion by probiotics may demonstrate their potential activities against pathogens and gut inflammation. In terms of intestinal symptoms regarding abdominal bloating and discomfort, Consti-Biome and Sensi-Biome also inhibited gas production. Thus, these two probiotic mixtures have great potential to be developed as dietary supplements to alleviate the intestinal disorders.

Amelioration of colitis progression by ginseng-derived exosome-like nanoparticles through suppression of inflammatory cytokines

  • Jisu Kim;Shuya Zhang ;Ying Zhu;Ruirui Wang;Jianxin Wang
    • Journal of Ginseng Research
    • /
    • 제47권5호
    • /
    • pp.627-637
    • /
    • 2023
  • Background: Damage to the healthy intestinal epithelial layer and regulation of the intestinal immune system, closely interrelated, are considered pivotal parts of the curative treatment for inflammatory bowel disease (IBD). Plant-based diets and phytochemicals can support the immune microenvironment in the intestinal epithelial barrier for a balanced immune system by improving the intestinal microecological balance and may have therapeutic potential in colitis. However, there have been only a few reports on the therapeutic potential of plant-derived exosome-like nanoparticles (PENs) and the underlying mechanism in colitis. This study aimed to assess the therapeutic effect of PENs from Panax ginseng, ginseng-derived exosome-like nanoparticles (GENs), in a mouse model of IBD, with a focus on the intestinal immune microenvironment. Method: To evaluate the anti-inflammatory effect of GENs on acute colitis, we treated GENs in Caco2 and lipopolysaccharide (LPS) -induced RAW 264.7 macrophages and analyzed the gene expression of proinflammatory cytokines and anti-inflammatory cytokines such as TNF-α, IL-6, and IL-10 by real-time PCR (RT-PCR). Furthermore, we further examined bacterial DNA from feces and determined the alteration of gut microbiota composition in DSS-induced colitis mice after administration of GENs through 16S rRNA gene sequencing analysis. Result: GENs with low toxicity showed a long-lasting intestinal retention effect for 48 h, which could lead to effective suppression of pro-inflammatory cytokines such as TNF-α and IL-6 production through inhibition of NF-κB in DSS-induced colitis. As a result, it showed longer colon length and suppressed thickening of the colon wall in the mice treated with GENs. Due to the amelioration of the progression of DSS-induced colitis with GENs treatment, the prolonged survival rate was observed for 17 days compared to 9 days in the PBS-treated group. In the gut microbiota analysis, the ratio of Firmicutes/Bacteroidota was decreased, which means GENs have therapeutic effectiveness against IBD. Ingesting GENs would be expected to slow colitis progression, strengthen the gut microbiota, and maintain gut homeostasis by preventing bacterial dysbiosis. Conclusion: GENs have a therapeutic effect on colitis through modulation of the intestinal microbiota and immune microenvironment. GENs not only ameliorate the inflammation in the damaged intestine by downregulating pro-inflammatory cytokines but also help balance the microbiota on the intestinal barrier and thereby improve the digestive system.

Failure of Fecal Microbiota Transplantation in a Three-Year-Old Child with Severe Refractory Ulcerative Colitis

  • Kumagai, Hideki;Yokoyama, Koji;Imagawa, Tomoyuki;Inoue, Shun;Tulyeu, Janyerkye;Tanaka, Mamoru;Yamagata, Takanori
    • Pediatric Gastroenterology, Hepatology & Nutrition
    • /
    • 제19권3호
    • /
    • pp.214-220
    • /
    • 2016
  • Fecal microbiota transplantation (FMT) is a treatment designed to correct gut dysbiosis by administration of feces from a healthy volunteer. It is still unclear whether FMT for children with ulcerative colitis (UC) is effective or hazardous. Here we describe a young patient to have received FMT for UC. A three-year-old girl was admitted to our hospital with severe active UC, and treated with aminosalicylates and various immunosuppressive drugs. As remission was not achieved, we decided to try FMT before colectomy. We administered donor fecal material a total of six times by retention enema (${\times}2$) and via a nasoduodenal tube (${\times}4$) within 10 days. The patient developed abdominal pain and pyrexia after each FMT session. Analyses revealed the transferred donor fecal microbiota had not been retained by the patient, who ultimately underwent colectomy. The severity of the UC and/or timing of FMT may have partly accounted for the poor outcome.

In Vitro Synergistic Antibacterial and Anti-Inflammatory Effects of Nisin and Lactic Acid in Yogurt against Helicobacter pylori and Human Gastric Cells

  • Seo Gu Han;Hyuk Cheol Kwon;Do Hyun Kim;Seong Joon Hong;Sung Gu Han
    • 한국축산식품학회지
    • /
    • 제43권5호
    • /
    • pp.751-766
    • /
    • 2023
  • Helicobacter pylori is a bacterium that naturally thrives in acidic environments and has the potential to induce various gastrointestinal disorders in humans. The antibiotic therapy utilized for treating H. pylori can lead to undesired side effects, such as dysbiosis in the gut microbiota. The objective of our study was to explore the potential antibacterial effects of nisin and lactic acid (LA) in yogurt against H. pylori. Additionally, we investigated the anti-inflammatory effects of nisin and LA in human gastric (AGS) cells infected with H. pylori. Nisin and LA combination showed the strongest inhibitory activity, with confirmed synergy at 0.375 fractional inhibitory concentration index. Also, post-fermented yogurt with incorporation of nisin exhibited antibacterial effect against H. pylori. The combination of nisin and LA resulted in a significant reduction of mRNA levels of bacterial toxins of H. pylori and pro-inflammatory cytokines in AGS cells infected with H. pylori. Furthermore, this also increased bacterial membrane damage, which led to DNA and protein leakage in H. pylori. Overall, the combination of nisin and LA shows promise as an alternative therapy for H. pylori infection. Additionally, the incorporation of nisin into foods containing LA presents a potential application. Further studies, including animal research, are needed to validate these findings and explore clinical applications.

Effects of natural mono- and di-saccharide as alternative sweeteners on inflammatory bowel disease: a narrative review

  • Eunju Kim
    • 대한지역사회영양학회지
    • /
    • 제28권3호
    • /
    • pp.181-191
    • /
    • 2023
  • Objectives: The incidence of inflammatory bowel disease (IBD) is increasing globally, and excessive added sugar consumption has been identified as one of the contributing factors. In the context of IBD, it is essential to explore functional sweeteners that can improve metabolic health and minimize the risk of IBD-related symptoms. This review article aims to shed light on the effects of natural mono- and di-saccharides as alternative sweeteners, specifically focusing on potential benefits for IBD. Methods: A comprehensive literature review was performed using PubMed and Google Scholar databases with articles published after the year 2000. The search terms 'IBD', 'added sugar', 'sweeteners', 'mono-saccharide', and 'di-saccharide' were combined to retrieve relevant articles. A total of 21 manuscripts, aligning with the objectives of the study, were selected. Papers focusing on artificial or high-intensity sweeteners were excluded to ensure relevant literature selection. Results: Multiple studies have emphasized the association between the high consumption of added sugars such as simple sugars and the increased risk of developing IBD. This is suggested to be attributed to the induction of pro-inflammatory cytokine productions and dysbiosis of the gut microbiota. Consequently, there is a growing demand for safe and functional sweeteners, in particular mono- and di-saccharides, that can serve as alternatives for IBD patients. Those functional sweeteners regulate inflammation, oxidative stress, and Intestinal barrier protection, and restore microbiome profiles in various IBD models including cells, animals, and humans. Conclusions: Understanding these mechanisms resolves the link between how sugar consumption and IBD, and highlights the beneficial effects of natural alternative sweeteners on IBD when they were administered by itself or as a replacement for simple sugar. Further, exploration of this relationship leads us to recognize the necessity of natural alternative sweeteners in dietary planning. This knowledge could potentially lead to more effective dietary strategies for individuals with IBD.