• 제목/요약/키워드: gut inflammation

검색결과 99건 처리시간 0.022초

Protective Effect of Lactobacillus fermentum LA12 in an Alcohol-Induced Rat Model of Alcoholic Steatohepatitis

  • Kim, Byoung-Kook;Lee, In-Ock;Tan, Pei-Lei;Eor, Ju-Young;Hwang, Jae-Kwan;Kim, Sae-Hun
    • 한국축산식품학회지
    • /
    • 제37권6호
    • /
    • pp.931-939
    • /
    • 2017
  • Alcoholic liver disease (ALD) is a complex multifaceted disease that involves oxidative stress and inflammation as the key mediators. Despite decades of intensive research, there are no FDA-approved therapies, and/or no effective cure is yet available. Probiotics have received increasing attention in the past few years due to their well-documented gastrointestinal health-promoting effects. Interestingly, emerging studies have suggested that certain probiotics may offer benefits beyond the gut. Lactobacillus fermentum LA12 has been previously demonstrated to play a role in inflammatory-related disease. However, the possible protective effect of L. fermentum LA12 on ALD still remain to be explored. Thus, the aim of this study was to evaluate the possible protective effect of L. fermentum LA12 on alcohol-induced gut barrier dysfunction and liver damage in a rat model of alcoholic steatohepatitis (ASH). Daily oral administration of L. fermentum LA12 in rat model of ASH for four weeks was shown to significantly reduced intestinal nitric oxide production and hyperpermeability. Moreover, small intestinal histological- and qRT-PCR analysis further revealed that L. fermentum LA12 treatment was capable of up-regulating the mRNA expression levels of tight junction proteins, thereby stimulating the restitution of barrier structure and function. Serum and hepatic analyses also revealed that the restoration of epithelial barrier function may prevent the leakage of endotoxin into the blood, subsequently improve liver function and hepatic steatosis in the L. fermentum LA12-treated rats. Altogether, results in this study suggest that L. fermentum LA12 may be used as a dietary adjunct for the prevention and treatment of ASH.

From Gut to Brain: Alteration in Inflammation Markers in the Brain of Dextran Sodium Sulfate-induced Colitis Model Mice

  • Do, Jongho;Woo, Jungmin
    • Clinical Psychopharmacology and Neuroscience
    • /
    • 제16권4호
    • /
    • pp.422-433
    • /
    • 2018
  • Objective: Neuropsychiatric manifestations like depression and cognitive dysfunction commonly occur in inflammatory bowel disease (IBD). In the context of the brain-gut axis model, colitis can lead to alteration of brain function in a bottom-up manner. Here, the changes in the response of the hypothalamic-pituitary-adrenal axis and inflammation-related markers in the brain in colitis were studied. Methods: Dextran sodium sulfate (DSS) was used to generate a mouse model of colitis. Mice were treated with DSS for 3 or 7 days and sacrificed. We analyzed the gene expression of brain-derived neurotrophic factor (BDNF), cyclooxygenase 2 (COX-2), and glial fibrillary acidic protein (GFAP), and the expression of GFAP, in the hippocampus, hypothalamus, and amygdala. Additionally, the levels of C-reactive protein (CRP) and serum cortisol/corticosterone were measured. Results: Alteration of inflammatory-related markers varied depending on the brain region and exposure time. In the hippocampus, COX-2 mRNA, GFAP mRNA, and GFAP expression were upregulated during exposure to DSS. However, in the hypothalamus, COX-2 mRNA was upregulated only 3 days after treatment. In the amygdala, BDNF and COX-2 mRNAs were downregulated. CRP and corticosterone expression increased with DSS treatment at day 7. Conclusion: IBD could lead to neuroinflammation in a bottom-up manner, and this effect varied according to brain region. Stress-related hormones and serum inflammatory markers, such as CRP, were upregulated from the third day of DSS treatment. Therefore, early and active intervention is required to prevent psychological and behavioral changes caused by IBD, and region-specific studies can help understand the precise mechanisms by which IBD affects the brain.

Development of a Novel ATP Bioluminescence Assay Based on Engineered Probiotic Saccharomyces boulardii Expressing Firefly Luciferase

  • Ji Sun Park;Young-Woo Kim;Hyungdong Kim;Sun-Ki Kim;Kyeongsoon Park
    • Journal of Microbiology and Biotechnology
    • /
    • 제33권11호
    • /
    • pp.1506-1512
    • /
    • 2023
  • Quantitative analysis of adenosine triphosphate (ATP) has been widely used as a diagnostic tool in the food and medical industries. Particularly, the pathogenesis of a few diseases including inflammatory bowel disease (IBD) is closely related to high ATP concentrations. A bioluminescent D-luciferin/luciferase system, which includes a luciferase (FLuc) from the firefly Photinus pyralis as a key component, is the most commonly used method for the detection and quantification of ATP. Here, instead of isolating FLuc produced in recombinant Escherichia coli, we aimed to develop a whole-cell biocatalyst system that does not require extraction and purification of FLuc. To this end, the gene coding for FLuc was introduced into the genome of probiotic Saccharomyces boulardii using the CRISPR/Cas9-based genome editing system. The linear relationship (r2 = 0.9561) between ATP levels and bioluminescence generated from the engineered S. boulardii expressing FLuc was observed in vitro. To explore the feasibility of using the engineered S. boulardii expressing FLuc as a whole-cell biosensor to detect inflammation biomarker (i.e., ATP) in the gut, a colitis mouse model was established using dextran sodium sulfate as a colitogenic compound. Our findings demonstrated that the whole-cell biosensor can detect elevated ATP levels during gut inflammation in mice. Therefore, the simple and powerful method developed herein could be applied for non-invasive IBD diagnosis.

Pear pomace alleviated atopic dermatitis in NC/Nga mice and inhibited LPS-induced inflammation in RAW 264.7 macrophages

  • You, Mikyoung;Wang, Ziyun;Kim, Hwa-Jin;Lee, Young-Hyun;Kim, Hyeon-A
    • Nutrition Research and Practice
    • /
    • 제16권5호
    • /
    • pp.577-588
    • /
    • 2022
  • BACKGROUND/OBJECTIVES: Poorly regulated inflammation is believed to be the most predominant factor that can result in a wide scope of diseases including atopic dermatitis (AD). Despite many studies on the effect of pear pomace in obesity-related disorders including dysregulated gut microbiota, the protective effect of pear pomace in AD is still unknown. This study aimed to evaluate the effect of pear pomace ethanol extract (PPE) on AD by inhibiting inflammation. MATERIALS/METHODS: In the in vivo experiment, 2, 4-dinitrochlorobenzene (DNCB) was applied to NC/Nga mice to induce AD-like skin lesions. After the induction, PPE was administered daily by oral gavage for 4 weeks. The clinical severity score, serum IgE levels, spleen weight, histological changes in dorsal skin, and inflammation-related proteins were measured. In the cell study, RAW 264.7 cells were pretreated with PPE before stimulation with lipopolysaccharide (LPS). Nitrite oxide (NO) production and nuclear factor kappa B (NF-𝛋B) protein expression were detected. RESULTS: Compared to the AD control (AD-C) group, IgE levels were dramatically decreased via PPE treatment. PPE significantly reduced scratching behavior, improved skin symptoms, and decreased ear thickness compared to the AD-C group. In addition, PPE inhibited the DNCB-induced expression of inducible nitrite oxide synthase (iNOS), the receptor for advanced glycation end products, extracellular signal-regulated kinase (ERK) 1/2, and NF-𝛋B. PPE inhibited the LPS-induced overproduction of NO and the enhanced expression of iNOS and cyclooxygenase-2. Moreover, the phosphorylation of ERK1/2 and NF-𝛋B in RAW 264.7 cells was suppressed by PPE. CONCLUSIONS: These results suggest that PPE could be explored as a therapeutic agent to prevent AD.

Zerumbone Restores Gut Microbiota Composition in ETBF Colonized AOM/DSS Mice

  • Cho, Hye-Won;Rhee, Ki-Jong;Eom, Yong-Bin
    • Journal of Microbiology and Biotechnology
    • /
    • 제30권11호
    • /
    • pp.1640-1650
    • /
    • 2020
  • Colorectal cancer (CRC) is the leading cause of common malignant neoplasm worldwide. Many studies have analyzed compositions of gut microbiota associated with various diseases such as inflammatory bowel diseases (IBD) and colon cancer. One of the most representative bacteria involved in CRC is enterotoxigenic Bacteroides fragilis (ETBF), a species belonging to phylum Bacteroidetes. We used ETBF colonized mice with azoxymethane (AOM)/dextran sulphate sodium (DSS) and zerumbone, a compound with anti-bacterial effect, to determine whether zerumbone could restore intestinal microbiota composition. Four experimental groups of mice were used: sham, ETBF colonized AOM/DSS group, ETBF colonized AOM/DSS group zerumbone 60 mg kg-1 (ETBF/AOM/DSS + Z (60)), and only zerumbone (60 mg kg-1)-treated group. We performed reversible dye terminators-based analysis of 16S rRNA gene region V3-V4 for group comparison. Microbiota compositions of ETBF/AOM/DSS + Z (60) group and ETBF colonized AOM/DSS group not given zerumbone were significantly different. There were more Bacteroides in ETBF/AOM/DSS + Z (60) group than those in ETBF colonized AOM/DSS group, suggesting that B. fragilis could be a normal flora activated by zerumbone. In addition, based on linear discriminant analysis of effect size (LEfSe) analysis, microbial diversity decreased significantly in the ETBF colonized AOM/DSS group. However, after given zerumbone, the taxonomic relative abundance was increased. These findings suggest that zerumbone not only influenced the microbial diversity and richness, but also could be helpful for enhancing the balance of gut microbial composition. In this work, we demonstrate that zerumbone could restore the composition of intestinal microbiota.

Effect of Probiotics Lactobacillus and Bifidobacterium on Gut-Derived Lipopolysaccharides and Inflammatory Cytokines: An In Vitro Study Using a Human Colonic Microbiota Model

  • Rodes, Laetitia;Khan, Afshan;Paul, Arghya;Coussa-Charley, Michael;Marinescu, Daniel;Tomaro-Duchesneau, Catherine;Shao, Wei;Kahouli, Imen;Prakash, Satya
    • Journal of Microbiology and Biotechnology
    • /
    • 제23권4호
    • /
    • pp.518-526
    • /
    • 2013
  • Gut-derived lipopolysaccharides (LPS) are critical to the development and progression of chronic low-grade inflammation and metabolic diseases. In this study, the effects of probiotics Lactobacillus and Bifidobacterium on gut-derived lipopolysaccharide and inflammatory cytokine concentrations were evaluated using a human colonic microbiota model. Lactobacillus reuteri, L. rhamnosus, L. plantarum, Bifidobacterium animalis, B. bifidum, B. longum, and B. longum subsp. infantis were identified from the literature for their anti-inflammatory potential. Each bacterial culture was administered daily to a human colonic microbiota model during 14 days. Colonic lipopolysaccharides, and Gram-positive and negative bacteria were quantified. RAW 264.7 macrophage cells were stimulated with supernatant from the human colonic microbiota model. Concentrations of TNF-${\alpha}$, IL-$1{\beta}$, and IL-4 cytokines were measured. Lipopolysaccharide concentrations were significantly reduced with the administration of B. bifidum ($-46.45{\pm}5.65%$), L. rhamnosus ($-30.40{\pm}5.08%$), B. longum ($-42.50{\pm}1.28%$), and B. longum subsp. infantis ($-68.85{\pm}5.32%$) (p < 0.05). Cell counts of Gram-negative and positive bacteria were distinctly affected by the probiotic administered. There was a probiotic strain-specific effect on immunomodulatory responses of RAW 264.7 macrophage cells. B. longum subsp. infantis demonstrated higher capacities to reduce TNF-${\alpha}$ concentrations ($-69.41{\pm}2.78%$; p < 0.05) and to increase IL-4 concentrations ($+16.50{\pm}0.59%$; p < 0.05). Colonic lipopolysaccharides were significantly correlated with TNF-${\alpha}$ and IL-$1{\beta}$ concentrations (p < 0.05). These findings suggest that specific probiotic bacteria, such as B. longum subsp. infantis, might decrease colonic lipopolysaccharide concentrations, which might reduce the proinflammatory tone. This study has noteworthy applications in the field of biotherapeutics for the prevention and/or treatment of inflammatory and metabolic diseases.

NQO1-Knockout Mice Are Highly Sensitive to Clostridium Difficile Toxin A-Induced Enteritis

  • Nam, Seung Taek;Hwang, Jung Hwan;Kim, Dae Hong;Lu, Li Fang;Hong, Ji;Zhang, Peng;Yoon, I Na;Hwang, Jae Sam;Chung, Hyo Kyun;Shong, Minho;Lee, Chul-Ho;Kim, Ho
    • Journal of Microbiology and Biotechnology
    • /
    • 제26권8호
    • /
    • pp.1446-1451
    • /
    • 2016
  • Clostridium difficile toxin A causes acute gut inflammation in animals and humans. It is known to downregulate the tight junctions between colonic epithelial cells, allowing luminal contents to access body tissues and trigger acute immune responses. However, it is not yet known whether this loss of the barrier function is a critical factor in the progression of toxin A-induced pseudomembranous colitis. We previously showed that NADH:quinone oxidoreductase 1 (NQO1) KO (knockout) mice spontaneously display weak gut inflammation and a marked loss of colonic epithelial tight junctions. Moreover, NQO1 KO mice exhibited highly increased inflammatory responses compared with NQO1 WT (wild-type) control mice when subjected to DSS-induced experimental colitis. Here, we tested whether toxin A could also trigger more severe inflammatory responses in NQO1 KO mice compared with NQO1 WT mice. Indeed, our results show that C. difficile toxin A-mediated enteritis is significantly enhanced in NQO1 KO mice compared with NQO1 WT mice. The levels of fluid secretion, villus disruption, and epithelial cell apoptosis were also higher in toxin A-treated NQO1 KO mice compared with WT mice. The previous and present results collectively show that NQO1 is involved in the formation of tight junctions in the small intestine, and that defects in NQO1 enhance C. difficile toxin A-induced acute inflammatory responses, presumably via the loss of epithelial cell tight junctions.

장내미생물과 인지기능은 서로 연관되어 있는가? (Does the Gut Microbiota Regulate a Cognitive Function?)

  • 최정현;진윤호;김주헌;홍용근
    • 생명과학회지
    • /
    • 제29권6호
    • /
    • pp.747-753
    • /
    • 2019
  • 인지기능 저하는 장 단기 기억 및 주의력 소실과 우울증, 불안증의 증가를 특징으로 한다. 또한, 인지기능 저하는 알츠하이머, 파킨슨병과 같은 다양한 퇴행성 뇌질환과 연관되어 있다. 경제적 부담, 안전 위협을 포함하는 인지기능 저하와 관련된 사회적 문제는 고령화가 진행됨에 따라 증가하고 있다. 이러한 문제를 해결하기 위해 전 세계적으로 많은 연구가 수행되고 있다. 일반적으로 인지기능 저하를 유발할 가능성이 있는 원인으로는 노화에 따른 대사 및 호르몬 불균형, 감염, 약물 오남용, 신경세포 손상 등이 알려져 있지만 다양한 요인이 관련되어 있으므로 원인 규명이 어려운 한계점 때문에 뚜렷한 치료전략 수립이 어려운 실정이다. 최근의 연구에 따르면 퇴행성 뇌질환 발생의 원인과 이에 대한 치료전략 수립에 있어서 장내미생물의 역할이 중요하게 제시되고 있다. 특히, 알츠하이머병과 파킨슨병에서 장내미생물 조성의 변화 및 이들에 의한 대사산물에 따른 분자생물학적, 신경행동학적 증상의 변화가 밝혀졌다. 알츠하이머병 동물모델에서 장내미생물의 변화는 NMDA 수용체와 글루탐산의 변화를 통해 기억능력 소실을 야기하였다. 반면, 알츠하이머병 동물모델에 프로바이오틱스를 투여하였을 때, 비정상적인 신경학적 행동이 유의적으로 감소하였다. 파킨슨병은 장내미생물 군집의 변화와 직접적인 연관성을 보였으며 이는 이차적 증상인 변비 발생에도 영향을 미치는 것으로 나타났다. 파킨슨병 동물모델에 투여한 프로바이오틱스는 단쇄지방산 중 하나인 뷰티르산 증가를 통한 신경세포 보호효과를 나타내었다. 또한, 알츠하이머병과 파킨슨병에서 뇌-혈관장벽의 기능이상이 밝혀졌으며, 뇌-혈관장벽 변화는 장내미생물 불균형에 의한 전신성 염증에 따른 미세소관의 파괴 및 투과성 증가와 연관된 것으로 나타났다. 더불어 장내미생물 대사과정에서 생성된 대사산물은 퇴행성 뇌질환의 발생과 치료에 영향을 미친다. 본 논문에서는 인지기능 저하의 진행을 지연시킴으로써 심화를 방지할 수 있는 효과적인 접근법을 제시하기 위하여 인지기능 저하와 장내미생물의 연관성을 심층적으로 고찰하여 치료적 대안으로 제시하고자 한다.

Expression of Toll-like Receptors, Pro-, and Anti-inflammatory Cytokines in Relation to Gut Microbiota in Irritable Bowel Syndrome: The Evidence for Its Micro-organic Basis

  • Shukla, Ratnakar;Ghoshal, Ujjala;Ranjan, Prabhat;Ghoshal, Uday C
    • Journal of Neurogastroenterology and Motility
    • /
    • 제24권4호
    • /
    • pp.628-642
    • /
    • 2018
  • Background/Aims A Subset of patients with irritable bowel syndrome (IBS) may have mild inflammation due to immune activation. Toll-like receptors (TLRs) and cytokines may cause intestinal inflammation. We studied their expression in relation to gut microbiota. Methods Expression of TLRs and cytokines was assessed in 47 IBS patients (Rome III) and 25 controls using quantitative real-time polymerase chain reaction. Immunohistochemistry was further performed to confirm the expression of TLR-4 and TLR-5. Results Of 47 patients with IBS, 20 had constipation (IBS-C), 20 diarrhea (IBS-D), and 7 unclassified (IBS-U). The mRNA levels of TLR-4 and TLR-5 were up-regulated in IBS patients than controls (P = 0.013 and P < 0.001, respectively). Expression of TLR-4 and TLR-5 at protein level was 4.2-folds and 6.6-folds higher in IBS-D than controls. The mRNA levels of IL-6 (P = 0.003), C-X-C motif chemokine ligand 11 (CXCL-11) (P < 0.001) and C-X-C motif chemokine receptor 3 (CXCR-3) (P < 0.001) were higher among IBS patients than controls. Expression of IL-6 (P = 0.002), CXCL-11 (P < 0.001), and CXCR-3 (P < 0.001) were up-regulated and IL-10 (P = 0.012) was down-regulated in IBS-D patients than controls. Positive correlation was seen between TLR-4 and IL-6 (P = 0.043), CXCR-3, and CXCL-11 (P = 0.047), and IL-6 and CXCR-3 (P = 0.003). Stool frequency per week showed positive correlation with mRNA levels of TLR-4 (P = 0.016) and CXCR-3 (P = 0.005), but inversely correlated with IL-10 (P = 0.002). Copy number of Lactobacillus (P = 0.045) and Bifidobacterium (P = 0.011) showed correlation with IL-10 in IBS-C, while Gram-positive (P = 0.031) and Gram-negative bacteria (P = 0.010) showed correlation with CXCL-11 in IBS-D patients. Conclusions Altered immune activation in response to dysbiotic microbiota may promote intestinal inflammation in a subset of patients with IBS.

Autophagy and Digestive Disorders: Advances in Understanding and Therapeutic Approaches

  • Thein, Wynn;Po, Wah Wah;Choi, Won Seok;Sohn, Uy Dong
    • Biomolecules & Therapeutics
    • /
    • 제29권4호
    • /
    • pp.353-364
    • /
    • 2021
  • The gastrointestinal (GI) tract is a series of hollow organs that is responsible for the digestion and absorption of ingested foods and the excretion of waste. Any changes in the GI tract can lead to GI disorders. GI disorders are highly prevalent in the population and account for substantial morbidity, mortality, and healthcare utilization. GI disorders can be functional, or organic with structural changes. Functional GI disorders include functional dyspepsia and irritable bowel syndrome. Organic GI disorders include inflammation of the GI tract due to chronic infection, drugs, trauma, and other causes. Recent studies have highlighted a new explanatory mechanism for GI disorders. It has been suggested that autophagy, an intracellular homeostatic mechanism, also plays an important role in the pathogenesis of GI disorders. Autophagy has three primary forms: macroautophagy, microautophagy, and chaperone-mediated autophagy. It may affect intestinal homeostasis, host defense against intestinal pathogens, regulation of the gut microbiota, and innate and adaptive immunity. Drugs targeting autophagy could, therefore, have therapeutic potential for treating GI disorders. In this review, we provide an overview of current understanding regarding the evidence for autophagy in GI diseases and updates on potential treatments, including drugs and complementary and alternative medicines.