Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 8.472
1.
Life Sci Alliance ; 7(7)2024 Jul.
Article En | MEDLINE | ID: mdl-38719750

Celiac disease (CD) is an autoimmune enteropathy resulting from an interaction between diet, genome, and immunity. Although many patients respond to a gluten-free diet, in a substantive number of individuals, the intestinal injury persists. Thus, other factors might amplify the ongoing inflammation. Candida albicans is a commensal fungus that is well adapted to the intestinal life. However, specific conditions increase Candida pathogenicity. The hypothesis that Candida may be a trigger in CD has been proposed after the observation of similarity between a fungal wall component and two CD-related gliadin T-cell epitopes. However, despite being implicated in intestinal disorders, Candida may also protect against immune pathologies highlighting a more intriguing role in the gut. Herein, we postulated that a state of chronic inflammation associated with microbial dysbiosis and leaky gut are favorable conditions that promote C. albicans pathogenicity eventually contributing to CD pathology via a mast cells (MC)-IL-9 axis. However, the restoration of immune and microbial homeostasis promotes a beneficial C. albicans-MC cross-talk favoring the attenuation of CD pathology to alleviate CD pathology and symptoms.


Candida albicans , Celiac Disease , Homeostasis , Mast Cells , Celiac Disease/immunology , Celiac Disease/microbiology , Celiac Disease/metabolism , Humans , Candida albicans/pathogenicity , Candida albicans/immunology , Mast Cells/immunology , Mast Cells/metabolism , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Candidiasis/immunology , Candidiasis/microbiology , Animals , Candida/pathogenicity , Candida/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism
2.
BMC Gastroenterol ; 24(1): 158, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720308

BACKGROUND: Primary sclerosing cholangitis (PSC) is a complex disease with pathogenic mechanisms that remain to be elucidated. Previous observational studies with small sample sizes have reported associations between PSC, dyslipidemia, and gut microbiota dysbiosis. However, the causality of these associations is uncertain, and there has been no systematic analysis to date. METHODS: The datasets comprise data on PSC, 179 lipid species, and 412 gut microbiota species. PSC data (n = 14,890) were sourced from the International PSC Study Group, while the dataset pertaining to plasma lipidomics originated from a study involving 7174 Finnish individuals. Data on gut microbiota species were derived from the Dutch Microbiome Project study, which conducted a genome-wide association study involving 7738 participants. Furthermore, we employed a two-step Mendelian randomization (MR) analysis to quantify the proportion of the effect of gut microbiota-mediated lipidomics on PSC. RESULTS: Following a rigorous screening process, our MR analysis revealed a causal relationship between higher levels of gene-predicted Phosphatidylcholine (O-16:1_18:1) (PC O-16:1_18:1) and an increased risk of developing PSC (inverse variance-weighted method, odds ratio (OR) 1.30, 95% confidence interval (CI) 1.03-1.63). There is insufficient evidence to suggest that gene-predicted PSC impacts the levels of PC O-16:1_18:1 (OR 1.01, 95% CI 0.98-1.05). When incorporating gut microbiota data into the analysis, we found that Eubacterium rectale-mediated genetic prediction explains 17.59% of the variance in PC O-16:1_18:1 levels. CONCLUSION: Our study revealed a causal association between PC O-16:1_18:1 levels and PSC, with a minor portion of the effect mediated by Eubacterium rectale. This study aims to further explore the pathogenesis of PSC and identify promising therapeutic targets. For patients with PSC who lack effective treatment options, the results are encouraging.


Cholangitis, Sclerosing , Gastrointestinal Microbiome , Lipidomics , Mendelian Randomization Analysis , Humans , Cholangitis, Sclerosing/blood , Cholangitis, Sclerosing/microbiology , Cholangitis, Sclerosing/genetics , Gastrointestinal Microbiome/genetics , Male , Genome-Wide Association Study , Female , Phosphatidylcholines/blood , Dysbiosis/blood , Middle Aged , Adult
3.
J Transl Med ; 22(1): 433, 2024 May 08.
Article En | MEDLINE | ID: mdl-38720361

Doxorubicin (DOX) is a broad-spectrum and highly efficient anticancer agent, but its clinical implication is limited by lethal cardiotoxicity. Growing evidences have shown that alterations in intestinal microbial composition and function, namely dysbiosis, are closely linked to the progression of DOX-induced cardiotoxicity (DIC) through regulating the gut-microbiota-heart (GMH) axis. The role of gut microbiota and its metabolites in DIC, however, is largely unelucidated. Our review will focus on the potential mechanism between gut microbiota dysbiosis and DIC, so as to provide novel insights into the pathophysiology of DIC. Furthermore, we summarize the underlying interventions of microbial-targeted therapeutics in DIC, encompassing dietary interventions, fecal microbiota transplantation (FMT), probiotics, antibiotics, and natural phytochemicals. Given the emergence of microbial investigation in DIC, finally we aim to point out a novel direction for future research and clinical intervention of DIC, which may be helpful for the DIC patients.


Cardiotoxicity , Doxorubicin , Gastrointestinal Microbiome , Gastrointestinal Microbiome/drug effects , Humans , Doxorubicin/adverse effects , Cardiotoxicity/etiology , Animals , Dysbiosis , Fecal Microbiota Transplantation
4.
Microbiome ; 12(1): 85, 2024 May 10.
Article En | MEDLINE | ID: mdl-38725043

BACKGROUND: Left ventricular diastolic dysfunction (LVDD) is an important precursor of heart failure (HF), but little is known about its relationship with gut dysbiosis and microbial-related metabolites. By leveraging the multi-omics data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), a study with population at high burden of LVDD, we aimed to characterize gut microbiota associated with LVDD and identify metabolite signatures of gut dysbiosis and incident LVDD. RESULTS: We included up to 1996 Hispanic/Latino adults (mean age: 59.4 years; 67.1% female) with comprehensive echocardiography assessments, gut microbiome, and blood metabolome data. LVDD was defined through a composite criterion involving tissue Doppler assessment and left atrial volume index measurements. Among 1996 participants, 916 (45.9%) had prevalent LVDD, and 212 out of 594 participants without LVDD at baseline developed incident LVDD over a median 4.3 years of follow-up. Using multivariable-adjusted analysis of compositions of microbiomes (ANCOM-II) method, we identified 7 out of 512 dominant gut bacterial species (prevalence > 20%) associated with prevalent LVDD (FDR-q < 0.1), with inverse associations being found for Intestinimonas_massiliensis, Clostridium_phoceensis, and Bacteroide_coprocola and positive associations for Gardnerella_vaginali, Acidaminococcus_fermentans, Pseudomonas_aeruginosa, and Necropsobacter_massiliensis. Using multivariable adjusted linear regression, 220 out of 669 circulating metabolites with detection rate > 75% were associated with the identified LVDD-related bacterial species (FDR-q < 0.1), with the majority being linked to Intestinimonas_massiliensis, Clostridium_phoceensis, and Acidaminococcus_fermentans. Furthermore, 46 of these bacteria-associated metabolites, mostly glycerophospholipids, secondary bile acids, and amino acids, were associated with prevalent LVDD (FDR-q < 0.1), 21 of which were associated with incident LVDD (relative risk ranging from 0.81 [p = 0.001, for guanidinoacetate] to 1.25 [p = 9 × 10-5, for 1-stearoyl-2-arachidonoyl-GPE (18:0/20:4)]). The inclusion of these 21 bacterial-related metabolites significantly improved the prediction of incident LVDD compared with a traditional risk factor model (the area under the receiver operating characteristic curve [AUC] = 0.73 vs 0.70, p = 0.001). Metabolite-based proxy association analyses revealed the inverse associations of Intestinimonas_massilliensis and Clostridium_phoceensis and the positive association of Acidaminococcus_fermentans with incident LVDD. CONCLUSION: In this study of US Hispanics/Latinos, we identified multiple gut bacteria and related metabolites linked to LVDD, suggesting their potential roles in this preclinical HF entity. Video Abstract.


Gastrointestinal Microbiome , Hispanic or Latino , Ventricular Dysfunction, Left , Humans , Female , Middle Aged , Male , Ventricular Dysfunction, Left/microbiology , Ventricular Dysfunction, Left/blood , United States , Dysbiosis/microbiology , Aged , Bacteria/classification , Bacteria/isolation & purification , Metabolome , Echocardiography
5.
BMC Microbiol ; 24(1): 161, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730357

Gestational diabetes mellitus (GDM) is characterized by insulin resistance and low-grade inflammation, and most studies have demonstrated gut dysbiosis in GDM pregnancies. Overall, they were manifested as a reduction in microbiome diversity and richness, depleted short chain fatty acid (SCFA)-producing genera and a dominant of Gram-negative pathogens releasing lipopolysaccharide (LPS). The SCFAs functioned as energy substance or signaling molecules to interact with host locally and beyond the gut. LPS contributed to pathophysiology of diseases through activating Toll-like receptor 4 (TLR4) and involved in inflammatory responses. The gut microbiome dysbiosis was not only closely related with GDM, it was also vital to fetal health through vertical transmission. In this review, we summarized gut microbiota signature in GDM pregnancies of each trimester, and presented a brief introduction of microbiome derived SCFAs. We then discussed mechanisms of microbiome-host interactions in the physiopathology of GDM and associated metabolic disorders. Finally, we compared offspring microbiota composition from GDM with that from normal pregnancies, and described the possible mechanism.


Diabetes, Gestational , Dysbiosis , Fatty Acids, Volatile , Gastrointestinal Microbiome , Diabetes, Gestational/microbiology , Diabetes, Gestational/metabolism , Humans , Pregnancy , Female , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Bacteria/classification , Bacteria/genetics , Bacteria/metabolism , Bacteria/isolation & purification , Host Microbial Interactions , Lipopolysaccharides/metabolism
6.
Respir Res ; 25(1): 204, 2024 May 10.
Article En | MEDLINE | ID: mdl-38730440

BACKGROUND: The impact of cigarette smoke (CS) on lung diseases and the role of microbiome dysbiosis in chronic obstructive pulmonary disease (COPD) have been previously reported; however, the relationships remain unclear. METHODS: Our research examined the effects of 20-week cigarette smoke (CS) exposure on the lung and intestinal microbiomes in C57BL/6JNarl mice, alongside a comparison with COPD patients' intestinal microbiome data from a public dataset. RESULTS: The study found that CS exposure significantly decreased forced vital capacity (FVC), thickened airway walls, and induced emphysema. Increased lung damage was observed along with higher lung keratinocyte chemoattractant (KC) levels by CS exposure. Lung microbiome analysis revealed a rise in Actinobacteriota, while intestinal microbiome showed significant diversity changes, indicating dysbiosis. Principal coordinate analysis highlighted distinct intestinal microbiome compositions between control and CS-exposed groups. In the intestinal microbiome, notable decreases in Patescibacteria, Campilobacterota, Defferibacterota, Actinobacteriota, and Desulfobacterota were observed. We also identified correlations between lung function and dysbiosis in both lung and intestinal microbiomes. Lung interleukins, interferon-É£, KC, and 8-isoprostane levels were linked to lung microbiome dysbiosis. Notably, dysbiosis patterns in CS-exposed mice were similar to those in COPD patients, particularly of Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage 4 patients. This suggests a systemic impact of CS exposure. CONCLUSION: In summary, CS exposure induces significant dysbiosis in lung and intestinal microbiomes, correlating with lung function decline and injury. These results align with changes in COPD patients, underscoring the important role of microbiome in smoke-related lung diseases.


Dysbiosis , Gastrointestinal Microbiome , Lung , Mice, Inbred C57BL , Pulmonary Disease, Chronic Obstructive , Animals , Pulmonary Disease, Chronic Obstructive/microbiology , Gastrointestinal Microbiome/physiology , Mice , Humans , Male , Lung/microbiology , Female , Middle Aged , Aged , Smoke/adverse effects
7.
Int J Mol Sci ; 25(9)2024 Apr 29.
Article En | MEDLINE | ID: mdl-38732048

HIV infection results in marked alterations in the gut microbiota (GM), such as the loss of microbial diversity and different taxonomic and metabolic profiles. Despite antiretroviral therapy (ART) partially ablating gastrointestinal alterations, the taxonomic profile after successful new ART has shown wide variations. Our objective was to determine the GM composition and functions in people living with HIV (PLWHIV) under ART in comparison to seronegative controls (SC). Fecal samples from 21 subjects (treated with integrase strand-transfer inhibitors, INSTIs) and 18 SC were included. We employed 16S rRNA amplicon sequencing, coupled with PICRUSt2 and fecal short-chain fatty acid (SCFA) quantification by gas chromatography. The INSTI group showed a decreased α-diversity (p < 0.001) compared to the SC group, at the expense of increased amounts of Pseudomonadota (Proteobacteria), Segatella copri, Lactobacillus, and Gram-negative bacteria. Concurrently, we observed an enrichment in Megasphaera and Butyricicoccus, both SCFA-producing bacteria, and significant elevations in fecal butyrate in this group (p < 0.001). Interestingly, gut dysbiosis in PLWHIV was characterized by a proinflammatory environment orchestrated by Pseudomonadota and elevated levels of butyrate associated with bacterial metabolic pathways, as well as the evident presence of butyrogenic bacteria. The role of this unique GM in PLWHIV should be evaluated, as well as the use of butyrate-based supplements and ART regimens that contain succinate, such as tenofovir disoproxil succinate. This mixed profile is described for the first time in PLWHIV from Mexico.


Feces , Gastrointestinal Microbiome , HIV Infections , RNA, Ribosomal, 16S , Humans , HIV Infections/microbiology , HIV Infections/drug therapy , Mexico , Female , Male , Adult , Middle Aged , Feces/microbiology , RNA, Ribosomal, 16S/genetics , Dysbiosis/microbiology , Fatty Acids, Volatile/metabolism , Fatty Acids, Volatile/analysis , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Butyrates/metabolism
8.
Food Res Int ; 186: 114404, 2024 Jun.
Article En | MEDLINE | ID: mdl-38729686

Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.


Autism Spectrum Disorder , Brain-Gut Axis , Flavonoids , Gastrointestinal Microbiome , Gastrointestinal Microbiome/drug effects , Humans , Autism Spectrum Disorder/microbiology , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/diet therapy , Flavonoids/pharmacology , Diet , Dysbiosis , Brain/metabolism , Animals , Antioxidants/pharmacology
9.
Gut Microbes ; 16(1): 2348441, 2024.
Article En | MEDLINE | ID: mdl-38706224

Colorectal cancer (CRC), a malignant tumor worldwide, is associated with gut microbiota. The influence of gut microbe-derived metabolites on CRC has attracted a lot of attention. However, the role of immunity mediated by commensal microbiota-derived metabolites in tumorigenesis of CRC is not intensively explored. Here we monitored the gut microbial dysbiosis in CRC mouse model (ApcMin/+ model) without dietary and pharmacological intervention, followed by characterized of metabolites enriched in CRC model mice. Profound changes of gut microbiome (bacteriome) were observed during intestinal disorders. Metabolomic profiling indicated that agmatine, derived from the gut bacteria i.e. Blautia, Odoribacter, Alistipes and Paraprevotella, could interact with Rnf128 to suppress the Rnf128-mediated ubiquitination of ß-catenin to further upregulate the downstream targets of ß-catenin including Cyclin D1, Lgr5, CD44 and C-myc, thus activating Wnt signaling. The activated Wnt signaling pathway promoted dysplasia of intestinal cells and inflammatory infiltration of lymphocytes via inducing the upregulation of pro-inflammatory cytokines (IL-6 and TNF-α) and downregulation of anti-inflammatory cytokine (IL-10), thereby contributing to colorectal carcinogenesis. Therefore, our study presented novel insights into the roles and mechanisms of gut microbiota in pathogenesis of CRC.


Carcinogenesis , Colorectal Neoplasms , Gastrointestinal Microbiome , Inflammation , Wnt Signaling Pathway , Animals , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/metabolism , Mice , Inflammation/metabolism , Inflammation/microbiology , Bacteria/classification , Bacteria/metabolism , Bacteria/isolation & purification , Bacteria/genetics , Mice, Inbred C57BL , beta Catenin/metabolism , Dysbiosis/microbiology , Humans , Disease Models, Animal , Cytokines/metabolism , Symbiosis , Male
10.
Front Cell Infect Microbiol ; 14: 1363276, 2024.
Article En | MEDLINE | ID: mdl-38707511

Introduction: Chronic kidney disease (CKD) is worldwide healthcare burden with growing incidence and death rate. Emerging evidence demonstrated the compositional and functional differences of gut microbiota in patients with CKD. As such, gut microbial features can be developed as diagnostic biomarkers and potential therapeutic target for CKD. Methods: To eliminate the outcome bias arising from factors such as geographical distribution, sequencing platform, and data analysis techniques, we conducted a comprehensive analysis of the microbial differences between patients with CKD and healthy individuals based on multiple samples worldwide. A total of 980 samples from six references across three nations were incorporated from the PubMed, Web of Science, and GMrepo databases. The obtained 16S rRNA microbiome data were subjected to DADA2 processing, QIIME2 and PICRUSt2 analyses. Results: The gut microbiota of patients with CKD differs significantly from that of healthy controls (HC), with a substantial decrease in the microbial diversity among the CKD group. Moreover, a significantly reduced abundance of bacteria Faecalibacterium prausnitzii (F. prausnitzii) was detected in the CKD group through linear discriminant analysis effect size (LEfSe) analysis, which may be associated with the alleviating effects against CKD. Notably, we identified CKD-depleted F. prausnitzii demonstrated a significant negative correlation with three pathways based on predictive functional analysis, suggesting its potential role in regulating systemic acidbase disturbance and pro-oxidant metabolism. Discussion: Our findings demonstrated notable alterations of gut microbiota in CKD patients. Specific gut-beneficial microbiota, especially F. prausnitzii, may be developed as a preventive and therapeutic tool for CKD clinical management.


Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Renal Insufficiency, Chronic , Gastrointestinal Microbiome/genetics , Humans , RNA, Ribosomal, 16S/genetics , Renal Insufficiency, Chronic/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Feces/microbiology , Phylogeny , Faecalibacterium prausnitzii/genetics , Biodiversity , Dysbiosis/microbiology
11.
Pharm Biol ; 62(1): 356-366, 2024 Dec.
Article En | MEDLINE | ID: mdl-38720666

CONTEXT: Yi-Shen-Hua-Shi (YSHS) is a traditional Chinese medicine that treats chronic kidney disease (CKD). However, its efficacy in reducing proteinuria and underlying mechanisms is unknown. OBJECTIVE: This single-center randomized controlled trial explored whether YSHS could improve proteinuria and modulate the gut microbiota. MATERIALS AND METHODS: 120 CKD patients were enrolled and randomized to receive the renin-angiotensin-aldosterone system (RAAS) inhibitor plus YSHS (n = 56) or RAAS inhibitor (n = 47) alone for 4 months, and 103 patients completed the study. We collected baseline and follow-up fecal samples and clinical outcomes from participants. Total bacterial DNA was extracted, and the fecal microbiome was analyzed using bioinformatics. RESULTS: Patients in the intervention group had a significantly higher decrease in 24-h proteinuria. After 4 months of the YSHS intervention, the relative abundance of bacteria that have beneficial effects on the body, such as Faecalibacterium, Lachnospiraceae, Lachnoclostridium, and Sutterella increased significantly, while pathogenic bacteria such as the Eggerthella and Clostridium innocuum group decreased. However, we could not find these changes in the control group. Redundancy analysis showed that the decline in 24-h proteinuria during follow-up was significantly correlated with various taxa of gut bacteria, such as Lachnospiraceae and the Lachnoclostridium genus in the YSHS group. KEGG analysis also showed the potential role of YSHS in regulating glycan, lipid, and vitamin metabolism. DISCUSSION AND CONCLUSION: The YSHS granule reduced proteinuria associated with mitigating intestinal microbiota dysbiosis in CKD patients. The definite mechanisms of YSHS to improve proteinuria need to be further explored. TRIAL REGISTRATION: ChiCTR2300076136, retrospectively registered.


Drugs, Chinese Herbal , Dysbiosis , Gastrointestinal Microbiome , Proteinuria , Renal Insufficiency, Chronic , Humans , Gastrointestinal Microbiome/drug effects , Male , Female , Renal Insufficiency, Chronic/microbiology , Renal Insufficiency, Chronic/drug therapy , Proteinuria/drug therapy , Proteinuria/microbiology , Middle Aged , Drugs, Chinese Herbal/pharmacology , Feces/microbiology , Aged , Adult , Medicine, Chinese Traditional/methods
12.
Sci Rep ; 14(1): 10394, 2024 05 06.
Article En | MEDLINE | ID: mdl-38710815

Tobacco use significantly influences the oral microbiome. However, less is known about how different tobacco products specifically impact the oral microbiome over time. To address this knowledge gap, we characterized the oral microbiome of cigarette users, smokeless tobacco users, and non-users over 4 months (four time points). Buccal swab and saliva samples (n = 611) were collected from 85 participants. DNA was extracted from all samples and sequencing was carried out on an Illumina MiSeq, targeting the V3-V4 region of the 16S rRNA gene. Cigarette and smokeless tobacco users had more diverse oral bacterial communities, including a higher relative abundance of Firmicutes and a lower relative abundance of Proteobacteria, when compared to non-users. Non-users had a higher relative abundance of Actinomyces, Granulicatella, Haemophilus, Neisseria, Oribacterium, Prevotella, Pseudomonas, Rothia, and Veillonella in buccal swab samples, compared to tobacco users. While the most abundant bacterial genera were relatively constant over time, some species demonstrated significant shifts in relative abundance between the first and last time points. In addition, some opportunistic pathogens were detected among tobacco users including Neisseria subflava, Bulleidia moorei and Porphyromonas endodontalis. Overall, our results provide a more holistic understanding of the structure of oral bacterial communities in tobacco users compared to non-users.


Dysbiosis , Microbiota , Mouth , RNA, Ribosomal, 16S , Tobacco, Smokeless , Humans , Tobacco, Smokeless/adverse effects , Male , Female , Dysbiosis/microbiology , Adult , RNA, Ribosomal, 16S/genetics , Mouth/microbiology , Saliva/microbiology , Middle Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Smokers , Young Adult , Cigarette Smoking/adverse effects , Mouth Mucosa/microbiology
13.
Clin Exp Rheumatol ; 42(5): 1097-1103, 2024 05.
Article En | MEDLINE | ID: mdl-38743445

The relationship between intestinal microbiota and arthritis has garnered significant attention, with emerging evidence suggesting a potential association between dysbiosis and various forms of inflammatory arthropathies. While observational studies have provided valuable insights into microbiota alterations in patients with arthritis, establishing causality remains challenging. Observational data, influenced by multiple confounders such as environmental factors, medication effects, and dietary habits, are insufficient to conclusively determine whether microbiota changes are somehow causally linked to arthritis. The heterogeneity of results across independent studies further complicates interpretation. To further support this hypothesis, interventional randomised trials are deemed necessary, yet their implementation in this area presents significant technical limitations. Experimental animal models offer insights into potential pathogenic mechanisms linking dysbiosis to arthritis, including compromised intestinal barrier function, the role of microbiota-derived metabolites and molecular mimicry. However, conflicting findings underscore the complexity of hostmicrobiota interactions and the challenges in establishing causality.Efforts to modulate the microbiota for arthritis treatment or prevention have shown promise, yet efficacy and applicability remains uncertain. Antibacterial drugs, dietary interventions, probiotics, and faecal microbiota transplantation have been explored, but their clinical utility awaits further validation. In conclusion, while the association between intestinal microbiota and arthritis is increasingly recognised, establishing causality remains elusive.


Dysbiosis , Gastrointestinal Microbiome , Humans , Animals , Probiotics/therapeutic use , Arthritis/microbiology , Fecal Microbiota Transplantation , Host-Pathogen Interactions , Risk Factors
14.
PLoS One ; 19(5): e0302726, 2024.
Article En | MEDLINE | ID: mdl-38743706

BACKGROUND: Dysbiosis during childhood impacts the configuration and maturation of the microbiota. The immaturity of the infant microbiota is linked with the development of inflammatory, allergic, and dysmetabolic diseases. AIMS: To identify taxonomic changes associated with age and GDM and classify the maturity of the intestinal microbiota of children of mothers with GDM and children without GDM (n-GDM). METHODS: Next-generation sequencing was used to analyze the V3-V4 region of 16S rRNA gene. QIIME2 and Picrust2 were used to determine the difference in the relative abundance of bacterial genera between the study groups and to predict the functional profile of the intestinal microbiota. RESULTS: According to age, the older GDM groups showed a lower alpha diversity and different abundance of Enterobacteriaceae, Veillonella, Clostridiales, and Bacteroides. Regarding the functional profile, PWY-7377 and K05895 associated with Vitamin B12 metabolism were reduced in GDM groups. Compared to n-GDM group, GDM offspring had microbiota immaturity as age-discriminatory taxa in random forest failed to classify GDM offspring according to developmental age (OOB error 81%). Conclusion. Offspring from mothers with GDM have a distinctive taxonomic profile related to taxa associated with gut microbiota immaturity.


Bacteroides , Diabetes, Gestational , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Veillonella , Humans , Diabetes, Gestational/microbiology , Female , Pregnancy , Bacteroides/genetics , RNA, Ribosomal, 16S/genetics , Veillonella/genetics , Infant , Adult , Male , Dysbiosis/microbiology , Feces/microbiology , Child, Preschool , High-Throughput Nucleotide Sequencing
15.
BMC Pulm Med ; 24(1): 235, 2024 May 14.
Article En | MEDLINE | ID: mdl-38745167

BACKGROUND: Emerging evidences have demonstrated that gut microbiota composition is associated with pulmonary arterial hypertension (PAH). However, the underlying causality between intestinal dysbiosis and PAH remains unresolved. METHOD: An analysis using the two-sample Mendelian randomization (MR) approach was conducted to examine the potential causal relationship between gut microbiota and PAH. To assess exposure data, genetic variants associated with 196 bacterial traits were extracted from the MiBioGen consortium, which included a sample size of 18,340 individuals. As for the outcomes, summary statistics for PAH were obtained from the NHGRI-EBI GWAS Catalog, which conducted a meta-analysis of four independent studies comprising a total of 11,744 samples. Causal effects were estimated employing various methods, including inverse variance weighted (IVW), MR-Egger, weighted median, weight mode and simple mode, with sensitivity analyses also being implemented with Cochran's Q test, MR-Egger intercept test, MR-PRESSO, leave-one-out analysis, and funnel plots. RESULTS: Following false discovery rate (FDR) correction, the genetically predicted genus Eubacterium fissicatena group (odds ratio (OR) 1.471, 95% confidence interval (CI) 1.178-1.837, q = 0.076) exhibited a causal association with PAH. In addition, the genus LachnospiraceaeUCG004 (OR 1.511, 95% CI 1.048-2.177) and genus RuminococcaceaeUCG002 (OR 1.407, 95% CI 1.040-1.905) showed a suggestive increased risk of PAH, while genus Eubacterium eligens group (OR 0.563, 95% CI 0.344-0.922), genus Phascolarctobacterium (OR 0.692, 95% CI 0.487-0.982), genus Erysipelatoclostridium (OR 0.757, 95% CI 0.579-0.989) and genus T-yzzerella3 (OR 0.768, 95% CI 0.624-0.945) were found to have nominal protective effect against PAH. CONCLUSION: The findings from our MR study have revealed a potential causal relationship between gut microbiota and PAH. Specifically, we have identified four types of gut microbiota that exhibit a protective effect on PAH, as well as three types that have a detrimental impact on PAH, thereby offering valuable insights for future mechanistic and clinical investigations in the field of PAH.


Gastrointestinal Microbiome , Mendelian Randomization Analysis , Humans , Gastrointestinal Microbiome/genetics , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/microbiology , Genome-Wide Association Study , Dysbiosis/genetics , Polymorphism, Single Nucleotide
16.
Int J Oral Sci ; 16(1): 39, 2024 May 13.
Article En | MEDLINE | ID: mdl-38740741

The aim of this study was to explore the impact of chronic apical periodontitis (CAP) on atherosclerosis in apoE-/- mice fed high-fat diet (HFD). This investigation focused on the gut microbiota, metabolites, and intestinal barrier function to uncover potential links between oral health and cardiovascular disease (CVD). In this study, CAP was shown to exacerbate atherosclerosis in HFD-fed apoE-/- mice, as evidenced by the increase in plaque size and volume in the aortic walls observed via Oil Red O staining. 16S rRNA sequencing revealed significant alterations in the gut microbiota, with harmful bacterial species thriving while beneficial species declining. Metabolomic profiling indicated disruptions in lipid metabolism and primary bile acid synthesis, leading to elevated levels of taurochenodeoxycholic acid (TCDCA), taurocholic acid (TCA), and tauroursodeoxycholic acid (TDCA). These metabolic shifts may contribute to atherosclerosis development. Furthermore, impaired intestinal barrier function, characterized by reduced mucin expression and disrupted tight junction proteins, was observed. The increased intestinal permeability observed was positively correlated with the severity of atherosclerotic lesions, highlighting the importance of the intestinal barrier in cardiovascular health. In conclusion, this research underscores the intricate interplay among oral health, gut microbiota composition, metabolite profiles, and CVD incidence. These findings emphasize the importance of maintaining good oral hygiene as a potential preventive measure against cardiovascular issues, as well as the need for further investigations into the intricate mechanisms linking oral health, gut microbiota, and metabolic pathways in CVD development.


Atherosclerosis , Diet, High-Fat , Dysbiosis , Gastrointestinal Microbiome , Animals , Diet, High-Fat/adverse effects , Atherosclerosis/metabolism , Mice , Male , Periapical Periodontitis/metabolism , Periapical Periodontitis/microbiology , Apolipoproteins E/metabolism , Mice, Inbred C57BL , RNA, Ribosomal, 16S
17.
Front Immunol ; 15: 1373504, 2024.
Article En | MEDLINE | ID: mdl-38715617

Cancer is a very aggressive disease and one of mankind's most important health problems, causing numerous deaths each year. Its etiology is complex, including genetic, gender-related, infectious diseases, dysbiosis, immunological imbalances, lifestyle, including dietary factors, pollution etc. Cancer patients also become immunosuppressed, frequently as side effects of chemotherapy and radiotherapy, and prone to infections, which further promote the proliferation of tumor cells. In recent decades, the role and importance of the microbiota in cancer has become a hot spot in human biology research, bringing together oncology and human microbiology. In addition to their roles in the etiology of different cancers, microorganisms interact with tumor cells and may be involved in modulating their response to treatment and in the toxicity of anti-tumor therapies. In this review, we present an update on the roles of microbiota in cancer with a focus on interference with anticancer treatments and anticancer potential.


Disease Progression , Neoplasms , Humans , Neoplasms/microbiology , Neoplasms/therapy , Neoplasms/immunology , Neoplasms/etiology , Animals , Antineoplastic Agents/therapeutic use , Microbiota , Gastrointestinal Microbiome/drug effects , Dysbiosis
18.
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
19.
Elife ; 122024 May 01.
Article En | MEDLINE | ID: mdl-38690990

Caesarean section scar diverticulum (CSD) is a significant cause of infertility among women who have previously had a Caesarean section, primarily due to persistent inflammatory exudation associated with this condition. Even though abnormal bacterial composition is identified as a critical factor leading to this chronic inflammation, clinical data suggest that a long-term cure is often unattainable with antibiotic treatment alone. In our study, we employed metagenomic analysis and mass spectrometry techniques to investigate the fungal composition in CSD and its interaction with bacteria. We discovered that local fungal abnormalities in CSD can disrupt the stability of the bacterial population and the entire microbial community by altering bacterial abundance via specific metabolites. For instance, Lachnellula suecica reduces the abundance of several Lactobacillus spp., such as Lactobacillus jensenii, by diminishing the production of metabolites like Goyaglycoside A and Janthitrem E. Concurrently, Clavispora lusitaniae and Ophiocordyceps australis can synergistically impact the abundance of Lactobacillus spp. by modulating metabolite abundance. Our findings underscore that abnormal fungal composition and activity are key drivers of local bacterial dysbiosis in CSD.


Bacteria , Cesarean Section , Cicatrix , Diverticulum , Female , Cesarean Section/adverse effects , Humans , Diverticulum/microbiology , Diverticulum/metabolism , Bacteria/metabolism , Bacteria/genetics , Cicatrix/microbiology , Cicatrix/metabolism , Dysbiosis/microbiology , Fungi/metabolism , Fungi/genetics , Fungi/physiology , Microbial Interactions , Microbiota
20.
World J Gastroenterol ; 30(16): 2191-2194, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38690026

This editorial explores the intricate relationship between microplastics (MPs) and gut microbiota, emphasizing the complexity and environmental health implications. The gut microbiota, a crucial component of gastrointestinal health, is examined in the context of potential microbial degradation of MPs. Furthermore, dysbiosis induced by MPs emerges as a consensus, disrupting the balance of gut microbiota and decreasing diversity. The mechanisms triggering dysbiosis, including physical interactions and chemical composition, are under investigation. Ongoing research addresses the consequences of MPs on immune fun-ction, nutrient metabolism, and overall host health. The bidirectional relationship between MPs and gut microbiota has significant implications for environmental and human health. Despite uncertainties, MPs negatively impact gut microbiota and health. Further research is essential to unravel the complex interactions and assess the long-term consequences of MPs on both environmental and human well-being.


Dysbiosis , Gastrointestinal Microbiome , Microplastics , Microplastics/adverse effects , Humans , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Environmental Health , Environmental Exposure/adverse effects , Animals
...