Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 2.065
1.
Food Res Int ; 186: 114322, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729712

Lactobacillus delbrueckii subsp. lactis CIDCA 133 is a health-promoting bacterium that can alleviate gut inflammation and improve the epithelial barrier in a mouse model of mucositis. Despite these beneficial effects, the protective potential of this strain in other inflammation models, such as inflammatory bowel disease, remains unexplored. Herein, we examined for the first time the efficacy of Lactobacillus delbrueckii CIDCA 133 incorporated into a fermented milk formulation in the recovery of inflammation, epithelial damage, and restoration of gut microbiota in mice with dextran sulfate sodium-induced colitis. Oral administration of Lactobacillus delbrueckii CIDCA 133 fermented milk relieved colitis by decreasing levels of inflammatory factors (myeloperoxidase, N-acetyl-ß-D-glucosaminidase, toll-like receptor 2, nuclear factor-κB, interleukins 10 and 6, and tumor necrosis factor), secretory immunoglobulin A levels, and intestinal paracellular permeability. This immunobiotic also modulated the expression of tight junction proteins (zonulin and occludin) and the activation of short-chain fatty acids-related receptors (G-protein coupled receptors 43 and 109A). Colonic protection was effectively associated with acetate production and restoration of gut microbiota composition. Treatment with Lactobacillus delbrueckii CIDCA 133 fermented milk increased the abundance of Firmicutes members (Lactobacillus genus) while decreasing the abundance of Proteobacteria (Helicobacter genus) and Bacteroidetes members (Bacteroides genus). These promising outcomes influenced the mice's mucosal healing, colon length, body weight, and disease activity index, demonstrating that this immunobiotic could be explored as an alternative approach for managing inflammatory bowel disease.


Colitis , Cultured Milk Products , Dextran Sulfate , Gastrointestinal Microbiome , Lactobacillus delbrueckii , Animals , Gastrointestinal Microbiome/drug effects , Colitis/microbiology , Colitis/chemically induced , Colitis/metabolism , Colitis/drug therapy , Lactobacillus delbrueckii/metabolism , Cultured Milk Products/microbiology , Mice , Probiotics/therapeutic use , Male , Mice, Inbred C57BL , Disease Models, Animal , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Inflammation , Colon/microbiology , Colon/metabolism , Lactobacillus
2.
Nat Commun ; 15(1): 3784, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710716

Probiotic and engineered microbe-based therapeutics are an emerging class of pharmaceutical agents. They represent a promising strategy for treating various chronic and inflammatory conditions by interacting with the host immune system and/or delivering therapeutic molecules. Here, we engineered a targeted probiotic yeast platform wherein Saccharomyces boulardii is designed to bind to abundant extracellular matrix proteins found within inflammatory lesions of the gastrointestinal tract through tunable antibody surface display. This approach enabled an additional 24-48 h of probiotic gut residence time compared to controls and 100-fold increased probiotic concentrations within the colon in preclinical models of ulcerative colitis in female mice. As a result, pharmacodynamic parameters including colon length, colonic cytokine expression profiles, and histological inflammation scores were robustly improved and restored back to healthy levels. Overall, these studies highlight the potential for targeted microbial therapeutics as a potential oral dosage form for the treatment of inflammatory bowel diseases.


Colitis, Ulcerative , Colon , Disease Models, Animal , Extracellular Matrix , Probiotics , Saccharomyces boulardii , Animals , Probiotics/administration & dosage , Female , Mice , Extracellular Matrix/metabolism , Colitis, Ulcerative/therapy , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/pathology , Colon/microbiology , Colon/metabolism , Colon/pathology , Mice, Inbred C57BL , Colitis/therapy , Colitis/microbiology , Colitis/pathology , Cytokines/metabolism , Humans
3.
Theranostics ; 14(7): 2719-2735, 2024.
Article En | MEDLINE | ID: mdl-38773969

Aim: To elucidate dynamics and functions in colonic macrophage subsets, and their regulation by Bifidobacterium breve (B. breve) and its associated metabolites in the initiation of colitis-associated colorectal cancer (CAC). Methods: Azoxymethane (AOM) and dextran sodium sulfate (DSS) were used to create a CAC model. The tumor-suppressive effect of B. breve and variations of macrophage subsets were evaluated. Intestinal macrophages were ablated to determine their role in the protective effects of B. breve. Efficacious molecules produced by B. breve were identified by non-targeted and targeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis. The molecular mechanism was further verified in murine bone marrow-derived macrophages (BMDMs), macrophages derived from human peripheral blood mononuclear cells (hPBMCs), and demonstrated in CAC mice. Results: B. breve alleviated colitis symptoms, delayed colonic tumorigenesis, and promoted phenotypic differentiation of immature inflammatory macrophages into mature homeostatic macrophages. On the contrary, the ablation of intestinal macrophages largely annulled the protective effects of B. breve. Microbial analysis of colonic contents revealed the enrichment of probiotics and the depletion of potential pathogens following B. breve supplementation. Moreover, indole-3-lactic acid (ILA) was positively correlated with B. breve in CAC mice and highly enriched in the culture supernatant of B. breve. Also, the addition of ILA directly promoted AKT phosphorylation and restricted the pro-inflammatory response of murine BMDMs and macrophages derived from hPBMCs in vitro. The effects of ILA in murine BMDMs and macrophages derived from hPBMCs were abolished by the aryl hydrocarbon receptor (AhR) antagonist CH-223191 or the AKT inhibitor MK-2206. Furthermore, ILA could protect against tumorigenesis by regulating macrophage differentiation in CAC mice; the AhR antagonist largely abrogated the effects of B. breve and ILA in relieving colitis and tumorigenesis. Conclusion: B. breve-mediated tryptophan metabolism ameliorates the precancerous inflammatory intestinal milieu to inhibit tumorigenesis by directing the differentiation of immature colonic macrophages.


Bifidobacterium breve , Cell Differentiation , Colitis , Indoles , Macrophages , Probiotics , Animals , Mice , Macrophages/metabolism , Macrophages/drug effects , Bifidobacterium breve/metabolism , Indoles/pharmacology , Indoles/metabolism , Humans , Colitis/chemically induced , Colitis/microbiology , Colitis/complications , Cell Differentiation/drug effects , Probiotics/pharmacology , Probiotics/administration & dosage , Disease Models, Animal , Carcinogenesis/drug effects , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/microbiology , Colitis-Associated Neoplasms/metabolism , Mice, Inbred C57BL , Colon/microbiology , Colon/pathology , Colon/metabolism , Dextran Sulfate , Male , Gastrointestinal Microbiome , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Azoxymethane
4.
J Clin Invest ; 134(9)2024 May 01.
Article En | MEDLINE | ID: mdl-38690730

The gut microbiota is an integral part of the human metaorganism that is required to shape physiologic host immune responses including host defense against pathogens. Disease-associated gut dysbiosis has been characterized by blooms of pathobionts, which are bacterial species that can drive disease under certain conditions. Pathobionts like Enterobacteriaceae often bloom during flares of inflammatory bowel disease (IBD) and are causally linked with IBD in murine models. In this issue of the JCI, Hecht and colleagues investigated how simple carbohydrates are causally linked to the bloom of the gut pathobiont Klebsiella pneumoniae, which belong to the Enterobacteriaceae family. Notably, the presence of fiber reduced the dissemination of K. pneumoniae into the blood and liver in a colitis model. Their findings provide a diet-related mechanism for gut dysbiosis, which has implications in the management of IBD and other conditions in which gut dysbiosis is an underlying factor.


Dysbiosis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Klebsiella pneumoniae , Humans , Animals , Inflammatory Bowel Diseases/microbiology , Mice , Dietary Carbohydrates/adverse effects , Klebsiella Infections , Colitis/chemically induced , Colitis/microbiology , Dietary Fiber
5.
Bull Exp Biol Med ; 176(5): 543-547, 2024 Mar.
Article En | MEDLINE | ID: mdl-38717566

We studied the dynamics of the main hemodynamic parameters in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) rats with visceral obesity and chemically induced colitis (CIC) against the background of probiotic therapy. Systolic BP, HR, and body temperature were recorded over 36 days using a wireless telemetry system. During 8 days (3 days before CIC induction and until the end of the experiment) the animals were intragastrically administered a probiotic based on Lactobacillus delbrueckii D5 strain. At baseline, systolic BP was significantly higher in the SHR group, while HR and body temperature did not differ in SHR and WKY rats. On day 8 after CIC induction, systolic BP, HR, and body temperature in SHR were significantly increased in comparison with the initial values. In the group of WKY rats, all indices at the end of the experiment remained at the initial levels. Probiotic therapy in SHR, in contrast to WKY rats, did not lead to normalization of body temperature and hemodynamic disorders resulting from CIC.


Body Temperature , Colitis , Hemodynamics , Probiotics , Rats, Inbred SHR , Rats, Inbred WKY , Animals , Probiotics/pharmacology , Probiotics/administration & dosage , Rats , Male , Colitis/chemically induced , Colitis/physiopathology , Colitis/microbiology , Hemodynamics/drug effects , Body Temperature/drug effects , Blood Pressure/drug effects , Blood Pressure/physiology , Heart Rate/drug effects , Lactobacillus delbrueckii , Obesity/physiopathology , Obesity, Abdominal/physiopathology , Obesity, Abdominal/chemically induced
6.
Nutrients ; 16(9)2024 Apr 25.
Article En | MEDLINE | ID: mdl-38732527

Ulcerative colitis (UC) is characterized by chronic inflammation and ulceration of the intestinal inner lining, resulting in various symptoms. Sea buckthorn berries contain a bioactive compound known as sea buckthorn polysaccharide (SBP). However, the precise mechanisms underlying the impact of SBP on UC remain unclear. In this study, we investigated the effects of pretreatment with SBP on colitis induced by DSS. Our findings demonstrate that SBP pretreatment effectively reduces inflammation, oxidative stress, and intestinal barrier damage associated with colitis. To further elucidate the role of SBP-modulated gut microbiota in UC, we performed fecal microbiota transplantation (FMT) on DSS-treated mice. The microbiota from SBP-treated mice exhibits notable anti-inflammatory and antioxidant effects, improves colonic barrier integrity, and increases the abundance of beneficial bacteria, as well as enhancing SCFA production. Collectively, these results strongly indicate that SBP-mediated amelioration of colitis is attributed to its impact on the gut microbiota, particularly through the promotion of SCFA-producing bacteria and subsequent elevation of SCFA levels. This study provides compelling evidence supporting the efficacy of pre-emptive SBP supplementation in alleviating colitis symptoms by modulating the gut microbiota, thereby offering novel insights into the potential of SBP as a regulator of the gut microbiota for colitis relief.


Gastrointestinal Microbiome , Hippophae , Polysaccharides , Animals , Hippophae/chemistry , Polysaccharides/pharmacology , Gastrointestinal Microbiome/drug effects , Mice , Colitis/drug therapy , Colitis/chemically induced , Colitis/microbiology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/drug therapy , Disease Models, Animal , Male , Mice, Inbred C57BL , Oxidative Stress/drug effects , Fecal Microbiota Transplantation , Colon/drug effects , Colon/microbiology , Colon/metabolism , Dextran Sulfate , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Fruit/chemistry , Fatty Acids, Volatile/metabolism
7.
J Int Med Res ; 52(5): 3000605241241000, 2024 May.
Article En | MEDLINE | ID: mdl-38749910

Ileostomy diverts the flow of feces, which can result in malnutrition in the distal part of the intestine. The diversity of the gut microbiota consequently decreases, ultimately leading to intestinal dysbiosis and dysfunction. This condition can readily result in diversion colitis (DC). Potential treatment strategies include interventions targeting the gut microbiota. In this case study, we effectively treated a patient with severe DC by ileostomy and allogeneic fecal microbiota transplantation (FMT). A 69-year-old man presented with a perforated malignant tumor in the descending colon and an iliac abscess. He underwent laparoscopic radical sigmoid colon tumor resection and prophylactic ileostomy. Follow-up colonoscopy 3 months postoperatively revealed diffuse intestinal mucosal congestion and edema along with granular inflammatory follicular hyperplasia, leading to a diagnosis of severe DC. After two rounds of allogeneic FMT, both the intestinal mucosal bleeding and edema significantly improved, as did the diversity of the gut microbiota. The positive outcome of allogeneic FMT in this case highlights the potential advantages that this procedure can offer patients with DC. However, few studies have focused on allogeneic FMT, and more in-depth research is needed to gain a better understanding.


Colitis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Ileostomy , Humans , Male , Aged , Fecal Microbiota Transplantation/methods , Colitis/microbiology , Colitis/therapy , Transplantation, Homologous/methods , Treatment Outcome , Colonoscopy
8.
Sci Rep ; 14(1): 11560, 2024 05 21.
Article En | MEDLINE | ID: mdl-38773299

IBD is a disorder which could be caused by oxidative stress. This investigation aims to determine if probiotics and postbiotics can control oxidative stress and inflammation and compare the effectiveness of these two probiotic and postbiotic mixtures of substances. 88 strains of Lactobacillus and Bifidobacterium were tested for antioxidant activity. Male wild-type C57BL/6 mice were divided into four experimental groups, namely high fat diet (HFD) + PBS, HFD + DSS, HFD + DSS + 109 cfu/ml of probiotics, and HFD + DSS + 109 cfu/ml of postbiotics. The phenotypical indices and pathological scores were assessed. The expression of genes related to NF-kB and Nrf2 signaling pathways and enzymes associated with oxidant/anti-oxidant activities, and proinflammatory/inflammatory cytokines were assessed. In contrast to the groups exposed to DSS, mice treated with probiotics mixture and postbiotics mixture alongside DSS displayed alleviation of DSS-induced adverse effects on phenotypical characteristics, as well as molecular indices such as the Nrf2 and NF-kB related genes, with a greater emphasis on the postbiotics component. In accordance with the findings of the present investigation, it can be inferred that even in using a high-fat dietary regimen as an inducer of oxidative stress, the emergence of inflammation can be effectively addressed through the utilization of probiotics and, more specifically, postbiotics.


Anti-Inflammatory Agents , Antioxidants , Colitis , Dextran Sulfate , Disease Models, Animal , Mice, Inbred C57BL , NF-E2-Related Factor 2 , NF-kappa B , Oxidative Stress , Probiotics , Signal Transduction , Animals , Probiotics/pharmacology , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Antioxidants/metabolism , Antioxidants/pharmacology , Male , Mice , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Anti-Inflammatory Agents/pharmacology , Signal Transduction/drug effects , Oxidative Stress/drug effects , Lactobacillus , Bifidobacterium , Diet, High-Fat/adverse effects
9.
Gut Microbes ; 16(1): 2356642, 2024.
Article En | MEDLINE | ID: mdl-38769708

Adherent-invasive Escherichia coli (AIEC) strain LF82, isolated from patients with Crohn's disease, invades gut epithelial cells, and replicates in macrophages contributing to chronic inflammation. In this study, we found that RstAB contributing to the colonization of LF82 in a mouse model of chronic colitis by promoting bacterial replication in macrophages. By comparing the transcriptomes of rstAB mutant- and wild-type when infected macrophages, 83 significant differentially expressed genes in LF82 were identified. And we identified two possible RstA target genes (csgD and asr) among the differentially expressed genes. The electrophoretic mobility shift assay and quantitative real-time PCR confirmed that RstA binds to the promoters of csgD and asr and activates their expression. csgD deletion attenuated LF82 intracellular biofilm formation, and asr deletion reduced acid tolerance compared with the wild-type. Acidic pH was shown by quantitative real-time PCR to be the signal sensed by RstAB to activate the expression of csgD and asr. We uncovered a signal transduction pathway whereby LF82, in response to the acidic environment within macrophages, activates transcription of the csgD to promote biofilm formation, and activates transcription of the asr to promote acid tolerance, promoting its replication within macrophages and colonization of the intestine. This finding deepens our understanding of the LF82 replication regulation mechanism in macrophages and offers new perspectives for further studies on AIEC virulence mechanisms.


Bacterial Adhesion , Biofilms , Escherichia coli Infections , Escherichia coli Proteins , Escherichia coli , Gene Expression Regulation, Bacterial , Macrophages , Macrophages/microbiology , Animals , Mice , Escherichia coli/genetics , Escherichia coli/pathogenicity , Escherichia coli/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Biofilms/growth & development , Escherichia coli Infections/microbiology , Humans , Hydrogen-Ion Concentration , Virulence , Colitis/microbiology , Crohn Disease/microbiology , Disease Models, Animal , Signal Transduction , Acids/metabolism
10.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G659-G675, 2024 Jun 01.
Article En | MEDLINE | ID: mdl-38591132

Probiotic-containing fermented dairy foods have the potential to benefit human health, but the importance of the dairy matrix for efficacy remains unclear. We investigated the capacity of Lacticaseibacillus paracasei BL23 in phosphate-buffered saline (BL23-PBS), BL23-fermented milk (BL23-milk), and milk to modify intestinal and behavioral responses in a dextran sodium sulfate (DSS, 3% wt/vol) mouse model of colitis. Significant sex-dependent differences were found such that female mice exhibited more severe colitis, greater weight loss, and higher mortality rates. Sex differences were also found for ion transport ex vivo, colonic cytokine and tight junction gene expression, and fecal microbiota composition. Measurements of milk and BL23 effects showed BL23-PBS consumption improved weight recovery in females, whereas milk resulted in better body weight recovery in males. Occludin and Claudin-2 gene transcript levels indicated barrier function was impaired in males, but BL23-milk was still found to improve colonic ion transport in those mice. Proinflammatory and anti-inflammatory gene expression levels were increased in both male and female mice fed BL23, and to a more variable extent, milk, compared with controls. The female mouse fecal microbiota contained high proportions of Akkermansia (average of 18.1%) at baseline, and females exhibited more changes in gut microbiota composition following BL23 and milk intake. Male fecal microbiota harbored significantly more Parasutterella and less Blautia and Roseburia after DSS treatment, independent of BL23 or milk consumption. These findings show the complex interplay between dietary components and sex-dependent responses in mitigating inflammation in the digestive tract.NEW & NOTEWORTHY Sex-dependent responses to probiotic Lacticaseibacillus paracasei and milk and the potential of the dairy matrix to enhance probiotic protection against colitis in this context have not been previously explored. Female mice were more sensitive than males to colonic injury, and neither treatment effectively alleviated inflammation in both sexes. These sex-dependent responses may result from differences in the higher baseline proportions of Akkermansia in the gut microbiome of female mice.


Colitis , Dextran Sulfate , Disease Models, Animal , Milk , Probiotics , Animals , Female , Probiotics/pharmacology , Male , Colitis/microbiology , Colitis/chemically induced , Colitis/metabolism , Mice , Gastrointestinal Microbiome , Mice, Inbred C57BL , Colon/metabolism , Colon/microbiology , Sex Factors , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology
11.
Nutrients ; 16(8)2024 Apr 12.
Article En | MEDLINE | ID: mdl-38674829

The intestinal epithelial barrier can prevent the invasion of pathogenic microorganisms and food antigens to maintain a consistent intestinal homeostasis. However, an imbalance in this barrier can result in various diseases, such as inflammatory bowel disease, malnutrition, and metabolic disease. Thus, the aim of this study was to select probiotic strains with epithelial barrier-enhancing ability in cell-based model and further investigate them for their improving effects on colitis mouse and weaned piglet models. The results showed that selected specific cell-free fermentation supernatants (CFSs) from Ligilactobacillus salivarius P1, Lactobacillus gasseri P12, and Limosilactobacillus reuteri G7 promoted intestinal epithelial cell growth and proliferation, strengthening the intestinal barrier in an intestinal epithelial cell line Caco-2 model. Further, the administration of CFSs of L. salivarius P1, L. gasseri P12, and L. reuteri G7 were found to ameliorate DSS-induced colitis in mice. Additionally, spray-dried powders of CFS from the three strains were examined in a weaned piglet model, only CFS powder of L. reuteri G7 could ameliorate the feed/gain ratio and serum levels of D-lactate and endotoxin. In conclusion, a new potential probiotic strain, L. reuteri G7, was selected and showed ameliorating effects in both colitis mouse and weaned piglet models.


Colitis , Disease Models, Animal , Fermentation , Intestinal Mucosa , Limosilactobacillus reuteri , Probiotics , Weaning , Animals , Probiotics/pharmacology , Colitis/chemically induced , Colitis/metabolism , Colitis/microbiology , Humans , Mice , Swine , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Caco-2 Cells , Ligilactobacillus salivarius , Lactobacillus gasseri , Dextran Sulfate , Male , Cell Proliferation/drug effects
12.
Surg Clin North Am ; 104(3): 545-556, 2024 Jun.
Article En | MEDLINE | ID: mdl-38677819

Clostridioides difficile colitis is an important source of hospital-acquired diarrhea associated with antibiotic use. Symptoms are profuse watery diarrhea, typically following a course of antibiotics; however, some cases of fulminant disease may manifest with shock, ileus, or megacolon. Nonfulminant colitis is treated with oral fidaxomicin. C difficile colitis has a high potential for recurrence, and recurrent episodes are also treated with fidaxomicin. Bezlotoxumab is another medication that may be used in populations at high risk for further recurrence. Fulminant disease is treated with maximal medical therapy and early surgical consultation. Antibiotic stewardship is critical to preventing C difficile colitis.


Anti-Bacterial Agents , Clostridioides difficile , Clostridium Infections , Colitis , Humans , Clostridium Infections/diagnosis , Clostridium Infections/therapy , Anti-Bacterial Agents/therapeutic use , Colitis/microbiology , Colitis/diagnosis , Colitis/therapy , Fidaxomicin/therapeutic use
13.
J Agric Food Chem ; 72(18): 10355-10365, 2024 May 08.
Article En | MEDLINE | ID: mdl-38620073

The genus Bifidobacterium has been widely used in functional foods for health promotion due to its beneficial effects on human health, especially in the gastrointestinal tract (GIT). In this study, we characterize the anti-inflammatory potential of the probiotic strain Bifidobacterium pseudocatenulatum G7, isolated from a healthy male adult. G7 secretion inhibited inflammatory response in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Moreover, oral administration of bacteria G7 alleviated the severity of colonic inflammation in dextran sulfate sodium (DSS)-treated colitis mice, which was evidenced by a decreased disease activity index (DAI) and enhanced structural integrity of the colon. The 16S rRNA gene sequencing result illustrated that the G7 alleviated DSS-induced gut microbiota dysbiosis, accompanied by the modulated bile acids and short-chain fatty acid (SCFA) levels. Overall, our results demonstrated the potential anti-inflammatory effects of Bifidobacterium pseudocatenulatum G7 on both in vitro and in vivo models, which provided a solid foundation for further development of a novel anti-inflammatory probiotic.


Anti-Inflammatory Agents , Bifidobacterium pseudocatenulatum , Colitis , Gastrointestinal Microbiome , Probiotics , Probiotics/administration & dosage , Probiotics/pharmacology , Mice , Animals , RAW 264.7 Cells , Male , Anti-Inflammatory Agents/administration & dosage , Humans , Colitis/microbiology , Colitis/therapy , Colitis/chemically induced , Bifidobacterium pseudocatenulatum/genetics , Bifidobacterium pseudocatenulatum/chemistry , Mice, Inbred C57BL , Macrophages/immunology , Fatty Acids, Volatile/metabolism , Colon/microbiology , Colon/immunology
14.
Appl Environ Microbiol ; 90(5): e0004624, 2024 May 21.
Article En | MEDLINE | ID: mdl-38563787

Dietary fiber metabolism by gut microorganisms plays important roles in host physiology and health. Alginate, the major dietary fiber of daily diet seaweeds, is drawing more attention because of multiple biological activities. To advance the understanding of alginate assimilation mechanism in the gut, we show the presence of unsaturated alginate oligosaccharides (uAOS)-specific alginate utilization loci (AUL) in human gut microbiome. As a representative example, a working model of the AUL from the gut microorganism Bacteroides clarus was reconstructed from biochemistry and transcriptome data. The fermentation of resulting monosaccharides through Entner-Doudoroff pathway tunes the metabolism of short-chain fatty acids and amino acids. Furthermore, we show that uAOS feeding protects the mice against dextran sulfate sodium-induced acute colitis probably by remodeling gut microbiota and metabolome. IMPORTANCE: Alginate has been included in traditional Chinese medicine and daily diet for centuries. Recently discovered biological activities suggested that alginate-derived alginate oligosaccharides (AOS) might be an active ingredient in traditional Chinese medicine, but how these AOS are metabolized in the gut and how it affects health need more information. The study on the working mechanism of alginate utilization loci (AUL) by the gut microorganism uncovers the role of unsaturated alginate oligosaccharides (uAOS) assimilation in tuning short-chain fatty acids and amino acids metabolism and demonstrates that uAOS metabolism by gut microorganisms results in a variation of cell metabolites, which potentially contributes to the physiology and health of gut.


Alginates , Gastrointestinal Microbiome , Oligosaccharides , Alginates/metabolism , Oligosaccharides/metabolism , Mice , Animals , Humans , Colitis/microbiology , Colitis/chemically induced , Mice, Inbred C57BL , Fatty Acids, Volatile/metabolism , Inflammation/metabolism , Dextran Sulfate , Dietary Fiber/metabolism
15.
Eur J Pharmacol ; 974: 176611, 2024 Jul 05.
Article En | MEDLINE | ID: mdl-38663540

Inflammatory bowel disease (IBD) is a chronic and relapsing inflammatory disorder of the gastrointestinal tract for which treatment options remain limited. In this study, we used a dual-luciferase-based screening of an FDA-approved drug library, identifying Bazedoxifene (BZA) as an inhibitor of the NF-κB pathway. We further investigated its therapeutic effects in a dextran sodium sulfate (DSS)-induced colitis model and explored its impact on gut microbiota regulation and the underlying molecular mechanisms. Our results showed that BZA significantly reduced DSS-induced colitis symptoms in mice, evidenced by decreased colon length shortening, lower histological scores, and increased expression of intestinal mucosal barrier-associated proteins, such as Claudin 1, Occludin, Zo-1, Mucin 2 (Muc2), and E-cadherin. Used independently, BZA showed therapeutic effects comparable to those of infliximab (IFX). In addition, BZA modulated the abundance of gut microbiota especially Bifidobacterium pseudolongum, and influenced microbial metabolite production. Crucially, BZA's alleviation of DSS-induced colitis in mice was linked to change in gut microbiota composition, as evidenced by in vivo gut microbiota depletion and fecal microbiota transplantation (FMT) mice model. Molecularly, BZA inhibited STAT3 and NF-κB activation in DSS-induced colitis in mice. In general, BZA significantly reduced DSS-induced colitis in mice through modulating the gut microbiota and inhibiting STAT3 and NF-κB activation, and its independent use demonstrated a therapeutic potential comparable to IFX. This study highlights gut microbiota's role in IBD drug development, offering insights for BZA's future development and its clinical applications.


Colitis , Dextran Sulfate , Gastrointestinal Microbiome , NF-kappa B , STAT3 Transcription Factor , Signal Transduction , Animals , NF-kappa B/metabolism , STAT3 Transcription Factor/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis/microbiology , Colitis/pathology , Gastrointestinal Microbiome/drug effects , Mice , Signal Transduction/drug effects , Indoles/pharmacology , Indoles/therapeutic use , Mice, Inbred C57BL , Disease Models, Animal , Colon/drug effects , Colon/pathology , Colon/metabolism , Colon/microbiology , Male , Humans
16.
Redox Biol ; 72: 103140, 2024 Jun.
Article En | MEDLINE | ID: mdl-38593629

Gut microbiota has been implicated in the initiation and progression of various diseases; however, the underlying mechanisms remain elusive and effective therapeutic strategies are scarce. In this study, we investigated the role and mechanisms of gut microbiota in TNBS-induced colitis and its associated kidney injury while evaluating the potential of dietary protein as a therapeutic intervention. The intrarectal administration of TNBS induced colitis in mice, concurrently with kidney damage. Interestingly, this effect was absent when TNBS was administered intraperitoneally, indicating a potential role of gut microbiota. Depletion of gut bacteria with antibiotics significantly attenuated the severity of TNBS-induced inflammation, oxidative damage, and tissue injury in the colon and kidneys. Mechanistic investigations using cultured colon epithelial cells and bone-marrow macrophages unveiled that TNBS induced cell oxidation, inflammation and injury, which was amplified by the bacterial component LPS and mitigated by thiol antioxidants. Importantly, in vivo administration of thiol-rich whey protein entirely prevented TNBS-induced colonic and kidney injury. Our findings suggest that gut bacteria significantly contribute to the initiation and progression of colitis and associated kidney injury, potentially through mechanisms involving LPS-induced exaggeration of oxidative cellular damage. Furthermore, our research highlights the potential of dietary thiol antioxidants as preventive and therapeutic interventions.


Colitis , Gastrointestinal Microbiome , Oxidative Stress , Trinitrobenzenesulfonic Acid , Animals , Gastrointestinal Microbiome/drug effects , Oxidative Stress/drug effects , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Mice , Trinitrobenzenesulfonic Acid/toxicity , Trinitrobenzenesulfonic Acid/adverse effects , Disease Models, Animal , Male , Antioxidants/pharmacology , Kidney/metabolism , Kidney/pathology , Kidney/drug effects
17.
Biomed Pharmacother ; 174: 116514, 2024 May.
Article En | MEDLINE | ID: mdl-38574618

Plant-derived nanovesicles (PDNVs) have recently emerged as natural delivery systems of biofunctional compounds toward mammalian cells. Considering their already described composition, anti-inflammatory properties, stability, and low toxicity, PDNVs offer a promising path for developing new preventive strategies for several inflammatory diseases, among which the inflammatory bowel disease (IBD). In this study, we explore the protective effects of industrially produced lemon vesicles (iLNVs) in a rat model of IBD. Characterization of iLNVs reveals the presence of small particles less than 200 nm in size and a profile of bioactive compounds enriched in flavonoids and organic acids with known beneficial properties. In vitro studies on human macrophages confirm the safety and anti-inflammatory effects of iLNVs, as evidenced by the reduced expression of pro-inflammatory cytokines and increased levels of anti-inflammatory markers. As evidenced by in vivo experiments, pre-treatment with iLNVs significantly alleviates symptoms and histological features in 2,4 dinitrobenzensulfuric acid (DNBS)-induced colitis in rats. Molecular pathway analysis reveals modulation of NF-κB and Nrf2, indicating anti-inflammatory and antioxidant effects. Finally, iLNVs affects gut microbiota composition, improving the consistent colitis-related alterations. Overall, we demonstrated the protective role of industrially produced lemon nanovesicles against colitis and emphasized their potential in managing IBD through multifaceted mechanisms.


Anti-Inflammatory Agents , Antioxidants , Citrus , Colitis , Gastrointestinal Microbiome , Animals , Anti-Inflammatory Agents/pharmacology , Citrus/chemistry , Colitis/pathology , Colitis/drug therapy , Colitis/chemically induced , Colitis/microbiology , Colitis/metabolism , Male , Antioxidants/pharmacology , Rats , Humans , Gastrointestinal Microbiome/drug effects , Nanoparticles/chemistry , Rats, Wistar , Disease Models, Animal , Cytokines/metabolism , NF-kappa B/metabolism
18.
Microbiome ; 12(1): 76, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38649950

BACKGROUND: The etiology of inflammatory bowel disease (IBD) is unclear but involves both genetics and environmental factors, including the gut microbiota. Indeed, exacerbated activation of the gastrointestinal immune system toward the gut microbiota occurs in genetically susceptible hosts and under the influence of the environment. For instance, a majority of IBD susceptibility loci lie within genes involved in immune responses, such as caspase recruitment domain member 9 (Card9). However, the relative impacts of genotype versus microbiota on colitis susceptibility in the context of CARD9 deficiency remain unknown. RESULTS: Card9 gene directly contributes to recovery from dextran sodium sulfate (DSS)-induced colitis by inducing the colonic expression of the cytokine IL-22 and the antimicrobial peptides Reg3ß and Reg3γ independently of the microbiota. On the other hand, Card9 is required for regulating the microbiota capacity to produce AhR ligands, which leads to the production of IL-22 in the colon, promoting recovery after colitis. In addition, cross-fostering experiments showed that 5 weeks after weaning, the microbiota transmitted from the nursing mother before weaning had a stronger impact on the tryptophan metabolism of the pups than the pups' own genotype. CONCLUSIONS: These results show the role of CARD9 and its effector IL-22 in mediating recovery from DSS-induced colitis in both microbiota-independent and microbiota-dependent manners. Card9 genotype modulates the microbiota metabolic capacity to produce AhR ligands, but this effect can be overridden by the implantation of a WT or "healthy" microbiota before weaning. It highlights the importance of the weaning reaction occurring between the immune system and microbiota for host metabolism and immune functions throughout life. A better understanding of the impact of genetics on microbiota metabolism is key to developing efficient therapeutic strategies for patients suffering from complex inflammatory disorders. Video Abstract.


CARD Signaling Adaptor Proteins , Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Interleukin-22 , Interleukins , Pancreatitis-Associated Proteins , Animals , CARD Signaling Adaptor Proteins/genetics , Colitis/microbiology , Colitis/genetics , Colitis/immunology , Mice , Pancreatitis-Associated Proteins/genetics , Interleukins/genetics , Interleukins/metabolism , Mice, Knockout , Genetic Predisposition to Disease , Disease Models, Animal , Mice, Inbred C57BL , Colon/microbiology , Colon/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/immunology , Female , Male
19.
J Food Sci ; 89(5): 3078-3093, 2024 May.
Article En | MEDLINE | ID: mdl-38605580

Human milk contains a variety of microorganisms that exert benefit for human health. In the current study, we isolated a novel Lactobacillus gasseri strain named Lactobacillus gasseri (L. gasseri) SHMB 0001 from human milk and aimed to evaluate the probiotic characteristics and protective effects on murine colitis of the strain. The results showed that L. gasseri SHMB 0001 possessed promising potential probiotic characteristics, including good tolerance against artificial gastric and intestinal fluids, adhesion to Caco-2 cells, susceptibility to antibiotic, no hemolytic activity, and without signs of toxicity or infection in mice. Administration of L. gasseri SHMB 0001 (1 × 108 CFU per gram of mouse weight per day) reduced weight loss, the disease activity index, and colon shortening in mice during murine colitis conditions. Histopathological analysis revealed that L. gasseri SHMB 0001 treatment attenuated epithelial damage and inflammatory infiltration in the colon. L. gasseri SHMB 0001 treatment increased the expression of colonic occludin and claudin-1 while decreasing the expression of pro-inflammatory cytokine genes. L. gasseri SHMB 0001 modified the composition and structure of the gut microbiota community and partially recovered the Clusters of Orthologous Groups (COG) and Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways altered by dextran sulfate sodium (DSS). Overall, our results indicated that the human breast milk-derived L. gasseri SHMB 0001 exhibited promising probiotic properties and ameliorative effect on DSS-induced colitis in mice. L. gasseri SHMB 0001 may be applied as a promising probiotic against intestinal inflammation in the future. PRACTICAL APPLICATION: L. gasseri SHMB 0001 isolated from human breast milk showed good tolerance to gastrointestinal environment, safety, and protective effect against DSS-induced mice colitis via enforcing gut barrier, downregulating pro-inflammatory cytokines, and modulating gut microbiota. L. gasseri SHMB 0001 may be a promising probiotic candidate for the treatment of intestinal inflammation.


Colitis , Dextran Sulfate , Gastrointestinal Microbiome , Lactobacillus gasseri , Milk, Human , Probiotics , Probiotics/pharmacology , Animals , Humans , Mice , Colitis/chemically induced , Colitis/therapy , Colitis/microbiology , Dextran Sulfate/adverse effects , Gastrointestinal Microbiome/drug effects , Caco-2 Cells , Female , Colon/microbiology , Colon/pathology , Colon/metabolism , Cytokines/metabolism , Disease Models, Animal
20.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article En | MEDLINE | ID: mdl-38673843

Neutrophil-myeloperoxidase (MPO) is a heme-containing peroxidase which produces excess amounts of hypochlorous acid during inflammation. While pharmacological MPO inhibition mitigates all indices of experimental colitis, no studies have corroborated the role of MPO using knockout (KO) models. Therefore, we investigated MPO deficient mice in a murine model of colitis. Wild type (Wt) and MPO-deficient mice were treated with dextran sodium sulphate (DSS) in a chronic model of experimental colitis with three acute cycles of DSS-induced colitis over 63 days, emulating IBD relapse and remission cycles. Mice were immunologically profiled at the gut muscoa and the faecal microbiome was assessed via 16S rRNA amplicon sequencing. Contrary to previous pharmacological antagonist studies targeting MPO, MPO-deficient mice showed no protection from experimental colitis during cyclical DSS-challenge. We are the first to report drastic faecal microbiota shifts in MPO-deficient mice, showing a significantly different microbiome profile on Day 1 of treatment, with a similar shift and distinction on Day 29 (half-way point), via qualitative and quantitative descriptions of phylogenetic distances. Herein, we provide the first evidence of substantial microbiome shifts in MPO-deficiency, which may influence disease progression. Our findings have significant implications for the utility of MPO-KO mice in investigating disease models.


Colitis , Dextran Sulfate , Disease Models, Animal , Gastrointestinal Microbiome , Mice, Knockout , Peroxidase , Animals , Peroxidase/metabolism , Peroxidase/genetics , Mice , Colitis/microbiology , Colitis/chemically induced , Colitis/genetics , Feces/microbiology , Gene Deletion , RNA, Ribosomal, 16S/genetics , Mice, Inbred C57BL
...