Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.529
Filtrer
1.
J Pineal Res ; 76(5): e12989, 2024 Aug.
Article de Anglais | MEDLINE | ID: mdl-38978438

RÉSUMÉ

Colistin is renowned as a last-resort antibiotic due to the emergence of multidrug-resistant pathogens. However, its potential toxicity significantly hampers its clinical utilization. Melatonin, chemically known as N-acetyl-5-hydroxytryptamine, is an endogenous hormone produced by the pineal gland and possesses diverse biological functions. However, the protective role of melatonin in alleviating antibiotic-induced intestinal inflammation remains unknown. Herein, we reveal that colistin stimulation markedly elevates intestinal inflammatory levels and compromises the gut barrier. In contrast, pretreatment with melatonin safeguards mice against intestinal inflammation and mucosal damage. Microbial diversity analysis indicates that melatonin supplementation prevents a reduction in the abundance of Erysipelotrichales and Bifidobacteriales, as well as an increase in Desulfovibrionales abundance, following colistin exposure. Remarkably, short-chain fatty acids (SCFAs) analysis shows that propanoic acid contributes to the protective effect of melatonin on colistin-induced intestinal inflammation. Furthermore, the protection effects of melatonin and propanoic acid on LPS-induced cellular inflammation in RAW 264.7 cells are confirmed. Mechanistic investigations suggest that intervention with melatonin and propanoic acid can repress the activation of the TLR4 signal and its downstream NF-κB and MAPK signaling pathways, thereby mitigating the toxic effects of colistin. Our work highlights the unappreciated role of melatonin in preventing the potential detrimental effects of colistin on intestinal health and suggests a combined therapeutic strategy to effectively manage intestinal infectious diseases.


Sujet(s)
Colistine , Dysbiose , Microbiome gastro-intestinal , Mélatonine , Mélatonine/pharmacologie , Animaux , Souris , Colistine/effets indésirables , Dysbiose/induit chimiquement , Dysbiose/métabolisme , Dysbiose/microbiologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Cellules RAW 264.7 , Inflammation/métabolisme , Inflammation/traitement médicamenteux , Inflammation/induit chimiquement , Mâle , Antibactériens/pharmacologie , Souris de lignée C57BL
2.
FASEB J ; 38(13): e23766, 2024 Jul 15.
Article de Anglais | MEDLINE | ID: mdl-38967214

RÉSUMÉ

Dysbiosis of gut microbiota may account for pathobiology in simple fatty liver (SFL), metabolic dysfunction-associated steatohepatitis (MASH), fibrotic progression, and transformation to MASH-associated hepatocellular carcinoma (MASH-HCC). The aim of the present study is to investigate gut dysbiosis in this progression. Fecal microbial rRNA-16S sequencing, absolute quantification, histopathologic, and biochemical tests were performed in mice fed high fat/calorie diet plus high fructose and glucose in drinking water (HFCD-HF/G) or control diet (CD) for 2, 16 weeks, or 14 months. Histopathologic examination verified an early stage of SFL, MASH, fibrotic, or MASH-HCC progression with disturbance of lipid metabolism, liver injury, and impaired gut mucosal barrier as indicated by loss of occludin in ileum mucosa. Gut dysbiosis occurred as early as 2 weeks with reduced α diversity, expansion of Kineothrix, Lactococcus, Akkermansia; and shrinkage in Bifidobacterium, Lactobacillus, etc., at a genus level. Dysbiosis was found as early as MAHS initiation, and was much more profound through the MASH-fibrotic and oncogenic progression. Moreover, the expansion of specific species, such as Lactobacillus johnsonii and Kineothrix alysoides, was confirmed by an optimized method for absolute quantification. Dynamic alterations of gut microbiota were characterized in three stages of early SFL, MASH, and its HCC transformation. The findings suggest that the extent of dysbiosis was accompanied with MASH progression and its transformation to HCC, and the shrinking or emerging of specific microbial species may account at least in part for pathologic, metabolic, and immunologic alterations in fibrogenic progression and malignant transition in the liver.


Sujet(s)
Carcinome hépatocellulaire , Dysbiose , Microbiome gastro-intestinal , Tumeurs du foie , Souris de lignée C57BL , Animaux , Souris , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/microbiologie , Carcinome hépatocellulaire/anatomopathologie , Carcinome hépatocellulaire/étiologie , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Tumeurs du foie/microbiologie , Tumeurs du foie/étiologie , Dysbiose/microbiologie , Mâle , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Stéatose hépatique/microbiologie , Alimentation riche en graisse/effets indésirables , Modèles animaux de maladie humaine , Évolution de la maladie , Métabolisme lipidique , Foie/métabolisme , Foie/anatomopathologie
3.
J Transl Med ; 22(1): 628, 2024 Jul 05.
Article de Anglais | MEDLINE | ID: mdl-38970045

RÉSUMÉ

BACKGROUND: Bladder cancer is a common malignancy with high recurrence rate. Early diagnosis and recurrence surveillance are pivotal to patients' outcomes, which require novel minimal-invasive diagnostic tools. The urinary microbiome is associated with bladder cancer and can be used as biomarkers, but the underlying mechanism is to be fully illustrated and diagnostic performance to be improved. METHODS: A total of 23 treatment-naïve bladder cancer patients and 9 non-cancerous subjects were enrolled into the Before group and Control group. After surgery, 10 patients from the Before group were further assigned into After group. Void mid-stream urine samples were collected and sent for 16S rDNA sequencing, targeted metabolomic profiling, and flow cytometry. Next, correlations were analyzed between microbiota, metabolites, and cytokines. Finally, receiver operating characteristic (ROC) curves of the urinary biomarkers were plotted and compared. RESULTS: Comparing to the Control group, levels of IL-6 (p < 0.01), IL-8 (p < 0.05), and IL-10 (p < 0.05) were remarkably elevated in the Before group. The α diversity of urine microbiome was also significantly higher, with the feature microbiota positively correlated to the level of IL-6 (r = 0.58, p < 0.01). Significant differences in metabolic composition were also observed between the Before and Control groups, with fatty acids and fatty acylcarnitines enriched in the Before group. After tumor resection, cytokine levels and the overall microbiome structure in the After group remained similar to that of the Before group, but fatty acylcarnitines were significantly reduced (p < 0.05). Pathway enrichment analysis revealed beta-oxidation of fatty acids was significantly involved (p < 0.001). ROC curves showed that the biomarker panel of Actinomycetaceae + arachidonic acid + IL-6 had superior diagnostic performance, with sensitivity of 0.94 and specificity of 1.00. CONCLUSIONS: Microbiome dysbiosis, proinflammatory environment and altered fatty acids metabolism are involved in the pathogenesis of bladder cancer, which may throw light on novel noninvasive diagnostic tool development.


Sujet(s)
Dysbiose , Acides gras , Inflammation , Microbiote , Tumeurs de la vessie urinaire , Humains , Tumeurs de la vessie urinaire/microbiologie , Tumeurs de la vessie urinaire/urine , Inflammation/microbiologie , Mâle , Dysbiose/microbiologie , Dysbiose/urine , Adulte d'âge moyen , Femelle , Acides gras/métabolisme , Acides gras/urine , Courbe ROC , Cytokines/métabolisme , ARN ribosomique 16S/génétique , Sujet âgé , Études cas-témoins
4.
Nat Commun ; 15(1): 5697, 2024 Jul 07.
Article de Anglais | MEDLINE | ID: mdl-38972900

RÉSUMÉ

Climate and environmental changes threaten human mental health, but the impacts of specific environmental conditions on neuropsychiatric disorders remain largely unclear. Here, we show the impact of a humid heat environment on the brain and the gut microbiota using a conditioned housing male mouse model. We demonstrate that a humid heat environment can cause anxiety-like behaviour in male mice. Microbial 16 S rRNA sequencing analysis reveals that a humid heat environment caused gut microbiota dysbiosis (e.g., decreased abundance of Lactobacillus murinus), and metabolomics reveals an increase in serum levels of secondary bile acids (e.g., lithocholic acid). Moreover, increased neuroinflammation is indicated by the elevated expression of proinflammatory cytokines in the serum and cortex, activated PI3K/AKT/NF-κB signalling and a microglial response in the cortex. Strikingly, transplantation of the microbiota from mice reared in a humid heat environment readily recapitulates these abnormalities in germ-free mice, and these abnormalities are markedly reversed by Lactobacillus murinus administration. Human samples collected during the humid heat season also show a decrease in Lactobacillus murinus abundance and an increase in the serum lithocholic acid concentration. In conclusion, gut microbiota dysbiosis induced by a humid heat environment drives the progression of anxiety disorders by impairing bile acid metabolism and enhancing neuroinflammation, and probiotic administration is a potential therapeutic strategy for these disorders.


Sujet(s)
Anxiété , Acides et sels biliaires , Dysbiose , Microbiome gastro-intestinal , Température élevée , Animaux , Mâle , Souris , Acides et sels biliaires/métabolisme , Humains , Dysbiose/microbiologie , Anxiété/microbiologie , Souris de lignée C57BL , Humidité , Acide lithocholique/métabolisme , Lactobacillus , Encéphale/métabolisme , Facteur de transcription NF-kappa B/métabolisme , ARN ribosomique 16S/génétique , Modèles animaux de maladie humaine , Troubles anxieux/métabolisme , Troubles anxieux/microbiologie , Troubles anxieux/étiologie , Transduction du signal , Cytokines/métabolisme
5.
Lipids Health Dis ; 23(1): 207, 2024 Jun 29.
Article de Anglais | MEDLINE | ID: mdl-38951816

RÉSUMÉ

BACKGROUND: Ketogenic diets are increasingly popular for addressing obesity, but their impacts on the gut microbiota and metabolome remain unclear. This paper aimed to investigate how a ketogenic diet affects intestinal microorganisms and metabolites in obesity. METHODS: Male mice were provided with one of the following dietary regimens: normal chow, high-fat diet, ketogenic diet, or high-fat diet converted to ketogenic diet. Body weight and fat mass were measured weekly using high-precision electronic balances and minispec body composition analyzers. Metagenomics and non-targeted metabolomics data were used to analyze differences in intestinal contents. RESULTS: Obese mice on the ketogenic diet exhibited notable improvements in weight and body fat. However, these were accompanied by a significant decrease in intestinal microbial diversity, as well as an increase in Firmicutes abundance and a 247% increase in the Firmicutes/Bacteroidetes ratio. The ketogenic diet also altered multiple metabolic pathways in the gut, including glucose, lipid, energy, carbohydrate, amino acid, ketone body, butanoate, and methane pathways, as well as bacterial secretion and colonization pathways. These changes were associated with increased intestinal inflammation and dysbiosis in obese mice. Furthermore, the ketogenic diet enhanced the secretion of bile and the synthesis of aminoglycoside antibiotics in obese mice, which may impair the gut microbiota and be associated with intestinal inflammation and immunity. CONCLUSIONS: The study suggest that the ketogenic diet had an unfavorable risk-benefit trade-off and may compromise metabolic homeostasis in obese mice.


Sujet(s)
Alimentation riche en graisse , Régime cétogène , Microbiome gastro-intestinal , Métagénomique , Obésité , Régime cétogène/effets indésirables , Animaux , Mâle , Souris , Obésité/métabolisme , Obésité/microbiologie , Obésité/étiologie , Alimentation riche en graisse/effets indésirables , Métagénomique/méthodes , Métabolomique/méthodes , Dysbiose/microbiologie , Dysbiose/métabolisme , Souris de lignée C57BL , Métabolome , Poids
6.
BMC Microbiol ; 24(1): 235, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38956452

RÉSUMÉ

BACKGROUND: Patients with pancreatic ductal adenocarcinoma (PDAC) display an altered oral, gastrointestinal, and intra-pancreatic microbiome compared to healthy individuals. However, knowledge regarding the bile microbiome and its potential impact on progression-free survival in PDACs remains limited. METHODS: Patients with PDAC (n = 45), including 20 matched pairs before and after surgery, and benign controls (n = 16) were included prospectively. The characteristics of the microbiomes of the total 81 bile were revealed by 16  S-rRNA gene sequencing. PDAC patients were divided into distinct groups based on tumor marker levels, disease staging, before and after surgery, as well as progression free survival (PFS) for further analysis. Disease diagnostic model was formulated utilizing the random forest algorithm. RESULTS: PDAC patients harbor a unique and diverse bile microbiome (PCoA, weighted Unifrac, p = 0.038), and the increasing microbial diversity is correlated with dysbiosis according to key microbes and microbial functions. Aliihoeflea emerged as the genus displaying the most significant alteration among two groups (p < 0.01). Significant differences were found in beta diversity of the bile microbiome between long-term PFS and short-term PFS groups (PCoA, weighted Unifrac, p = 0.005). Bacillota and Actinomycetota were identified as altered phylum between two groups associated with progression-free survival in all PDAC patients. Additionally, we identified three biomarkers as the most suitable set for the random forest model, which indicated a significantly elevated likelihood of disease occurrence in the PDAC group (p < 0.0001). The area under the receiver operating characteristic (ROC) curve reached 80.8% with a 95% confidence interval ranging from 55.0 to 100%. Due to the scarcity of bile samples, we were unable to conduct further external verification. CONCLUSION: PDAC is characterized by an altered microbiome of bile ducts. Biliary dysbiosis is linked with progression-free survival in all PDACs. This study revealed the alteration of the bile microbiome in PDACs and successfully developed a diagnostic model for PDAC.


Sujet(s)
Bile , Carcinome du canal pancréatique , Microbiote , Tumeurs du pancréas , Humains , Carcinome du canal pancréatique/microbiologie , Carcinome du canal pancréatique/mortalité , Carcinome du canal pancréatique/anatomopathologie , Bile/microbiologie , Mâle , Femelle , Tumeurs du pancréas/microbiologie , Tumeurs du pancréas/mortalité , Tumeurs du pancréas/anatomopathologie , Microbiote/génétique , Adulte d'âge moyen , Sujet âgé , Dysbiose/microbiologie , Survie sans progression , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Études prospectives , ARN ribosomique 16S/génétique
7.
J Infect Dev Ctries ; 18(6): 909-918, 2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-38990992

RÉSUMÉ

INTRODUCTION: Despite the numerous studies demonstrating gut microbiota dysbiosis in obese subjects, there is no data on the association between obesity and gastric microbiota. The aim of this study was to address this gap in literature by comparing the composition of gastric microbiota in obese patients and a control group which included normal weight volunteers diagnosed with functional dyspepsia (FD). METHODOLOGY: A total of 19 obese patients, and 18 normal weight subjects with FD and normal endoscopy results were included in the study. The gastric tissue samples were collected from participants in both groups by bariatric surgery and endoscopy, respectively, and profiled using 16S ribosomal RNA gene sequencing. RESULTS: There was no significant difference in the α-diversity scores, while distinct gastric microbial compositions were detected in both groups. Significantly lower levels of Bacteroidetes and Fusobacteria, and higher Firmicutes/Bacteroidetes ratio were recorded in the obese patients. A total of 15 bacterial genera exhibited significant difference in gastric abundance with Prevotella_7, Veillonella, Cupriavidus, and Acinetobacter, present in frequencies higher than 3% in at least one subject group. CONCLUSIONS: Our study suggests a significant association between obesity and gastric microbiome composition. Future studies with larger sample size and gastric samples from subjects without any gastrointestinal complications are required to confirm our conclusions.


Sujet(s)
Dyspepsie , Microbiome gastro-intestinal , Obésité , ARN ribosomique 16S , Humains , Dyspepsie/microbiologie , Obésité/microbiologie , Obésité/complications , Adulte , Mâle , Femelle , ARN ribosomique 16S/génétique , Adulte d'âge moyen , Estomac/microbiologie , Dysbiose/microbiologie , Bactéries/classification , Bactéries/isolement et purification , Bactéries/génétique , Jeune adulte
8.
Cell Host Microbe ; 32(7): 1048-1049, 2024 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-38991502

RÉSUMÉ

Gestational diabetes mellitus (GDM) is associated with increased risk of metabolic and neurodevelopmental disorders in offspring. In this issue of Cell Host & Microbe, Wang et al. provide evidence that changes in the gut microbiome of mothers with GDM may lead to dysbiosis in their infants and altered development in a sex-dependent manner.


Sujet(s)
Diabète gestationnel , Dysbiose , Microbiome gastro-intestinal , Diabète gestationnel/microbiologie , Diabète gestationnel/métabolisme , Grossesse , Microbiome gastro-intestinal/physiologie , Humains , Femelle , Dysbiose/microbiologie , Nourrisson , Mâle , Nouveau-né
9.
Gan To Kagaku Ryoho ; 51(6): 597-602, 2024 Jun.
Article de Japonais | MEDLINE | ID: mdl-39009513

RÉSUMÉ

In recent years, the human microbiota, especially the gut microbiota, has been attracting attention in various fields, and it is one of the topics in the field of oncology. The human microbiota is known to act directly or indirectly on host immunity, and the gut and lung microbiota influence each other through the"gut-lung axis". It has been suggested that dysbiosis, a condition in which the symbiosis of the human microbiota is disrupted, induces lung inflammation and various respiratory diseases, and is also implicated in the immune microenvironment of lung cancer. It is also widely known that the gut microbiota modulates the efficacy of cancer immunotherapy, a major pillar of lung cancer treatment, and many clinical trials targeting the gut microbiota, such as fecal microbiome transplantation and biotics intervention, are currently being conducted. In the future, research on lung carcinogenesis mechanisms and lung cancer treatment focusing on the human microbiota will become increasingly active.


Sujet(s)
Microbiome gastro-intestinal , Immunothérapie , Tumeurs du poumon , Animaux , Humains , Carcinogenèse/immunologie , Dysbiose/immunologie , Dysbiose/thérapie , Dysbiose/microbiologie , Microbiome gastro-intestinal/immunologie , Immunothérapie/méthodes , Tumeurs du poumon/immunologie , Tumeurs du poumon/thérapie , Tumeurs du poumon/microbiologie
10.
Gan To Kagaku Ryoho ; 51(6): 608-612, 2024 Jun.
Article de Japonais | MEDLINE | ID: mdl-39009515

RÉSUMÉ

The gut microbiome is involved in host physiology, including nutrition, metabolism, and immunity. It was recently known that dysbiosis of the gut microbiome has been implicated in several human diseases such as inflammatory bowel disease. It is altered by environmental factors such as diet, habit and lifestyle and has been directly and indirectly linked to the development and progression of colorectal cancer(CRC). Fusobacterium(F.)nucleatum, which causes periodontal disease, has been shown to play an important role in the initiation and development of CRC, although not as clearly as Helicobacter(H.) pylori in gastric cancer. Therefore, gut bacteria hold promise as a potential therapeutic approach to prevent or treat CRC. Although its clinical usefulness has not yet been demonstrated, future research such as metagenomics may open new avenues for CRC treatment with gut bacteria. Here, we reviewed the role of the gut microbial community in the development, progression, and prevention of colorectal carcinogenesis.


Sujet(s)
Tumeurs colorectales , Microbiome gastro-intestinal , Humains , Tumeurs colorectales/microbiologie , Tumeurs colorectales/thérapie , Animaux , Dysbiose/microbiologie
11.
Benef Microbes ; 15(4): 397-410, 2024 Jul 03.
Article de Anglais | MEDLINE | ID: mdl-38955351

RÉSUMÉ

Previous studies reporting the association between gut microbiota dysbiosis and maternal obesity were mostly confined at the phylum level or at postpartum period. This study aimed to investigate the dynamic changes in gut microbial communities associated with maternal obesity at different time points of pregnancy. We performed 16S rRNA gene V3-V4 amplicon sequencing on stool samples from 110 women in all three trimesters and 1-month postpartum. Maternal gut microbial communities associated with maternal pre-pregnancy body mass index (BMI) and gestational weight gain (GWG) were explored. The influence of maternal obesity on gut microbiota trajectories was determined based on longitudinal shifts in community clusters across the trimesters. The richness index of alpha diversity decreased with the progression of pregnancy, particularly in women with excessive GWG. The evenness index in 2nd trimester was found inversely associated with GWG. Various taxonomic differences in 1st trimester were associated with excessive GWG, whereas limited taxonomic differences in 2nd and 3rd trimesters were associated with pre-pregnancy BMI or GWG. Meanwhile, the gut microbiota trajectory with especially depleted genus Faecalibacterium in 1st trimester was associated with excessive GWG (adjusted odds ratio 5.7, 95% confidence interval 1.2-28.1). Moreover, the longitudinal abundances of genus Lachnospiraceae ND3007 group across gestations were depleted in women with overweight/obese pre-pregnancy BMI, while genus Bifidobacterium enriched in women with excessive GWG. Our study shows that dysbiosis of the gut microbiota in early pregnancy may have a significant impact on excess GWG. The abundance of the genus Faecalibacterium in 1st trimester may be a potential risk factor. Clinical trial number: NCT03785093 (https://classic.clinicaltrials.gov/ct2/show/NCT03785093).


Sujet(s)
Indice de masse corporelle , Dysbiose , Fèces , Microbiome gastro-intestinal , Prise de poids pendant la grossesse , ARN ribosomique 16S , Humains , Femelle , Grossesse , Adulte , Fèces/microbiologie , ARN ribosomique 16S/génétique , Dysbiose/microbiologie , Bactéries/classification , Bactéries/génétique , Bactéries/isolement et purification , Obésité/microbiologie , Jeune adulte , Obésité maternelle , Faecalibacterium/génétique
12.
Environ Microbiol ; 26(7): e16676, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39010309

RÉSUMÉ

Just as the human gut microbiome is colonized by a variety of microbes, so too is the rhizosphere of plants. An imbalance in this microbial community, known as dysbiosis, can have a negative impact on plant health. This study sought to explore the effect of rhizosphere dysbiosis on the health of tomato plants (Solanum lycopersicum L.), using them and the foliar bacterial spot pathogen Xanthomonas perforans as model organisms. The rhizospheres of 3-week-old tomato plants were treated with either streptomycin or water as a control, and then spray-inoculated with X. perforans after 24 h. Half of the plants that were treated with both streptomycin and X. perforans received soil microbiome transplants from uninfected plant donors 48 h after the streptomycin was applied. The plants treated with streptomycin showed a 26% increase in disease severity compared to those that did not receive the antibiotic. However, the plants that received the soil microbiome transplant exhibited an intermediate level of disease severity. The antibiotic-treated plants demonstrated a reduced abundance of rhizobacterial taxa such as Cyanobacteria from the genus Cylindrospermum. They also showed a down-regulation of genes related to plant primary and secondary metabolism, and an up-regulation of plant defence genes associated with induced systemic resistance. This study highlights the vital role that beneficial rhizosphere microbes play in disease resistance, even against foliar pathogens.


Sujet(s)
Dysbiose , Maladies des plantes , Rhizosphère , Microbiologie du sol , Solanum lycopersicum , Transcriptome , Maladies des plantes/microbiologie , Dysbiose/microbiologie , Solanum lycopersicum/microbiologie , Xanthomonas/génétique , Feuilles de plante/microbiologie , Microbiote , Résistance à la maladie/génétique , Racines de plante/microbiologie , Antibactériens/pharmacologie , Streptomycine/pharmacologie
13.
Pol Merkur Lekarski ; 52(3): 300-303, 2024.
Article de Anglais | MEDLINE | ID: mdl-39007468

RÉSUMÉ

OBJECTIVE: Aim: The aim of the study was to determine the quantitative and qualitative characteristics of the microbiota of dento-gingival plaque in children to improve the quality of treatment of chronic catarrhal gingivitis. PATIENTS AND METHODS: Materials and Methods: It was examined 16 children aged 9-16 years with a diagnosis of K05.1: chronic gingivitis and 10 persons with intact gums were taken as a comparison group. A clinical dental examination was performed on the study participants and a sample was taken to determine the bacteria in the periodontal plaque. RESULTS: Results: The results of statistical processing of the research data allowed us to establish that in patients with chronic gingivitis, quantitative indicators of the total bacterial mass, Lactobacillus spp., Enterobacteriaceae, Gardnerella vaginalis/Prevotella bivia/Porphyromonas spp. in the sample of periodontal plaque significantly exceeded the indicators of healthy patients. It was determined that the examined children with chronic gingivitis, the total number of Lactobacillus spp. significantly exceeds its amount in people with intact gums. CONCLUSION: Conclusions: The changes in the quantitative and qualitative characteristics of the main representatives of the microf i lm of dento-gingival plaque, which characterize dysbiosis, are of signif i cant clinical signif i cance. Study of the quantitative characteristics of Lactobacterium spp., Enterobacterium spp., Streptococcacea spp., Gardnerella spp., Prevotella spp., Porphyromonas spp., Eubacteridacea spp., Mycoplasma (hominis + genitalium), Candida spp. is a diagnostic factor in determining the condition of the mucous membrane of the oral cavity.


Sujet(s)
Dysbiose , Gingivite , Humains , Enfant , Gingivite/microbiologie , Gingivite/diagnostic , Adolescent , Dysbiose/microbiologie , Femelle , Mâle , Maladie chronique , Plaque dentaire/microbiologie , Réaction de polymérisation en chaine en temps réel , Microbiote , Réaction de polymérisation en chaine multiplex
14.
Int J Mol Sci ; 25(13)2024 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-39000333

RÉSUMÉ

Breast cancer represents the most prevalent form of cancer and the leading cause of cancer-related mortality among females worldwide. It has been reported that several risk factors contribute to the appearance and progression of this disease. Despite the advancements in breast cancer treatment, a significant portion of patients with distant metastases still experiences no cure. The extracellular matrix represents a potential target for enhanced serum biomarkers in breast cancer. Furthermore, extracellular matrix degradation and epithelial-mesenchymal transition constitute the primary stages of local invasion during tumorigenesis. Additionally, the microbiome has a potential influence on diverse physiological processes. It is emerging that microbial dysbiosis is a significant element in the development and progression of various cancers, including breast cancer. Thus, a better understanding of extracellular matrix and microbiome interactions could provide novel alternatives to breast cancer treatment and management. In this review, we summarize the current evidence regarding the intricate relationship between breast cancer with the extracellular matrix and the microbiome. We discuss the arising associations and future perspectives in this field.


Sujet(s)
Tumeurs du sein , Matrice extracellulaire , Microbiote , Humains , Tumeurs du sein/microbiologie , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Matrice extracellulaire/métabolisme , Femelle , Transition épithélio-mésenchymateuse , Animaux , Dysbiose/microbiologie , Microenvironnement tumoral
15.
J Extracell Vesicles ; 13(7): e12474, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-39001704

RÉSUMÉ

Gut microbiome dysbiosis is a major contributing factor to several pathological conditions. However, the mechanistic understanding of the communication between gut microbiota and extra-intestinal organs remains largely elusive. Extracellular vesicles (EVs), secreted by almost every form of life, including bacteria, could play a critical role in this inter-kingdom crosstalk and are the focus of present study. Here, we present a novel approach for isolating lipopolysaccharide (LPS)+ bacterial extracellular vesicles (bEVLPS) from complex biological samples, including faeces, plasma and the liver from lean and diet-induced obese (DIO) mice. bEVLPS were extensively characterised using nanoparticle tracking analyses, immunogold labelling coupled with transmission electron microscopy, flow cytometry, super-resolution microscopy and 16S sequencing. In liver tissues, the protein expressions of TLR4 and a few macrophage-specific biomarkers were assessed by immunohistochemistry, and the gene expressions of inflammation-related cytokines and their receptors (n = 89 genes) were measured using a PCR array. Faecal samples from DIO mice revealed a remarkably lower concentration of total EVs but a significantly higher percentage of LPS+ EVs. Interestingly, DIO faecal bEVLPS showed a higher abundance of Proteobacteria by 16S sequencing. Importantly, in DIO mice, a higher number of total EVs and bEVLPS consistently entered the hepatic portal vein and subsequently reached the liver, associated with increased expression of TLR4, macrophage markers (F4/80, CD86 and CD206), cytokines and receptors (Il1rn, Ccr1, Cxcl10, Il2rg and Ccr2). Furthermore, a portion of bEVLPS escaped liver and entered the peripheral circulation. In conclusion, bEV could be the key mediator orchestrating various well-established biological effects induced by gut bacteria on distant organs.


Sujet(s)
Vésicules extracellulaires , Microbiome gastro-intestinal , Lipopolysaccharides , Foie , Veine porte , Animaux , Vésicules extracellulaires/métabolisme , Foie/métabolisme , Lipopolysaccharides/métabolisme , Souris , Veine porte/métabolisme , Souris de lignée C57BL , Mâle , Bactéries/métabolisme , Récepteur de type Toll-4/métabolisme , Obésité/métabolisme , Obésité/microbiologie , Fèces/microbiologie , Dysbiose/métabolisme , Dysbiose/microbiologie
16.
Theranostics ; 14(9): 3760-3776, 2024.
Article de Anglais | MEDLINE | ID: mdl-38948060

RÉSUMÉ

Rationale: Currently, there are occasional reports of health problems caused by sleep deprivation (SD). However, to date, there remains a lack of in-depth research regarding the effects of SD on the growth and development of oocytes in females. The present work aimed to investigate whether SD influences ovarian folliculogenesis in adolescent female mice. Methods: Using a dedicated device, SD conditions were established in 3-week old female mice (a critical stage of follicular development) for 6 weeks and gut microbiota and systemic metabolomics were analyzed. Analyses were related to parameters of folliculogenesis and reproductive performance of SD females. Results: We found that the gut microbiota and systemic metabolomics were severely altered in SD females and that these were associated with parameters of premature ovarian insufficiency (POI). These included increased granulosa cell apoptosis, reduced numbers of primordial follicles (PmFs), correlation with decreased AMH, E2, and increased LH in blood serum, and a parallel increased number of growing follicles and changes in protein expression compatible with PmF activation. SD also reduced oocyte maturation and reproductive performance. Notably, fecal microbial transplantation from SD females into normal females induced POI parameters in the latter while niacinamide (NAM) supplementation alleviated such symptoms in SD females. Conclusion: Gut microbiota and alterations in systemic metabolomics caused by SD induced POI features in juvenile females that could be counteracted with NAM supplementation.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Métabolomique , Insuffisance ovarienne primitive , Privation de sommeil , Animaux , Femelle , Insuffisance ovarienne primitive/métabolisme , Souris , Dysbiose/microbiologie , Dysbiose/métabolisme , Métabolomique/méthodes , Privation de sommeil/complications , Privation de sommeil/métabolisme , Follicule ovarique/métabolisme , Ovocytes/métabolisme , Transplantation de microbiote fécal , Modèles animaux de maladie humaine , Apoptose
17.
Nutrients ; 16(13)2024 Jun 21.
Article de Anglais | MEDLINE | ID: mdl-38999736

RÉSUMÉ

The etiology and mechanisms of autism and autism spectrum disorder (ASD) are not yet fully understood. There is currently no treatment for ASD for providing significant improvement in core symptoms. Recent studies suggest, however, that ASD is associated with gut dysbiosis, indicating that modulation of gut microbiota in children with ASD may thus reduce the manifestation of ASD symptoms. The aim of this pilot study (prospective randomized, double-blinded, placebo-controlled) was to evaluate efficacy of the biological response modifier Juvenil in modulating the microbiome of children with ASD and, in particular, whether Juvenil is able to alleviate the symptoms of ASD. In total, 20 children with ASD and 12 neurotypical children were included in our study. Supplementation of ASD children lasted for three months. To confirm Juvenil's impact on the gut microbiome, stool samples were collected from all children and the microbiome's composition was analyzed. This pilot study demonstrated that the gut microbiome of ASD children differed significantly from that of healthy controls and was converted by Juvenil supplementation toward a more neurotypical microbiome that positively modulated children's autism symptoms.


Sujet(s)
Trouble du spectre autistique , Compléments alimentaires , Microbiome gastro-intestinal , Humains , Projets pilotes , Méthode en double aveugle , Mâle , Femelle , Trouble du spectre autistique/microbiologie , Enfant , Fèces/microbiologie , Enfant d'âge préscolaire , Études prospectives , Trouble autistique/microbiologie , Dysbiose/microbiologie
18.
Int J Mol Sci ; 25(13)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-39000013

RÉSUMÉ

Obesity is a global health concern implicated in numerous chronic degenerative diseases, including type 2 diabetes, dyslipidemia, and neurodegenerative disorders. It is characterized by chronic low-grade inflammation, gut microbiota dysbiosis, insulin resistance, glucose intolerance, and lipid metabolism disturbances. Here, we investigated the therapeutic potential of environmental enrichment (EE) to prevent the progression of gut dysbiosis in mice with high-fat diet (HFD)-induced metabolic syndrome. C57BL/6 male mice with obesity and metabolic syndrome, continuously fed with an HFD, were exposed to EE. We analyzed the gut microbiota of the mice by sequencing the 16s rRNA gene at different intervals, including on day 0 and 12 and 24 weeks after EE exposure. Fasting glucose levels, glucose tolerance, insulin resistance, food intake, weight gain, lipid profile, hepatic steatosis, and inflammatory mediators were evaluated in serum, adipose tissue, and the colon. We demonstrate that EE intervention prevents the progression of HFD-induced dysbiosis, reducing taxa associated with metabolic syndrome (Tepidimicrobium, Acidaminobacteraceae, and Fusibacter) while promoting those linked to healthy physiology (Syntrophococcus sucrumutans, Dehalobacterium, Prevotella, and Butyricimonas). Furthermore, EE enhances intestinal barrier integrity, increases mucin-producing goblet cell population, and upregulates Muc2 expression in the colon. These alterations correlate with reduced systemic lipopolysaccharide levels and attenuated colon inflammation, resulting in normalized glucose metabolism, diminished adipose tissue inflammation, reduced liver steatosis, improved lipid profiles, and a significant reduction in body weight gain despite mice's continued HFD consumption. Our findings highlight EE as a promising anti-inflammatory strategy for managing obesity-related metabolic dysregulation and suggest its potential in developing probiotics targeting EE-modulated microbial taxa.


Sujet(s)
Alimentation riche en graisse , Dysbiose , Microbiome gastro-intestinal , Souris de lignée C57BL , Obésité , Animaux , Alimentation riche en graisse/effets indésirables , Dysbiose/microbiologie , Souris , Obésité/métabolisme , Obésité/microbiologie , Mâle , Glucose/métabolisme , Souris obèse , Insulinorésistance , Syndrome métabolique X/métabolisme , Syndrome métabolique X/étiologie , Syndrome métabolique X/microbiologie
19.
Sci Rep ; 14(1): 16158, 2024 Jul 12.
Article de Anglais | MEDLINE | ID: mdl-38997299

RÉSUMÉ

Juvenile dermatomyositis (JDM) is a rare immune-mediated disease of childhood with putative links to microbial exposures. In this multi-center, prospective, observational cohort study, we evaluated whether JDM is associated with discrete oral and gut microbiome signatures. We generated 16S rRNA sequencing data from fecal, saliva, supragingival, and subgingival plaque samples from JDM probands (n = 28). To control for genetic and environmental determinants of microbiome community structure, we also profiled microbiomes of unaffected family members (n = 27 siblings, n = 26 mothers, and n = 17 fathers). Sample type (oral-vs-fecal) and nuclear family unit were the predominant variables explaining variance in microbiome diversity, more so than having a diagnosis of JDM. The oral and gut microbiomes of JDM probands were more similar to their own unaffected siblings than they were to the microbiomes of other JDM probands. In a sibling-paired within-family analysis, several potentially immunomodulatory bacterial taxa were differentially abundant in the microbiomes of JDM probands compared to their unaffected siblings, including Faecalibacterium (gut) and Streptococcus (oral cavity). While microbiome features of JDM are often shared by unaffected family members, the loss or gain of specific fecal and oral bacteria may play a role in disease pathogenesis or be secondary to immune dysfunction in susceptible individuals.


Sujet(s)
Dermatomyosite , Fèces , Microbiome gastro-intestinal , Bouche , ARN ribosomique 16S , Humains , Fèces/microbiologie , Dermatomyosite/microbiologie , Dermatomyosite/génétique , Femelle , Mâle , Enfant , Bouche/microbiologie , ARN ribosomique 16S/génétique , Microbiome gastro-intestinal/génétique , Études prospectives , Dysbiose/microbiologie , Microbiote/génétique , Enfant d'âge préscolaire , Adolescent , Salive/microbiologie , Adulte
20.
Aging (Albany NY) ; 16(13): 10882-10904, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38968172

RÉSUMÉ

BACKGROUND: Chronic heart failure (CHF) impairs cognitive function, yet its effects on brain structure and underlying mechanisms remain elusive. This study aims to explore the mechanisms behind cognitive impairment. METHODS: CHF models in rats were induced by ligation of the left anterior descending coronary artery. Cardiac function was analyzed by cardiac ultrasound and hemodynamics. ELISA, immunofluorescence, Western blot, Golgi staining and transmission electron microscopy were performed on hippocampal tissues. The alterations of intestinal flora under the morbid state were investigated via 16S rRNA sequencing. The connection between neuroinflammation and synapses is confirmed by a co-culture system of BV2 microglia and HT22 cells in vitro. Results: CHF rats exhibited deteriorated cognitive behaviors. CHF induced neuronal structural disruption, loss of Nissl bodies, and synaptic damage, exhibiting alterations in multiple parameters. CHF rats showed increased hippocampal levels of inflammatory cytokines and activated microglia and astrocytes. Furthermore, the study highlights dysregulated PDE4-dependent cAMP signaling and intestinal flora dysbiosis, closely associated with neuroinflammation, and altered synaptic proteins. In vitro, microglial neuroinflammation impaired synaptic plasticity via PDE4-dependent cAMP signaling. CONCLUSIONS: Neuroinflammation worsens CHF-related cognitive impairment through neuroplasticity disorder, tied to intestinal flora dysbiosis. PDE4 emerges as a potential therapeutic target. These findings provide insightful perspectives on the heart-gut-brain axis.


Sujet(s)
Dysfonctionnement cognitif , Dysbiose , Microbiome gastro-intestinal , Défaillance cardiaque , Maladies neuro-inflammatoires , Plasticité neuronale , Animaux , Défaillance cardiaque/microbiologie , Défaillance cardiaque/physiopathologie , Dysfonctionnement cognitif/microbiologie , Dysbiose/microbiologie , Rats , Mâle , Hippocampe/métabolisme , Hippocampe/anatomopathologie , Rat Sprague-Dawley , Modèles animaux de maladie humaine , Maladie chronique , Microglie/métabolisme
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE