Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 9.096
Filter
1.
Cent Eur J Public Health ; 32(3): 200-204, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39352096

ABSTRACT

OBJECTIVES: The dietary composition is able to rapidly and significantly influence the diversity of the gut microbiome. This article focuses on how various types of diet affect the composition of the gut microbiome and how dietary changes are able to prevent or slow down the development of non-communicable diseases including obesity, type 2 diabetes mellitus, cardiovascular diseases, and low-grade inflammation. METHODS: A review in PubMed and a hand search using references in identified articles were performed. Studies published in English from 2000 to 2024 were included. RESULTS: The studies showed the significant effect of diet on the development of non-communicable diseases dependent on the state of the gut microbiota and molecules it produces. The Western diet that continues to gain in popularity for Czech people, leads to dysbiosis and production of bacterial lipopolysaccharide or trimethylamine N-oxide causing systemic chronic inflammation in the body and thus promoting the development of non-communicable diseases. CONCLUSIONS: Findings from this review emphasize the importance of healthy eating habits in the prevention of intestinal dysbiosis and still increasing prevalence and incidence of obesity and other non-communicable diseases.


Subject(s)
Diet , Gastrointestinal Microbiome , Noncommunicable Diseases , Humans , Gastrointestinal Microbiome/physiology , Noncommunicable Diseases/epidemiology , Noncommunicable Diseases/prevention & control , Obesity/microbiology , Obesity/epidemiology , Dysbiosis , Diabetes Mellitus, Type 2/prevention & control , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/epidemiology , Czech Republic/epidemiology , Inflammation
2.
World J Gastroenterol ; 30(36): 4078-4082, 2024 Sep 28.
Article in English | MEDLINE | ID: mdl-39351246

ABSTRACT

This letter emphasizes the need to expand discussions on gut microbiome's role in inflammatory bowel disease (IBD) and colorectal cancer (CRC) by including the often-overlooked non-bacterial components of the human gut flora. It highlights how viral, fungal and archaeal inhabitants of the gut respond towards gut dys-biosis and contribute to disease progression. Viruses such as bacteriophages target certain bacterial species and modulate the immune system. Other viruses found associated include Epstein-Barr virus, human papillomavirus, John Cunningham virus, cytomegalovirus, and human herpes simplex virus type 6. Fungi such as Candida albicans and Malassezia contribute by forming tissue-invasive filaments and producing inflammatory cytokines, respectively. Archaea, mainly metha-nogens are also found altering the microbial fermentation pathways. This corres-pondence, thus underscores the significance of considering the pathological and physiological mechanisms of the entire spectrum of the gut microbiota to develop effective therapeutic interventions for both IBD and CRC.


Subject(s)
Colorectal Neoplasms , Disease Progression , Dysbiosis , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Humans , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Colorectal Neoplasms/immunology , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/immunology , Dysbiosis/immunology , Bacteria , Fungi/immunology , Fungi/pathogenicity
3.
Front Cell Infect Microbiol ; 14: 1477143, 2024.
Article in English | MEDLINE | ID: mdl-39359935

ABSTRACT

Oral mucositis is a common and debilitating oral complication in head and neck cancer patients undergoing radiotherapy, resulting in diminished quality of life and potential treatment disruptions. Oral microbiota has long been recognized as a contributing factor in the initiation and progression of radiation-induced oral mucositis (RIOM). Numerous studies have indicated that the radiation-induced oral microbial dysbiosis promotes the occurrence and severity of oral mucositis. Therefore, approaches that modulate oral microbial ecology are promising for the management of RIOM. Probiotics as a relatively predicable and safe measure that modulates microecology have garnered significant interest. In this review, we discussed the correlation between RIOM and oral microbiota, with a particular focus on the efficacy of probiotics in the control of RIOM, in order to provide novel paradigm for the management of this disease.


Subject(s)
Dysbiosis , Probiotics , Radiation Injuries , Stomatitis , Probiotics/therapeutic use , Humans , Stomatitis/etiology , Stomatitis/microbiology , Stomatitis/therapy , Stomatitis/prevention & control , Radiation Injuries/therapy , Microbiota , Head and Neck Neoplasms/radiotherapy , Radiotherapy/adverse effects , Mouth/microbiology , Quality of Life
4.
Int J Chron Obstruct Pulmon Dis ; 19: 2153-2167, 2024.
Article in English | MEDLINE | ID: mdl-39360021

ABSTRACT

Background: Recent evidence suggests that the gut microbiome and metabolites are intricately involved in Chronic Obstructive Pulmonary Disease (COPD) pathogenesis, yet the precise causal relationships remain unclear due to confounding factors and reverse causation. This study employs bidirectional two-sample Mendelian Randomization (MR) to clarify these connections. Methods: Summary data from publicly available Genome-Wide Association Studies (GWAS) concerning the gut microbiome, metabolites, and COPD were compiled. The selection of genetic instrumental variables (Single Nucleotide Polymorphisms, or SNPs) for MR analysis was conducted meticulously, primarily utilizing the Inverse Variance Weighting (IVW) method, supplemented by MR-Egger regression and the Weighted Median (WM) approach. The evaluation of heterogeneity and horizontal pleiotropy was performed using Cochran's Q test, the MR-Egger intercept test, and the MR-PRESSO global test. Sensitivity analyses, including leave-one-out tests, were conducted to verify the robustness of our results. And the mediation effect of gut microbiota-mediated changes in metabolites on the causal relationship with COPD was analyzed. Results: Our study identified nine significant gut microbiota taxa and thirteen known metabolites implicated in COPD pathogenesis. Moreover, associations between the onset of COPD and the abundance of five bacterial taxa, as well as the concentration of three known metabolites, were established. These findings consistently withstood sensitivity analyses, reinforcing their credibility. Additionally, our results revealed that gut microbiota contribute to the development of COPD by mediating changes in metabolites. Conclusion: Our bidirectional Two-Sample Mendelian Randomization analysis has revealed reciprocal causal relationships between the abundance of gut microbiota and metabolite concentrations in the context of COPD. This research holds promise for identifying biomarkers for early COPD diagnosis and monitoring disease progression, thereby opening new pathways for prevention and treatment. Further investigation into the underlying mechanisms is essential to improve our understanding of COPD onset.


Subject(s)
Gastrointestinal Microbiome , Genome-Wide Association Study , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Pulmonary Disease, Chronic Obstructive , Pulmonary Disease, Chronic Obstructive/microbiology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/diagnosis , Humans , Risk Factors , Genetic Predisposition to Disease , Lung/microbiology , Lung/physiopathology , Phenotype , Risk Assessment , Dysbiosis , Bacteria/genetics , Bacteria/isolation & purification
5.
J Obes ; 2024: 5846674, 2024.
Article in English | MEDLINE | ID: mdl-39360185

ABSTRACT

Objective: The importance of early microbial dysbiosis in later development of obesity and metabolic disorders has been a subject of debate. Here we tested cause and effect in mice. Methods: Germ-free male Swiss Webster mice were colonized in a specific-pathogen-free (SPF) facility at 1 week (1W) and 3 weeks (3W) of age. They were challenged with a high-fat diet and their responses were compared with SPF mice. Gut microbiota was analyzed by 16S rRNA gene sequencing. Moreover, RNA sequencing of the liver was performed on additional 3W and SPF mice on a regular chow diet. Results: There were no significant differences in weight, food consumption, epididymal fat weight, HbA1c levels, and serum insulin and leptin, whereas the early germ-free period resulted in mice with impaired glucose tolerance. Both the 1W and 3W group peaked 56% (p < 0.05) and 66% (p < 0.01) higher in blood glucose than the SPF control group, respectively. This was accompanied by a 45% reduction in the level of the anti-inflammatory cytokine IL-10 in the 1W mice (p < 0.05). There were no differences in the gut microbiota between the groups, indicating that all mice colonized fully after the germ-free period. Marked effects on hepatic gene expression (728 differentially expressed genes with adjusted p < 0.05 and a fold change ± 1.5) suggested a potential predisposition to a higher risk of developing insulin resistance in the 3W group. Conclusions: Lack of microbes early in life had no impact on adiposity but led to insulin resistance and altered liver gene expression related to glucose metabolism in mice. The study strongly supports the notion that microbial signaling to the liver in the beginning of life can alter the host's risk of developing metabolic disorder later in life.


Subject(s)
Adiposity , Gastrointestinal Microbiome , Insulin Resistance , Liver , Obesity , Animals , Mice , Male , Liver/metabolism , Obesity/microbiology , Mice, Obese , Diet, High-Fat , Dysbiosis , Gene Expression
6.
J Med Microbiol ; 73(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39383061

ABSTRACT

Introduction. The human lower gastrointestinal tract microbiome is complex, dynamic and prone to disruption occurring during critical illness.Hypothesis or gap statement. The characteristics of lower gastrointestinal tract microbiome disruption and its association with clinical outcomes in patients with prolonged intensive care stay remain uncertain.Aim. To systematically review studies describing lower gastrointestinal tract molecular sequencing in patients with prolonged intensive care stay and explore associations with clinical outcomes.Methodology. This systematic review was prospectively registered and follows the Preferred Reporting Items for Systematic Reviews and Meta-analyses guidelines. OVID MEDLINE, EMBASE and The Cochrane Central Register of Controlled Trials databases were searched for eligible studies describing adults and/or children who underwent molecular sequencing of stool or rectal samples taken on or after 10 days of intensive care.Results. There were 13 studies with 177 patients included. The overall certainty of evidence was low, and no studies reported mortality. Reduced alpha diversity was observed in nine out of nine studies but was not associated with clinical outcomes in four out of four studies. Longitudinal alpha diversity decreased in five out of six studies, and inter-individual beta diversity increased in five out of five studies. After approximately one week of intensive care unit admission, rapid fluctuations in dominant taxa stabilized with trajectories of either recovery or deterioration in five studies. Pathogenic enrichment and commensal depletion were reported in all 13 studies and associated with clinical outcomes in two studies.Conclusion. Lower gastrointestinal tract microbiome disruption is highly prevalent and has consistent characteristics in patients with prolonged intensive care stay. Amongst reported metrics, only relative taxon abundance was associated with clinical outcomes.


Subject(s)
Critical Illness , Dysbiosis , Gastrointestinal Microbiome , Humans , Dysbiosis/microbiology , Lower Gastrointestinal Tract/microbiology , Intensive Care Units , Feces/microbiology , Adult
7.
Cell Host Microbe ; 32(10): 1641-1643, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39389025

ABSTRACT

The role of fatty acids in shaping vaginal microbiota remains unclear. In an issue of Cell, Zhu et al. use genomic and transcriptomic analyses to reveal that oleic acid (OA) selectively inhibits L. iners while promoting L. crispatus, suggesting new strategies for the treatment of bacterial vaginosis (BV).


Subject(s)
Dysbiosis , Homeostasis , Oleic Acid , Vagina , Vaginosis, Bacterial , Female , Vagina/microbiology , Humans , Dysbiosis/microbiology , Oleic Acid/metabolism , Oleic Acid/pharmacology , Vaginosis, Bacterial/microbiology , Vaginosis, Bacterial/drug therapy , Lactobacillus/metabolism , Lactobacillus/genetics , Microbiota
8.
Front Immunol ; 15: 1482214, 2024.
Article in English | MEDLINE | ID: mdl-39391302

ABSTRACT

Rheumatoid arthritis is a widely prevalent autoimmune bone disease that imposes a significant burden on global healthcare systems due to its increasing incidence. In recent years, attention has focused on the interaction between gut homeostasis and the immune system, particularly in relation to bone health. Dysbiosis, which refers to an imbalance in the composition and function of the gut microbiota, has been shown to drive immune dysregulation through mechanisms such as the release of pro-inflammatory metabolites, increased gut permeability, and impaired regulatory T cell function. These factors collectively contribute to immune system imbalance, promoting the onset and progression of Rheumatoid arthritis. Dysbiosis induces both local and systemic inflammatory responses, activating key pro-inflammatory cytokines such as tumor necrosis factor-alpha, Interleukin-6, and Interleukin-17, which exacerbate joint inflammation and damage. Investigating the complex interactions between gut homeostasis and immune regulation in the context of Rheumatoid arthritis pathogenesis holds promise for identifying new therapeutic targets, revealing novel mechanisms of disease progression, and offering innovative strategies for clinical treatment.


Subject(s)
Arthritis, Rheumatoid , Dysbiosis , Gastrointestinal Microbiome , Homeostasis , Humans , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/etiology , Gastrointestinal Microbiome/immunology , Dysbiosis/immunology , Animals , Immune System/immunology , Immune System/metabolism , Cytokines/metabolism , Cytokines/immunology
9.
Front Immunol ; 15: 1439176, 2024.
Article in English | MEDLINE | ID: mdl-39391303

ABSTRACT

The human gastrointestinal (GI) tract microbiome is a complex and all-encompassing ecological system of trillions of microorganisms. It plays a vital role in digestion, disease prevention, and overall health. When this delicate balance is disrupted, it can lead to various health issues. Fecal microbiota transplantation (FMT) is an emerging therapeutic intervention used as an adjuvant therapy for many diseases, particularly those with dysbiosis as their underlying cause. Its goal is to restore this balance by transferring fecal material from healthy donors to the recipients. FMT has an impressive reported cure rate between 80% and 90% and has become a favored treatment for many diseases. While FMT may have generally mild to moderate transient adverse effects, rare severe complications underscore the importance of rigorous donor screening and standardized administration. FMT has enormous potential as a practical therapeutic approach; however, additional research is required to further determine its potential for clinical utilization, as well as its safety and efficiency in different patient populations. This comprehensive literature review offers increased confidence in the safety and effectiveness of FMT for several diseases affecting the intestines and other systems, including diabetes, obesity, inflammatory and autoimmune illness, and other conditions.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Humans , Fecal Microbiota Transplantation/methods , Fecal Microbiota Transplantation/adverse effects , Dysbiosis/therapy , Animals , Treatment Outcome
11.
Front Cell Infect Microbiol ; 14: 1357178, 2024.
Article in English | MEDLINE | ID: mdl-39391883

ABSTRACT

Sepsis, a disease with high incidence, mortality, and treatment costs, has a complex interaction with the gut microbiota. With advances in high-throughput sequencing technology, the relationship between sepsis and intestinal dysbiosis has become a new research focus. However, owing to the intricate interplay between critical illness and clinical interventions, it is challenging to establish a causal relationship between sepsis and intestinal microbiota imbalance. In this review, the correlation between intestinal microecology and sepsis was summarized, and new therapies for sepsis intervention based on microecological target therapy were proposed, and the shortcomings of bacterial selection and application timing in clinical practice were addressed. In conclusion, current studies on metabolomics, genomics and other aspects aimed at continuously discovering potential probiotics are all providing theoretical basis for restoring intestinal flora homeostasis for subsequent treatment of sepsis.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Sepsis , Sepsis/microbiology , Humans , Probiotics/therapeutic use , Animals , Metabolomics , Intestines/microbiology
12.
Front Cell Infect Microbiol ; 14: 1450310, 2024.
Article in English | MEDLINE | ID: mdl-39391885

ABSTRACT

Infertility is a disease of impaired fertility. With socioeconomic development, changes in human lifestyles, and increased environmental pollution, the problem of low human fertility has become increasingly prominent. The incidence of global infertility is increasing every year. Many factors lead to infertility, and common female factors include tubal factors, ovulation disorders, endometriosis, and immune factors. The gut microbiota is involved in many physiological processes, such as nutrient absorption, intestinal mucosal growth, glycolipid metabolism, and immune system regulation. An altered gut flora is associated with female infertility disorders such as polycystic ovary syndrome (PCOS), endometriosis (EMs), and premature ovarian failure (POF). Dysbiosis of the gut microbiota directly or indirectly contributes to the development of female infertility disorders, which also affect the homeostasis of the gut microbiota. Identifying the etiology and pathogenesis of infertility in patients is the focus of reproductive medicine physicians. We studied the developmental mechanism between the gut microbiota and PCOS, EMs, and POF from a new perspective, providing new ideas for diagnosing and treating female infertility diseases and specific reference values for eugenics.


Subject(s)
Biomarkers , Dysbiosis , Gastrointestinal Microbiome , Infertility, Female , Polycystic Ovary Syndrome , Humans , Female , Dysbiosis/microbiology , Infertility, Female/microbiology , Polycystic Ovary Syndrome/microbiology , Polycystic Ovary Syndrome/complications , Endometriosis/microbiology , Endometriosis/complications
13.
Front Immunol ; 15: 1414653, 2024.
Article in English | MEDLINE | ID: mdl-39376557

ABSTRACT

Background: Previous studies have shown that gut microbiota dysbiosis could affect clinical prognosis through an unknown mechanism. However, the causal relationship between the gut microbiota and the functional outcome after ischemic stroke remains unclear. We aimed to investigate the causal association between the gut microbiota and the functional outcome after ischemic stroke using Mendelian randomization (MR). Methods: Genetic instrumental variables associated with 211 bacterial traits were obtained from the MiBioGen consortium (N = 18,340). Data from genome-wide association studies (GWAS) for functional outcome after ischemic stroke were obtained from two phenotypes (i.e., overall stroke outcome and motor recovery). The inverse variance weighted method was used to estimate the causal association. Enrichment analysis was conducted based on the results of the MR analyses. Results: The genetically predicted family Peptostreptococcaceae (OR = 0.63, 95% CI = 0.41-0.98, p = 0.038) and the genera LachnospiraceaeNK4A136 group (OR = 0.65, 95% CI = 0.43-1.00, p = 0.048), LachnospiraceaeUCG004 (OR = 0.54, 95% CI = 0.33-0.90, p = 0.017), and Odoribacter (OR = 0.40, 95% CI = 0.21-0.77, p = 0.006) presented a suggestive association with favorable functional outcome, while the genera Eubacterium oxidoreducens group (OR = 1.77, 95% CI = 1.11-2.84, p = 0.018) and RuminococcaceaeUCG005 (OR = 1.85, 95% CI = 1.15-2.96, p = 0.010) were associated with unfavorable functional outcome. The genetically predicted family Oxalobacteraceae (OR = 2.12, 95% CI = 1.10-4.11, p = 0.025) and the genus RuminococcaceaeUCG014 (OR = 4.17, 95% CI = 1.29-13.52, p = 0.017) showed a suggestive association with motor recovery, while the order Enterobacteriales (OR = 0.14, 95% CI = 0.02-0.87, p = 0.035) and the family Enterobacteriaceae (OR = 0.14, 95% CI = 0.02-0.87, p = 0.035) were associated with motor weakness. Enrichment analysis revealed that regulation of the synapse structure or activity may be involved in the effect of the gut microbiota on the functional outcome after ischemic stroke. Conclusions: This study provides genetic support that the gut microbiota, especially those associated with short-chain fatty acids, could affect stroke prognosis by mediating synapse function. Our findings suggest that modifying the composition of the gut microbiota could improve the prognosis of ischemic stroke.


Subject(s)
Gastrointestinal Microbiome , Genome-Wide Association Study , Ischemic Stroke , Mendelian Randomization Analysis , Humans , Gastrointestinal Microbiome/genetics , Ischemic Stroke/microbiology , Ischemic Stroke/genetics , Dysbiosis , Recovery of Function
14.
NPJ Biofilms Microbiomes ; 10(1): 104, 2024 Oct 10.
Article in English | MEDLINE | ID: mdl-39389989

ABSTRACT

Gynecologic cancers develop from the female reproductive organs. Microbial dysbiosis in the gut and oral cavity can communicate with each other through various ways, leading to mucosal destruction, inflammatory response, genomic instability, and ultimately inducing cancer and worsening. Here, we introduce the mechanisms of interactions between gut and oral microbiota and their changes in the development of gynecologic tumors. In addition, new therapeutic approaches based on microbiota modulation are discussed.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Genital Neoplasms, Female , Mouth , Humans , Female , Genital Neoplasms, Female/microbiology , Dysbiosis/microbiology , Mouth/microbiology , Microbiota
15.
Clin Liver Dis ; 28(4): 663-679, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39362714

ABSTRACT

Alcohol-associated liver disease (ALD) poses a significant global public health challenge, with high patient mortality rates and economic burden. The gut microbiome plays an important role in the onset and progression of alcohol-associated liver disease. Excessive alcohol consumption disrupts the intestinal barrier, facilitating the entry of harmful microbes and their products into the liver, exacerbating liver damage. Dysbiosis, marked by imbalance in gut bacteria, correlates with ALD severity. Promising microbiota-centered therapies include probiotics, phages, and fecal microbiota transplantation. Clinical trials demonstrate the potential of these interventions to improve liver function and patient outcomes, offering a new frontier in ALD treatment.


Subject(s)
Dysbiosis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Liver Diseases, Alcoholic , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Liver Diseases, Alcoholic/microbiology , Liver Diseases, Alcoholic/therapy , Probiotics/therapeutic use , Bacteriophages
16.
J Headache Pain ; 25(1): 171, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39379796

ABSTRACT

BACKGROUND: Recently, there has been increasing interest in the possible role of the gut microbiota (GM) in the onset of migraine. Our aim was to verify whether bacterial populations associated with intestinal dysbiosis are found in pediatric patients with migraine. We looked for which metabolic pathways, these bacteria were involved and whether they might be associated with gut inflammation and increased intestinal permeability. METHODS: Patients aged between 6 and 17 years were recruited. The GM profiling was performed by the 16S rRNA metataxonomics of faecal samples from 98 patients with migraine and 98 healthy subjects. Alpha and beta diversity analyses and multivariate and univariate analyses were applied to compare the gut microbiota profiles between the two group. To predict functional metabolic pathways, we used phylogenetic analysis of communities. The level of indican in urine was analyzed to investigate the presence of metabolic dysbiosis. To assess gut inflammation, increased intestinal permeability and the mucosal immune activation, we measured the plasmatic levels of lipopolysaccharide, occludin and IgA, respectively. RESULTS: The α-diversity analysis revealed a significant increase of bacterial richness in the migraine group. The ß-diversity analysis showed significant differences between the two groups indicating gut dysbiosis in patients with migraine. Thirty-seven metabolic pathways were increased in the migraine group, which includes changes in tryptophan and phenylalanine metabolism. The presence of metabolic dysbiosis was confirmed by the increased level of indican in urine. Increased levels of plasmatic occludin and IgA indicated the presence of intestinal permeability and mucosal immune activation. The plasmatic LPS levels showed a low intestinal inflammation in patients with migraine. CONCLUSIONS: Pediatric patients with migraine present GM profiles different from healthy subjects, associated with metabolic pathways important in migraine.


Subject(s)
Dysbiosis , Gastrointestinal Microbiome , Migraine Disorders , Humans , Dysbiosis/epidemiology , Dysbiosis/microbiology , Child , Migraine Disorders/microbiology , Migraine Disorders/metabolism , Gastrointestinal Microbiome/physiology , Adolescent , Female , Male , Inflammation/microbiology , Feces/microbiology , RNA, Ribosomal, 16S/analysis , RNA, Ribosomal, 16S/genetics
17.
Microbiology (Reading) ; 170(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39405098

ABSTRACT

Adherent-invasive Escherichia coli (AIEC) has been implicated in the aetiology of Crohn's disease (CD), a chronic inflammatory disorder of the gastrointestinal tract. The presence of Enterobacteriaceae, including AIEC, is heightened in the intestines of CD patients. Therefore, inhibiting AIEC colonization in the gastrointestinal tract could be a promising therapeutic intervention for CD. This study aims to assess the potential of EnvC as a novel therapeutic target, examining how disrupting EnvC activity through the deletion of the envC gene decreases AIEC gut colonization levels. EnvC serves as a catalyst for peptidoglycan (also called murein) amidases, facilitating bacterial cell division. An AIEC mutant lacking the envC gene exhibited impaired cell division. Furthermore, envC deletion led to a diminished outer membrane barrier, as seen in our finding that the envC mutant became susceptible to vancomycin. Finally, we found that the envC mutant is impaired in competitive gut colonization in a dysbiotic mouse model. The colonization defects might be attributable to reduced resistance to colonic bile acids, as evidenced by our finding that increased colonic levels of bile acids inhibited the colonization of the gastrointestinal tract by AIEC strains. The present findings suggest that targeting bacterial cell division through the inhibition of EnvC activity could represent a promising intervention for CD.


Subject(s)
Crohn Disease , Escherichia coli Proteins , Escherichia coli , Crohn Disease/microbiology , Animals , Escherichia coli/genetics , Escherichia coli/metabolism , Mice , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Gene Deletion , Escherichia coli Infections/microbiology , Bacterial Outer Membrane/metabolism , Humans , Disease Models, Animal , Bile Acids and Salts/metabolism , Bacterial Outer Membrane Proteins/genetics , Bacterial Outer Membrane Proteins/metabolism , Gastrointestinal Tract/microbiology , Anti-Bacterial Agents/pharmacology , Dysbiosis/microbiology , Gastrointestinal Microbiome , Mice, Inbred C57BL , Female
18.
Microbiome ; 12(1): 202, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39407320

ABSTRACT

BACKGROUND: Entomopathogenic fungal infection-induced dysbiosis of host microbiota offers a window into understanding the complex interactions between pathogenic fungi and host symbionts. Such insights are critical for enhancing the efficacy of mycoinsecticides. However, the utilization of these interactions in pest control remains largely unexplored. RESULTS: Here, we found that infection by the host-specialist fungus Metarhizium acridum alters the composition of the symbiotic microbiota and increases the dominance of some bacterial symbionts in locusts. Meanwhile, M. acridum also effectively limits the overgrowth of the predominant bacteria. Comparative transcriptomic screening revealed that the fungus upregulates the production of MaCFEM1, an iron-binding protein, in the presence of bacteria. This protein sequesters iron, thereby limiting its availability. Functionally, overexpression of MaCFEM1 in the fungus induces iron deprivation, which significantly suppresses bacterial growth. Conversely, MaCFEM1 knockout relieves the restriction on bacterial iron availability, resulting in iron reallocation. Upon ΔMaCFEM1 infection, some host bacterial symbionts proliferate uncontrollably, turning into opportunistic pathogens and significantly accelerating host death. CONCLUSIONS: This study elucidates the critical role of pathogenic fungal-dominated iron allocation in mediating the shift of host microbes from symbiosis to pathogenicity. It also highlights a unique biocontrol strategy that jointly exploits pathogenic fungi and bacterial symbionts to increase host mortality. Video Abstract.


Subject(s)
Bacteria , Fungal Proteins , Iron , Metarhizium , Symbiosis , Iron/metabolism , Animals , Metarhizium/genetics , Metarhizium/metabolism , Metarhizium/pathogenicity , Metarhizium/physiology , Fungal Proteins/genetics , Fungal Proteins/metabolism , Bacteria/metabolism , Bacteria/genetics , Bacteria/classification , Grasshoppers/microbiology , Host-Pathogen Interactions , Dysbiosis/microbiology
19.
Front Cell Infect Microbiol ; 14: 1402389, 2024.
Article in English | MEDLINE | ID: mdl-39380726

ABSTRACT

Introduction: Genitourinary syndrome of menopause (GSM) describes the symptoms and signs resulting from the effect of estrogen deficiency on the female genitourinary tract, including genital, urinary, and sexual symptoms. However, besides estrogen deficiency, little is known about the etiology of GSM. The objective of this study was to investigate the effects of vaginal microbiota dysbiosis on the occurrence and development of GSM in perimenopausal and postmenopausal women. Methods: In total, 96 women were enrolled in this cross-sectional study and clinical data were collected. GSM symptoms were divided into three types: genital, urological, and sexual symptoms. Full-length 16S rRNA gene sequencing using the third-generation PacBio sequencing technology was performed to analyze the vaginal microbiome using vaginal swabs of non-GSM and GSM women with different types of GSM symptoms. Live Lactobacillus Capsule for Vaginal Use (LLCVU) treatment was used to verify the effects of Lactobacillus on GSM symptoms. Results: We found that 83.58% (56/67) of women experienced GSM symptoms in the perimenopausal and postmenopausal stages. Among these women with GSM, 23.21% (13/56), 23.21% (13/56), and 53.57% (30/56) had one type, two types, and three types of GSM symptoms, respectively. The richness and diversity of vaginal microbiota gradually increased from reproductive to postmenopausal women. There were significant differences in vaginal microbial community among non-GSM women and GSM women with different types of symptoms. Lactobacillus was found to be negatively associated with the onset, severity, and type of GSM while some bacteria, such as Escherichia-shigella, Anaerococcus, Finegoldia, Enterococcus, Peptoniphilus_harei, and Streptococcus, were found to be positively associated with these aspects of GSM, and these bacteria were especially associated with the types of genital and sexual symptoms in GSM women. LLCVU significantly relieved genital symptoms and improved the sexual life of GSM women in shortterm observation. Conclusions: The onset, severity, and type of GSM symptoms may be associated with changes in vaginal microbiota in perimenopausal and postmenopausal women. Vaginal microbiota dysbiosis probably contributes to the occurrence and development of GSMsymptoms, especially vaginal and sexual symptoms. Lactobacillus used in the vagina may be a possible option for non-hormonal treatment of GSM women with genital and sexual symptoms. Clinical Trial Registration: https://www.chictr.org.cn/indexEN.html, identifier ChiCTR2100044237.


Subject(s)
Dysbiosis , Lactobacillus , Menopause , Microbiota , RNA, Ribosomal, 16S , Vagina , Adult , Female , Humans , Middle Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Cross-Sectional Studies , Dysbiosis/microbiology , Female Urogenital Diseases/microbiology , Lactobacillus/isolation & purification , Lactobacillus/genetics , Postmenopause , RNA, Ribosomal, 16S/genetics , Syndrome , Vagina/microbiology
20.
Alzheimers Res Ther ; 16(1): 222, 2024 Oct 14.
Article in English | MEDLINE | ID: mdl-39396997

ABSTRACT

BACKGROUND: Emerging evidence suggested the association between gut dysbiosis and Alzheimer's disease (AD) progression. However, it remained unclear how the gut microbiome and neuroinflammation in the brain mutually interact or how these interactions affect brain functioning and cognition. Here we hypothesized that "gut-brain" axis mediated by microbial derived metabolites was expected to novel breakthroughs in the fields of AD research and development. METHODS: Multiple technologies, such as immunofluorescence, 16s rDNA sequencing, mass spectrometry-based metabolomics (LC-QQQ-MS and GC-MS), were used to reveal potential link between gut microbiota and the metabolism and cognition of the host. RESULTS: Microbial depletion induced by the antibiotics mix (ABX) verified that "gut-brain" can transmit information bidirectionally. Short-chain fatty acid-producing (SCFAs-producing) bacteria and amino acid-producing bacteria fluctuated greatly in 5×FAD mice, especially the reduction sharply of the Bifidobacteriaceae and the increase of the Lachnospiraceae family. Concentrations of several Tryptophan-kynurenine intermediates, lactic acid, CD4+ cell, and CD8+ cells were higher in serum of 5×FAD mice, whilst TCA cycle intermediates and Th1/Th2 were lower. In addition, the levels of iso-butyric acid (IBA) in feces, serum, and brain of 5×FAD mice were increased compared with WT-M mice, especially in serum. And IBA in the brain was positively correlated with Aß and proinflammatory factors. CONCLUSION: Together, our finding highlighted that the alternation in gut microbiota affected the effective communication between the "gut-brain" axis in 5×FAD mice by regulating the immune system, carbohydrate, and energy metabolism.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Neuroinflammatory Diseases , Animals , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Mice , Neuroinflammatory Diseases/metabolism , Alzheimer Disease/metabolism , Dysbiosis/metabolism , Brain/metabolism , Mice, Transgenic , Drugs, Chinese Herbal/pharmacology , Disease Models, Animal , Mice, Inbred C57BL , Medicine, Chinese Traditional/methods
SELECTION OF CITATIONS
SEARCH DETAIL