Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.951
Filter
1.
Nutrients ; 16(13)2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38999840

ABSTRACT

Inflammatory bowel diseases (IBDs), such as Crohn's disease (CD) and ulcerative colitis (UC), are chronic diseases of the digestive system with a multifactorial and not fully understood etiology. There is research suggesting that they may be initiated by genetic, immunological, and lifestyle factors. In turn, all of these factors play an important role in the modulation of intestinal microflora, and a significant proportion of IBD patients struggle with intestinal dysbiosis, which leads to the conclusion that intestinal microflora disorders may significantly increase the risk of developing IBD. Additionally, in IBD patients, Toll-like receptors (TLRs) produced by intestinal epithelial cells and dendritic cells treat intestinal bacterial antigens as pathogens, which causes a disruption of the immune response, resulting in the development of an inflammatory process. This may result in the occurrence of intestinal dysbiosis, which IBD patients are significantly vulnerable to. In this study, we reviewed scientific studies (in particular, systematic reviews with meta-analyses, being studies with the highest level of evidence) regarding the microflora of patients with IBD vs. the microflora in healthy people, and the use of various strains in IBD therapy.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/immunology , Crohn Disease/microbiology , Crohn Disease/immunology , Probiotics/therapeutic use , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/immunology
2.
Nat Commun ; 15(1): 5778, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987259

ABSTRACT

Antimicrobial proteins contribute to host-microbiota interactions and are associated with inflammatory bowel disease (IBD), but our understanding on antimicrobial protein diversity and functions remains incomplete. Ribonuclease 4 (Rnase4) is a potential antimicrobial protein with no known function in the intestines. Here we find that RNASE4 is expressed in intestinal epithelial cells (IEC) including Paneth and goblet cells, and is detectable in human and mouse stool. Results from Rnase4-deficient mice and recombinant protein suggest that Rnase4 kills Parasutterella to modulate intestinal microbiome, thereby enhancing indoleamine-2,3-dioxygenase 1 (IDO1) expression and subsequently kynurenic and xanthurenic acid production in IECs to reduce colitis susceptibility. Furthermore, deceased RNASE4 levels are observed in the intestinal tissues and stool from patients with IBD, correlating with increased stool Parasutterella. Our results thus implicate Rnase4 as an intestinal antimicrobial protein regulating gut microbiota and metabolite homeostasis, and as a potential diagnostic biomarker and therapeutic target for IBD.


Subject(s)
Gastrointestinal Microbiome , Homeostasis , Inflammatory Bowel Diseases , Mice, Inbred C57BL , Gastrointestinal Microbiome/physiology , Animals , Humans , Mice , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Colitis/microbiology , Colitis/metabolism , Colitis/chemically induced , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Knockout , Ribonucleases/metabolism , Male , Feces/microbiology , Female , Intestines/microbiology , Antimicrobial Peptides/metabolism
3.
BMC Microbiol ; 24(1): 233, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951788

ABSTRACT

BACKGROUND: Inflammatory Bowel Diseases (IBD) are a major public health issue with unclear aetiology. Changes in the composition and functionality of the intestinal microbiota are associated with these pathologies, including the depletion of strict anaerobes such as Feacalibacterium prausnitzii. Less evidence is observed for depletion in other anaerobes, among which bifidobacteria. This study characterized the taxonomic and functional diversity of bifidobacteria isolated from the human intestinal microbiota in active and non-active IBD patients by a culturomics approach and evaluated if these bifidobacteria might be used as probiotics for gut health. RESULTS: A total of 341 bifidobacteria were isolated from the intestinal microbiota of IBD patients (52 Crohn's disease and 26 ulcerative colitis patients), with a high proportion of Bifidobacterium dentium strains (28% of isolated bifidobacteria). In ulcerative colitis, the major species identified was B. dentium (39% of isolated bifidobacteria), in active and non-active ulcerative colitis. In Crohn's disease, B. adolescentis was the major species isolated from non-active patients (40%), while similar amounts of B. dentium and B. adolescentis were found in active Crohn's disease patients. The relative abundance of B. dentium was increased with age, both in Crohn's disease and ulcerative colitis and active and non-active IBD patients. Antibacterial capacities of bifidobacteria isolated from non-active ulcerative colitis against Escherichia coli LF82 and Salmonella enterica ATCC 14028 were observed more often compared to strains isolated from active ulcerative colitis. Finally, B. longum were retained as strains with the highest probiotic potential as they were the major strains presenting exopolysaccharide synthesis, antibacterial activity, and anti-inflammatory capacities. Antimicrobial activity and EPS synthesis were further correlated to the presence of antimicrobial and EPS gene clusters by in silico analysis. CONCLUSIONS: Different bifidobacterial taxonomic profiles were identified in the microbiota of IBD patients. The most abundant species were B. dentium, mainly associated to the microbiota of ulcerative colitis patients and B. adolescentis, in the intestinal microbiota of Crohn's disease patients. Additionally, the relative abundance of B. dentium significantly increased with age. Furthermore, this study evidenced that bifidobacteria with probiotic potential (antipathogenic activity, exopolysaccharide production and anti-inflammatory activity), especially B. longum strains, can be isolated from the intestinal microbiota of both active and non-active Crohn's disease and ulcerative colitis patients.


Subject(s)
Bifidobacterium , Gastrointestinal Microbiome , Probiotics , Humans , Bifidobacterium/isolation & purification , Bifidobacterium/classification , Bifidobacterium/genetics , Adult , Female , Male , Middle Aged , Inflammatory Bowel Diseases/microbiology , Young Adult , Aged , Colitis, Ulcerative/microbiology , Crohn Disease/microbiology , Phylogeny , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Phenotype , Adolescent , Anti-Bacterial Agents/pharmacology
4.
BMC Pediatr ; 24(1): 417, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951792

ABSTRACT

BACKGROUND: The relationship between Helicobacter-pylori(Hp)infection and inflammatory-bowel-disease(IBD) in pediatric-patients remains controversial. We aimed to assess the Hp-infection occurrence in newly-diagnosed pediatric-patients with IBD compared to no-IBD patients. Additionally, we aimed to examine differences in clinical-activity-index(CAI) and endoscopic-severity-score(ESS)between IBD-patients with and without Hp-infection, at baseline and at 1-year-follow-up(FU), after eradication-therapy(ET). METHODS: IBD diagnosis was based on Porto-criteria, and all patients underwent gastroscopy at baseline and 1-year FU. For Crohn's-disease(CD) and ulcerative colitis(UC), IBD-CAI and -ESS were classified using PCDAI/SES-CD and PUCAI/UCEIS, respectively. RESULTS: 76 IBD-patients were included in the study[35 F(46.1%),median-age 12(range 2-17)]. CD and UC were diagnosed in 29(38.2%) and 45(59.2%)patients, respectively, and unclassified-IBD in two(2.6%)patients. Non-IBD patients were 148[71 F(48.0%),median-age 12(range 1-17)]. Hp-infection at baseline was reported in 7(9.2%) and 18(12.2%)IBD and non-IBD patients, respectively(p = 0.5065). The 7 IBD patients with Hp infection were compared to 69 IBD patients without Hp-infection at baseline evaluation, and no significant differences were reported considering CAI and ESS in these two groups. At 1-year FU, after ET, IBD patients with Hp infection improved, both for CAI and ESS, but statistical significance was not reached. CONCLUSION: The occurrence of Hp-infection did not differ between IBD and no-IBD patients. No differences in CAI or ESS were observed at the diagnosis, and after ET no worsening of CAI or ESS was noted at one-year FU, between Hp-positive and -negative IBD patients.


Subject(s)
Colitis, Ulcerative , Helicobacter Infections , Helicobacter pylori , Humans , Helicobacter Infections/complications , Helicobacter Infections/diagnosis , Child , Male , Female , Adolescent , Prospective Studies , Child, Preschool , Colitis, Ulcerative/complications , Colitis, Ulcerative/microbiology , Crohn Disease/complications , Crohn Disease/microbiology , Severity of Illness Index , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/microbiology , Gastroscopy , Follow-Up Studies , Anti-Bacterial Agents/therapeutic use
5.
Gut Microbes ; 16(1): 2379633, 2024.
Article in English | MEDLINE | ID: mdl-39024479

ABSTRACT

Gut microbiota-derived extracellular vesicles (mEVs) are reported to regulate inflammatory response by delivering bacterial products into host cells. The complement receptor of the immunoglobulin superfamily macrophages (CRIg+ Mφ) could clear invading bacteria and their derivatives. Here, we investigate the role of CRIg+ Mφ and the mechanism by which mEVs regulate intestinal inflammation. We found that it is exacerbated in IBD patients and colitis mice by mEVs' leakage from disturbed gut microbiota, enriching microbial DNA in the intestinal mucosa. CRIg+ Mφ significantly decrease in IBD patients, allowing the spread of mEVs into the mucosa. The microbial DNA within mEVs is the key trigger for inflammation and barrier function damage. The cGAS/STING pathway is crucial in mEVs-mediated inflammatory injury. Blocking cGAS/STING signaling effectively alleviates inflammation caused by mEVs leakage and CRIg+ Mφ deficiency. Microbial DNA-containing mEVs, along with CRIg+ Mφ deficiency, stimulate inflammation in IBD, with the cGAS/STING pathway playing a crucial role.


Subject(s)
DNA, Bacterial , Extracellular Vesicles , Gastrointestinal Microbiome , Inflammation , Inflammatory Bowel Diseases , Intestinal Mucosa , Macrophages , Membrane Proteins , Nucleotidyltransferases , Extracellular Vesicles/metabolism , Extracellular Vesicles/immunology , Animals , Mice , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/immunology , Humans , Membrane Proteins/metabolism , Membrane Proteins/genetics , Nucleotidyltransferases/metabolism , Nucleotidyltransferases/genetics , Intestinal Mucosa/microbiology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Mucosa/immunology , Inflammation/microbiology , Inflammation/metabolism , DNA, Bacterial/genetics , Mice, Inbred C57BL , Male , Female , Signal Transduction , Colitis/microbiology , Colitis/pathology
6.
Sci Rep ; 14(1): 16613, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026025

ABSTRACT

Investigating the gut microbiome and metabolome frequently requires faecal samples, which can be difficult to obtain. Previous studies have shown that rectal swabs are comparable to faecal samples for analysing gut microbiota composition and key metabolites. In this study, 3D printed rectal swabs were compared with conventional flocked swabs and faecal samples, due to the potential advantages 3D printing as a technique offers for swab production and development. 16S rRNA gene sequencing, qPCR and metabolite profiling (using 1H-NMR spectroscopy) were performed on swab and faecal samples from healthy participants. Faecal calprotectin and total protein analysis were performed on samples from inflammatory bowel disease (IBD) patients. There were no significant differences between both swab types and faecal samples when assessing key measures of alpha and beta diversity, and differences in the abundance of major phyla. There was a strong correlation between both swab types and faecal samples for all combined metabolites detected by NMR. In IBD patients, there was no significant difference in faecal calprotectin and total protein levels between both swab types and faecal samples. These data lead us to conclude that 3D printed swabs are equivalent to flocked swabs for the analysis of the gut microbiome, metabolome and inflammation.


Subject(s)
Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Metabolome , Printing, Three-Dimensional , RNA, Ribosomal, 16S , Humans , Feces/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , RNA, Ribosomal, 16S/genetics , Male , Female , Adult , Rectum/microbiology , Rectum/metabolism , Leukocyte L1 Antigen Complex/metabolism , Leukocyte L1 Antigen Complex/analysis , Inflammation/microbiology , Inflammation/metabolism , Middle Aged , Specimen Handling/methods
7.
Microbiome ; 12(1): 130, 2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39026313

ABSTRACT

BACKGROUND: The gut virome has been implicated in inflammatory bowel disease (IBD), yet a full understanding of the gut virome in IBD patients, especially across diverse geographic populations, is lacking. RESULTS: In this study, we conducted a comprehensive gut virome-wide association study in a Chinese cohort of 71 IBD patients (15 with Crohn's disease and 56 with ulcerative colitis) and 77 healthy controls via viral-like particle (VLP) and bulk virome sequencing of their feces. By utilizing an integrated gut virus catalog tailored to the IBD virome, we revealed fundamental alterations in the gut virome in IBD patients. These characterized 139 differentially abundant viral signatures, including elevated phages predicted to infect Escherichia, Klebsiella, Enterococcus_B, Streptococcus, and Veillonella species, as well as IBD-depleted phages targeting Prevotella, Ruminococcus_E, Bifidobacterium, and Blautia species. Remarkably, these viral signatures demonstrated high consistency across diverse populations such as those in Europe and the USA, emphasizing their significance and broad relevance in the disease context. Furthermore, fecal virome transplantation experiments verified that the colonization of these IBD-characterized viruses can modulate experimental colitis in mouse models. CONCLUSIONS: Building upon these insights into the IBD gut virome, we identified potential biomarkers for prognosis and therapy in IBD patients, laying the foundation for further exploration of viromes in related conditions. Video Abstract.


Subject(s)
Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Virome , Humans , Gastrointestinal Microbiome/genetics , Animals , Feces/virology , Feces/microbiology , Mice , Inflammatory Bowel Diseases/virology , Inflammatory Bowel Diseases/microbiology , Female , Male , Adult , Middle Aged , Crohn Disease/virology , Crohn Disease/microbiology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Colitis, Ulcerative/virology , Colitis, Ulcerative/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , China , Fecal Microbiota Transplantation , Case-Control Studies , Viruses/classification , Viruses/isolation & purification , Viruses/genetics
8.
Curr Opin Gastroenterol ; 40(4): 258-267, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38841848

ABSTRACT

PURPOSE OF REVIEW: The role of the microbiome and dysbiosis is increasingly recognized in the pathogenesis of inflammatory bowel disease (IBD). Intestinal microbiota transplant (IMT), previously termed fecal microbiota transplant has demonstrated efficacy in restoring a healthy microbiome and promoting gut health in recurrent Clostridioides difficile infection. Several randomized trials (RCTs) highlighted IMT's potential in treating ulcerative colitis, while smaller studies reported on its application in managing Crohn's disease and pouchitis. RECENT FINDINGS: This review delves into the current understanding of dysbiosis in IBD, highlighting the distinctions in the microbiota of patients with IBD compared to healthy controls. It explores the mechanisms by which IMT can restore a healthy microbiome and provides a focused analysis of recent RCTs using IMT for inducing and maintaining remission in IBD. Lastly, we discuss the current knowledge gaps that limit its widespread use. SUMMARY: The body of evidence supporting the use of IMT in IBD is growing. The lack of a standardized protocol impedes its application beyond clinical trials. Further research is needed to identify patient profile and disease phenotypes that benefit from IMT, to delineate key donor characteristics, optimize the delivery route, dosage, and frequency.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Dysbiosis/therapy , Dysbiosis/microbiology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Randomized Controlled Trials as Topic
9.
Biochem Biophys Res Commun ; 724: 150243, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38857558

ABSTRACT

Inflammatory Bowel Disease (IBD) is an enduring inflammatory disease of the gastrointestinal tract (GIT). The complexity of IBD, its profound impact on patient's quality of life, and its burden on healthcare systems necessitate continuing studies to elucidate its etiology, refine care strategies, improve treatment outcomes, and identify potential targets for novel therapeutic interventions. The discovery of a connection between IBD and gut bacterial quorum sensing (QS) molecules has opened exciting opportunities for research into IBD pathophysiology. QS molecules are small chemical messengers synthesized and released by bacteria based on population density. These chemicals are sensed not only by the microbial species but also by host cells and are essential in gut homeostasis. QS molecules are now known to interact with inflammatory pathways, therefore rendering them potential therapeutic targets for IBD management. Given these intriguing developments, the most recent research findings in this area are herein reviewed. First, the global burden of IBD and the disruptions of the gut microbiota and intestinal barrier associated with the disease are assessed. Next, the general QS mechanism and signaling molecules in the gut are discussed. Then, the roles of QS molecules and their connection with IBD are elucidated. Lastly, the review proposes potential QS-based therapeutic targets for IBD, offering insights into the future research trajectory in this field.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Quorum Sensing , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Humans , Animals , Bacteria/metabolism
10.
World J Gastroenterol ; 30(21): 2744-2747, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38899327

ABSTRACT

There is considerable controversy on the role of physical activity in irritable bowel disease (IBD) since published reports are conflicting. It is well known that there is known relapse with specific treatment in IBD. This, in addition to onset of extraintestinal symptoms creates a need to think of alternate approaches. In this context, the current article describes the need of a multi-institutional study with standard protocol of physical activity for documenting its effect on both the primary disease and the extra alimentary manifestations. This paper also points out the possibility of using adjuvant complementary medicine such as yoga, whose effects have been documented in other diseases like irritable bowel syndrome. A third approach could be to focus on the intestinal dysbiosis in IBD and concentrate on research on restoring the microbial flora to normal, to see whether the extra-intestinal symptoms are alleviated.


Subject(s)
Dysbiosis , Exercise , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Yoga , Humans , Exercise/physiology , Inflammatory Bowel Diseases/therapy , Inflammatory Bowel Diseases/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/physiopathology , Exercise Therapy/methods , Irritable Bowel Syndrome/physiopathology , Irritable Bowel Syndrome/therapy , Irritable Bowel Syndrome/microbiology , Treatment Outcome , Multicenter Studies as Topic
11.
Medicine (Baltimore) ; 103(25): e38551, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905376

ABSTRACT

This research investigates the causal relationships among gut microbiota, inflammatory proteins, and inflammatory bowel disease (IBD), including crohn disease (CD) and ulcerative colitis (UC), and identifies the role of inflammatory proteins as potential mediators. Our study analyzed gut microbiome data from 13,266 samples collected by the MiBioGen alliance, along with inflammatory protein data from recent research by Zhao et al, and genetic data on CD and UC from the International Inflammatory Bowel Disease Genetics Consortium (IIBDGC). We used Mendelian randomization (MR) to explore the associations, complemented by replication, meta-analysis, and multivariable MR techniques for enhanced accuracy and robustness. Our analysis employed several statistical methods, including inverse-variance weighting, MR-Egger, and the weighted median method, ensuring comprehensive and precise evaluation. After MR analysis, replication and meta-analysis, we revealed significant associations between 11 types of gut microbiota and 17 inflammatory proteins were associated with CD and UC. Mediator MR analysis and multivariable MR analysis showed that in CD, the CD40L receptor mediated the causal effect of Defluviitaleaceae UCG-011 on CD (mediation ratio 8.3%), and the Hepatocyte growth factor mediated the causal effect of Odoribacter on CD (mediation ratio 18%). In UC, the C-C motif chemokine 4 mediated the causal effect of Ruminococcus2 on UC (mediation ratio 4%). This research demonstrates the interactions between specific gut microbiota, inflammatory proteins, and CD and UC. Furthermore, the CD40L receptor may mediate the relationship between Defluviitaleaceae UCG-011 and CD; the Hepatocyte growth factor may mediate the relationship between Odoribacter and CD; and the C-C motif chemokine 4 may mediate the relationship between Ruminococcus2 and UC. The identified associations and mediation effects offer insights into potential therapeutic approaches targeting the gut microbiome for managing CD and UC.


Subject(s)
Gastrointestinal Microbiome , Mendelian Randomization Analysis , Humans , Gastrointestinal Microbiome/genetics , Crohn Disease/microbiology , Crohn Disease/genetics , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/genetics , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics
12.
Nutrients ; 16(11)2024 May 29.
Article in English | MEDLINE | ID: mdl-38892628

ABSTRACT

This comprehensive review delineates the extensive roles of Akkermansia muciniphila in various health domains, spanning from metabolic and inflammatory diseases to neurodegenerative disorders. A. muciniphila, known for its ability to reside in the mucous layer of the intestine, plays a pivotal role in maintaining gut integrity and interacting with host metabolic processes. Its influence extends to modulating immune responses and potentially easing symptoms across several non-communicable diseases, including obesity, diabetes, inflammatory bowel disease, and cancer. Recent studies highlight its capacity to interact with the gut-brain axis, suggesting a possible impact on neuropsychiatric conditions. Despite the promising therapeutic potential of A. muciniphila highlighted in animal and preliminary human studies, challenges remain in its practical application due to stability and cultivation issues. However, the development of pasteurized forms and synthetic mediums offers new avenues for its use in clinical settings, as recognized by regulatory bodies like the European Food Safety Authority. This narrative review serves as a crucial resource for understanding the broad implications of A. muciniphila across different health conditions and its potential integration into therapeutic strategies.


Subject(s)
Akkermansia , Gastrointestinal Microbiome , Noncommunicable Diseases , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Probiotics/therapeutic use , Animals , Noncommunicable Diseases/prevention & control , Noncommunicable Diseases/therapy , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Verrucomicrobia , Brain-Gut Axis/physiology , Obesity/microbiology , Obesity/therapy , Neoplasms/therapy , Neoplasms/microbiology , Diabetes Mellitus/therapy , Diabetes Mellitus/microbiology
13.
Sci Rep ; 14(1): 13513, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866914

ABSTRACT

Fecal calprotectin is an established marker of gut inflammation in inflammatory bowel disease (IBD). Elevated levels of fecal calprotectin as well as gut microbial dysbiosis have also been observed in other clinical conditions. However, systemic and multi-omics alterations linked to elevated fecal calprotectin in older individuals remain unclear. This study comprehensively investigated the relationship between fecal calprotectin levels, gut microbiome composition, serum inflammation and targeted metabolomics markers, and relevant lifestyle and medical data in a large sample of older individuals (n = 735; mean age ± SD: 68.7 ± 6.3) from the TREND cohort study. Low (0-50 µg/g; n = 602), moderate (> 50-100 µg/g; n = 64) and high (> 100 µg/g; n = 62) fecal calprotectin groups were stratified. Several pro-inflammatory gut microbial genera were significantly increased and short-chain fatty acid producing genera were decreased in high vs. low calprotectin groups. In serum, IL-17C, CCL19 and the toxic metabolite indoxyl sulfate were increased in high vs. low fecal calprotectin groups. These changes were partially mediated by the gut microbiota. Moreover, the high fecal calprotectin group showed increased BMI and a higher disease prevalence of heart attack and obesity. Our findings contribute to the understanding of fecal calprotectin as a marker of gut dysbiosis and its broader systemic and clinical implications in older individuals.


Subject(s)
Biomarkers , Dysbiosis , Feces , Gastrointestinal Microbiome , Leukocyte L1 Antigen Complex , Humans , Leukocyte L1 Antigen Complex/analysis , Leukocyte L1 Antigen Complex/metabolism , Feces/microbiology , Feces/chemistry , Dysbiosis/diagnosis , Aged , Female , Male , Biomarkers/blood , Biomarkers/analysis , Middle Aged , Cohort Studies , Inflammatory Bowel Diseases/blood , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology
14.
Gut ; 73(8): 1292-1301, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38839272

ABSTRACT

OBJECTIVE: There is a strong clinical association between IBD and primary sclerosing cholangitis (PSC), a chronic disease of the liver characterised by biliary inflammation that leads to strictures and fibrosis. Approximately 60%-80% of people with PSC will also develop IBD (PSC-IBD). One hypothesis explaining this association would be that PSC drives IBD. Therefore, our aim was to test this hypothesis and to decipher the underlying mechanism. DESIGN: Colitis severity was analysed in experimental mouse models of colitis and sclerosing cholangitis, and people with IBD and PSC-IBD. Foxp3+ Treg-cell infiltration was assessed by qPCR and flow cytometry. Microbiota profiling was carried out from faecal samples of people with IBD, PSC-IBD and mouse models recapitulating these diseases. Faecal microbiota samples collected from people with IBD and PSC-IBD were transplanted into germ-free mice followed by colitis induction. RESULTS: We show that sclerosing cholangitis attenuated IBD in mouse models. Mechanistically, sclerosing cholangitis causes an altered intestinal microbiota composition, which promotes Foxp3+ Treg-cell expansion, and thereby protects against IBD. Accordingly, sclerosing cholangitis promotes IBD in the absence of Foxp3+ Treg cells. Furthermore, people with PSC-IBD have an increased Foxp3+ expression in the colon and an overall milder IBD severity. Finally, by transplanting faecal microbiota into gnotobiotic mice, we showed that the intestinal microbiota of people with PSC protects against colitis. CONCLUSION: This study shows that PSC attenuates IBD and provides a comprehensive insight into the mechanisms involved in this effect.


Subject(s)
Cholangitis, Sclerosing , Disease Models, Animal , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , T-Lymphocytes, Regulatory , Cholangitis, Sclerosing/immunology , Cholangitis, Sclerosing/complications , Cholangitis, Sclerosing/microbiology , Animals , Mice , T-Lymphocytes, Regulatory/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/immunology , Humans , Forkhead Transcription Factors/metabolism , Colitis/microbiology , Colitis/complications , Male , Fecal Microbiota Transplantation , Female , Feces/microbiology , Mice, Inbred C57BL
15.
Front Biosci (Elite Ed) ; 16(2): 13, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38939911

ABSTRACT

Adverse intestinal microbiome profiles described as a dysbiotic gut are a complicit etiological operative factor that can progress and maintain inflammatory sequelae in the intestines. The disruption of the gut microbiome that ensues with intestinal dysbiosis is, for example, posited by decreases in the alpha-diversity of the gut microbiome, which is characterized by significant reductions in the abundance of bacterial members from the Bacteroidetes and Firmicutes phyla. Proteobacteria have often been recognized as gut microbial signatures of disease. For example, this happens with observed increases in abundance of the phyla Proteobacteria and Gammaproteobacteria, such as the adherent-invasive Escherichia coli strain, which has been significantly linked with maintaining inflammatory bowel diseases. Research on the administration of probiotics, often identified as gut-functional foods, has demonstrated safety, tolerability, and efficacy issues in treating inflammatory bowel diseases (IBDs). In this narrative review, we explore the efficacy of probiotics in treating IBDs with bacterial strain- and dose-specific characteristics and the association with multi-strain administration.


Subject(s)
Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Probiotics , Probiotics/therapeutic use , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Humans , Functional Food
16.
J Nanobiotechnology ; 22(1): 371, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-38918805

ABSTRACT

The gut microbiota is one of the essential contributors of the pathogenesis and progress of inflammatory bowel disease (IBD). Compared with first-line drug therapy, probiotic supplementation has emerged as a viable and secure therapeutic approach for managing IBD through the regulation of both the immune system and gut microbiota. Nevertheless, the efficacy of oral probiotic supplements is hindered by their susceptibility to the gastrointestinal barrier, leading to diminished bioavailability and restricted intestinal colonization. Here, we developed a bacteria-microalgae symbiosis system (EcN-SP) for targeted intestinal delivery of probiotics and highly effective treatment of colitis. The utilization of mircroalge Spirulina platensis (SP) as a natural carrier for the probiotic Escherichia coli Nissle 1917 (EcN) demonstrated potential benefits in promoting EcN proliferation, facilitating effective intestinal delivery and colonization. The alterations in the binding affinity of EcN-SP within the gastrointestinal environment, coupled with the distinctive structural properties of the SP carrier, served to overcome gastrointestinal barriers, minimizing transgastric EcN loss and enabling sustained intestinal retention and colonization. The oral administration of EcN-SP could effectively treat IBD by reducing the expression of intestinal inflammatory factors, maintaining the intestinal barrier and regulating the balance of gut microbiota. This probiotic delivery approach is inspired by symbiotic interactions found in nature and offers advantages in terms of feasibility, safety, and efficacy, thus holding significant promise for the management of gastrointestinal disorders.


Subject(s)
Escherichia coli , Gastrointestinal Microbiome , Microalgae , Probiotics , Spirulina , Symbiosis , Animals , Mice , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/therapy , Humans , Colitis , Mice, Inbred C57BL , Male , Drug Delivery Systems/methods
17.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928402

ABSTRACT

The gut microbial and metabolic characteristics of intestinal Behçet's disease (BD), a condition sharing many clinical similarities with ulcerative colitis (UC) and Crohn's disease (CD), are largely unexplored. This study investigated the gut microbial and metabolic characteristics of intestinal BD as well as potential biomarkers, comparing them with those in UC, CD, and healthy controls. Colon tissue and stool samples from 100 patients (35 UC, 30 CD, and 35 intestinal BD) and 41 healthy volunteers were analyzed using 16S ribosomal RNA sequencing to assess microbial diversity, taxonomic composition, and functional profiling. Plasma metabolomic analyses were performed using gas chromatography and ultra-performance liquid chromatography-mass spectrometry. Results indicated reduced microbial diversity in CD but not in intestinal BD, with intestinal BD showing fewer changes compared to controls yet distinct taxonomic features from UC, CD, and controls. Common alterations across all diseases included a reduction in beneficial bacteria producing short-chain fatty acids. Intestinal BD-specific changes featured a decreased abundance of Bacteroides fragilis. Metabolomic profiles in intestinal BD were similar to those in CD but distinct from those in UC, displaying significant changes in energy metabolism and genetic information processing. This integrative analysis revealed both shared and unique profiles in intestinal BD compared with UC, CD, and controls, advancing our understanding of the distinctive features of these diseases.


Subject(s)
Behcet Syndrome , Gastrointestinal Microbiome , Metabolome , Humans , Behcet Syndrome/microbiology , Behcet Syndrome/metabolism , Male , Female , Adult , Middle Aged , RNA, Ribosomal, 16S/genetics , Crohn Disease/microbiology , Crohn Disease/metabolism , Metabolomics/methods , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Biomarkers , Feces/microbiology , Colitis, Ulcerative/microbiology , Colitis, Ulcerative/metabolism , Case-Control Studies
18.
Cell Host Microbe ; 32(7): 1147-1162.e12, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38917808

ABSTRACT

Gut bacteria are implicated in inflammatory bowel disease (IBD), but the strains driving these associations are unknown. Large-scale studies of microbiome evolution could reveal the imprint of disease on gut bacteria, thus pinpointing the strains and genes that may underlie inflammation. Here, we use stool metagenomes of thousands of IBD patients and healthy controls to reconstruct 140,000 strain genotypes, revealing hundreds of lineages enriched in IBD. We demonstrate that these strains are ancient, taxonomically diverse, and ubiquitous in humans. Moreover, disease-associated strains outcompete their healthy counterparts during inflammation, implying long-term adaptation to disease. Strain genetic differences map onto known axes of inflammation, including oxidative stress, nutrient biosynthesis, and immune evasion. Lastly, the loss of health-associated strains of Eggerthella lenta was predictive of fecal calprotectin, a biomarker of disease severity. Our work identifies reservoirs of strain diversity that may impact inflammatory disease and can be extended to other microbiome-associated diseases.


Subject(s)
Feces , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Inflammatory Bowel Diseases/microbiology , Feces/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Metagenome , Phylogeny , Genotype
19.
Proc Natl Acad Sci U S A ; 121(27): e2315944121, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38917002

ABSTRACT

Chronic inflammation is epidemiologically linked to the pathogenesis of gastrointestinal diseases, including inflammatory bowel disease (IBD) and colorectal cancer (CRC). However, our understanding of the molecular mechanisms controlling gut inflammation remains insufficient, hindering the development of targeted therapies for IBD and CRC. In this study, we uncovered C15ORF48/miR-147 as a negative regulator of gut inflammation, operating through the modulation of epithelial cell metabolism. C15ORF48/miR-147 encodes two molecular products, C15ORF48 protein and miR-147-3p microRNA, which are predominantly expressed in the intestinal epithelium. C15ORF48/miR-147 ablation leads to gut dysbiosis and exacerbates chemically induced colitis in mice. C15ORF48 and miR-147-3p work together to suppress colonocyte metabolism and inflammation by silencing NDUFA4, a subunit of mitochondrial complex IV (CIV). Interestingly, the C15ORF48 protein, a structural paralog of NDUFA4, contains a unique C-terminal α-helical domain crucial for displacing NDUFA4 from CIV and its subsequent degradation. NDUFA4 silencing hinders NF-κB signaling activation and consequently attenuates inflammatory responses. Collectively, our findings have established the C15ORF48/miR-147-NDUFA4 molecular axis as an indispensable regulator of gut homeostasis, bridging mitochondrial metabolism and inflammation.


Subject(s)
Energy Metabolism , Gastrointestinal Microbiome , Inflammation , MicroRNAs , Animals , Humans , Mice , Colitis/metabolism , Colitis/microbiology , Colitis/genetics , Colitis/chemically induced , Dysbiosis/metabolism , Dysbiosis/microbiology , Energy Metabolism/genetics , Inflammation/metabolism , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/genetics , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Mice, Inbred C57BL , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...