Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
2.
J Gastroenterol Hepatol ; 39(2): 280-288, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37961007

ABSTRACT

BACKGROUND AND AIM: Serotonin affects the balance and integrity of the gut microbiome; however, studies have confirmed the influence of selective serotonin reuptake inhibitors (SSRIs) on irritable bowel syndrome (IBS). We evaluated the association between SSRI use and subsequent IBS occurrence in a real-world setting. METHODS: A multivariate Cox proportional hazard model was adopted, and the National Health Insurance Service cohort claims database between 2010 and 2019 was used. Non-SSRI users were selected using the propensity score matching method. Subgroup analyses were performed using the point of use, cumulative dose, and duration of SSRI use. Additional analysis was performed using a control group without psychiatric medications. RESULTS: We included 2901 SSRI users and 2727 non-SSRI users. After adjusting covariates, the risk of developing IBS in SSRI users was 1.54 times that in non-SSRI users (95% confidence interval [CI]: 1.01-2.33). The hazard ratio (HR) of the recent, heavy, and short-term user groups were 3.19 (95% CI: 2.03-4.99), 2.22 (95% CI: 1.50-3.29), and 4.83 (95% CI: 3.02-7.73), respectively, compared with that of non-users. In patients without a history of psychiatric medications, the risk of IBS incidence after SSRI use increased significantly (HR: 1.61, 95% CI: 1.06-2.42), whereas HR was insignificant in patients with a history of psychiatric medications (HR: 1.25, 95% CI: 0.98-1.60). CONCLUSIONS: The risk of subsequent IBS occurrence following SSRI use was high in patients who initially took a heavy SSRI dose and those who did not have a history of psychiatric drug use.


Subject(s)
Irritable Bowel Syndrome , Selective Serotonin Reuptake Inhibitors , Humans , Selective Serotonin Reuptake Inhibitors/adverse effects , Retrospective Studies , Irritable Bowel Syndrome/chemically induced , Irritable Bowel Syndrome/epidemiology , Irritable Bowel Syndrome/drug therapy , Incidence , Proportional Hazards Models
3.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-1012717

ABSTRACT

Chronic constipation (CC) is one of the most common functional gastrointestinal diseases. At present, the overall therapeutic effect of CC is still not satisfactory worldwide, which seriously affects the quality of life and social function of patients. The etiology and pathophysiological mechanism of constipation are still unclear. It involves comprehensive factors such as heredity, social psychology, diet, intestinal flora imbalance, intestinal motility disorder, visceral sensitivity change, pelvic floor muscle group dysfunction and enteric nervous system (ENS) disorder. Among them, the abnormal factors of the brain-gut-microbiome axis are particularly significant. The brain-gut-microbiome axis is a complex network of interactions between the intestine and the brain, integrating and coordinating the physiological functions and pathological processes of the gastrointestinal tract. Microorganisms in the intestinal lumen play an important role in it, and can communicate with the intestinal tract and the central nervous system through nerve, endocrine and immune pathways. As a key brain-gut peptide in the regulation pathway of the brain-gut-microbiome axis, 5-hydroxytryptamine (5-HT) is involved in the regulation of gastrointestinal motility, sensation and secretion. The abnormal conduction of the 5-HT signaling pathway is closely related to the occurrence and development of constipation. Traditional Chinese medicine(TCM) is a unique precious resource in China, which has good curative effects and safety. In recent years, it has received extensive attention in the treatment of constipation. TCM and active ingredients, acupuncture and massage specifically regulate 5-HT signal transmission, so that the expressions of related molecules tend to be suitable for individual disease state levels to effectively improve constipation symptoms, with unique advantages. Therefore, this study used ''constipation'', ''intestinal flora'', ''5-HT'', and ''traditional Chinese medicine'' as the keywords to search PubMed, China National Knowledge Infrastructure (CNKI) and other literature databases. The correlation between 5-HT and constipation as well as brain-gut-microbiome axis and the research progress of TCM intervention in the 5-HT signaling pathway in the treatment of constipation were reviewed in order to explore the potential therapeutic value of 5-HT system in this disease and provide references for subsequent research.

4.
Appl Environ Microbiol ; 89(10): e0105023, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37815342

ABSTRACT

Comamonas testosteroni TA441 is capable of aerobically degrading steroids through the aromatization and cleavage of the A- and B-rings, followed by D- and C-ring cleavage via ß-oxidation. While most of the degradation steps have been previously characterized, a few intermediate compounds remained unidentified. In this study, we proposed that the cleavage of the D-ring at C13-17 required the ScdY hydratase, followed by C-ring cleavage via the ScdL1L2 transferase. The anticipated reaction was expected to yield 6-methyl-3,7-dioxo-decane-1,10-dioic acid-coenzyme A (CoA) ester. To confirm this hypothesis, we constructed a plasmid enabling the induction of targeted genes in TA441 mutant strains. Induction experiments of ScdL1L2 revealed that the major product was 3-hydroxy-6-methyl-7-oxo-decane-1,10-dioic acid-CoA ester. Similarly, induction experiments of ScdY demonstrated that the substrate of ScdY was a geminal diol, 17-dihydroxy-9-oxo-1,2,3,4,5,6,10,19-octanorandrost-8(14)-en-7-oic acid-CoA ester. These findings suggest that ScdY catalyzes the addition of a water molecule at C14 of 17-dihydroxy-9-oxo-1,2,3,4,5,6,10,19-octanorandrost-8(14)-en-7-oic acid-CoA ester, leading to D-ring cleavage at C13-17. Subsequently, the C9 ketone of the D-ring cleavage product is converted to a hydroxyl group, followed by C-ring cleavage, resulting in the production of 3-hydroxy-6-methyl-7-oxo-decane-1,10-dioic acid-CoA ester.IMPORTANCEStudies on bacterial steroid degradation were initiated more than 50 years ago primarily to obtain substrates for steroid drugs. In recent years, the role of steroid-degrading bacteria in relation to human health has gained significant attention, as emerging evidence suggests that the intestinal microflora plays a crucial role in human health. Furthermore, cholic acid, a major component of bile acid secreted in the intestines, is closely associated with the gut microbiota. While Comamonas testosteroni TA441 is recognized as the leading bacterial model for aerobic steroid degradation, the involvement of aerobic steroid degradation in the intestinal microflora remains largely unexplored. Nonetheless, the presence of C. testosteroni in the cecum suggests the potential influence of aerobic steroid degradation on gut microbiota. To establish essential information about the role of these bacteria, here, we identified the missing compounds and propose more details of C-, and D-ring cleavage, which have remained unclear until now.


Subject(s)
Comamonas testosteroni , Humans , Comamonas testosteroni/metabolism , Steroids/metabolism , Oxidation-Reduction , Esters/metabolism
5.
Appl Environ Microbiol ; 89(10): e0014323, 2023 10 31.
Article in English | MEDLINE | ID: mdl-37815361

ABSTRACT

Comamonas testosteroni is one of the representative aerobic steroid-degrading bacteria. We previously revealed the mechanism of steroidal A,B,C,D-ring degradation by C. testosteroni TA441. The corresponding genes are located in two clusters at both ends of a mega-cluster of steroid degradation genes. ORF7 and ORF6 are the only two genes in these clusters, whose function has not been determined. Here, we characterized ORF7 as encoding the dehydrase responsible for converting the C12ß hydroxyl group to the C10(12) double bond on the C-ring (SteC), and ORF6 as encoding the hydrogenase responsible for converting the C10(12) double bond to a single bond (SteD). SteA and SteB, encoded just upstream of SteC and SteD, are in charge of oxidizing the C12α hydroxyl group to a ketone group and of reducing the latter to the C12ß hydroxyl group, respectively. Therefore, the C12α hydroxyl group in steroids is removed with SteABCD via the C12 ketone and C12ß hydroxyl groups. Given the functional characterization of ORF6 and ORF7, we disclose the entire pathway of steroidal A,B,C,D-ring breakdown by C. testosteroni TA441.IMPORTANCEStudies on bacterial steroid degradation were initiated more than 50 years ago, primarily to obtain materials for steroid drugs. Now, their implications for the environment and humans, especially in relation to the infection and the brain-gut-microbiota axis, are attracting increasing attention. Comamonas testosteroni TA441 is the leading model of bacterial aerobic steroid degradation with the ability to break down cholic acid, the main component of bile acids. Bile acids are known for their variety of physiological activities according to their substituent group(s). In this study, we identified and functionally characterized the genes for the removal of C12 hydroxyl groups and provided a comprehensive summary of the entire A,B,C,D-ring degradation pathway by C. testosteroni TA441 as the representable bacterial aerobic degradation process of the steroid core structure.


Subject(s)
Comamonas testosteroni , Humans , Comamonas testosteroni/genetics , Comamonas testosteroni/metabolism , Oxidoreductases/metabolism , Steroids/metabolism , Cholic Acid/metabolism , Ketones/metabolism
6.
Biomedicines ; 11(6)2023 Jun 20.
Article in English | MEDLINE | ID: mdl-37371865

ABSTRACT

The relationship between the gut-brain-microbiome axis has gained great importance in the study of psychiatric disorders, as it may represent a new target for their treatment. To date, the available literature suggests that the microbiota may influence the pathophysiology of several diseases, including psychosis. The aim of this review is to summarize the clinical and preclinical studies that have evaluated the differences in microbiota as well as the metabolic consequences related to psychosis. Current data suggest that the genera Lactobacillus and Megasphaera are increased in schizophrenia (SZ), as well as alterations in the glutamate-glutamine-GABA cycle, serum levels of tryptophan, kynurenic acid (KYNA), and short-chain fatty acids (SCFAs). There are still very few studies on early-onset psychosis, thus more studies are needed to be able to propose targeted therapies for a point when the disease has just started or has not yet progressed.

7.
Brain Res Bull ; 199: 110665, 2023 07.
Article in English | MEDLINE | ID: mdl-37192716

ABSTRACT

The brain-gut-microbiome axis (BGMA) is a pivotal contributor to human health. A large body of research, especially from animal models, has revealed bidirectional, causal relationships between the BGMA and sex. In particular, sex steroids appear to be affected by the BGMA, to influence the BGMA, and to moderate environmental effects on the BGMA. However, animal research on the relationship between sex and the BGMA has not translated well to human models. We contend that this is due in part to an oversimplified approach to sex: although BGMA researchers have traditionally approached sex as a unidimensional, dichotomous variable, it is in fact multidimensional and is comprised of both multi-categorical and continuous dimensions. We also contend that research on the BGMA in humans should approach gender as a variable that is distinct from sex and that gender may influence the BGMA through pathways that are independent from the effects of sex alone. Research practices that consider the complexity and distinctiveness of sex and gender in relation to the human BGMA will not only yield improved understanding of this consequential system, but will also enhance the development of treatments for adverse health outcomes with BGMA-related etiologies. We conclude with recommendations for the implementation of such practices.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Male , Animals , Female , Humans , Brain
8.
Transl Stroke Res ; 2023 May 04.
Article in English | MEDLINE | ID: mdl-37140808

ABSTRACT

Ischemic stroke, a disease with high mortality and disability rate worldwide, currently has no effective treatment. The systemic inflammation response to the ischemic stroke, followed by immunosuppression in focal neurologic deficits and other inflammatory damage, reduces the circulating immune cell counts and multiorgan infectious complications such as intestinal and gut dysfunction dysbiosis. Evidence showed that microbiota dysbiosis plays a role in neuroinflammation and peripheral immune response after stroke, changing the lymphocyte populations. Multiple immune cells, including lymphocytes, engage in complex and dynamic immune responses in all stages of stroke and may be a pivotal moderator in the bidirectional immunomodulation between ischemic stroke and gut microbiota. This review discusses the role of lymphocytes and other immune cells, the immunological processes in the bidirectional immunomodulation between gut microbiota and ischemic stroke, and its potential as a therapeutic strategy for ischemic stroke.

9.
Sci Total Environ ; 881: 163481, 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37068676

ABSTRACT

The present study investigated the toxic effects of IMI on brain and gut of zebrafish (Danio rerio) by a combination of transcriptome and microbiome analysis. In addition, the involvement of light/dark period was also evaluated. An acute toxic test was conducted on adult zebrafish weighing 0.45 ± 0.02 g with 4 experimental groups (n = 15): 1) IMI group (Light: Dark = 12: 12 h), 2) prolonged light group (Light: Dark = 20: 4 h), 3) prolonged darkness group (Light: Dark = 4: 20 h) which received 20 mg/L of IMI, and 4) control group, which was not treated with IMI (Light: Dark = 12: 12 h). The results showed that prolonged darkness improved the survival rate of zebrafish upon IMI exposure for 96 h. In the sub-chronic test, zebrafish were divided into the same 4 groups and exposed to IMI at 1 mg/L for 14 d (n = 30). The results showed that IMI induced oxidative stress in both IMI and prolonged light groups by inhibition of antioxidant activities and accumulation of oxidative products. Transcriptome analysis revealed a compromise of antioxidation and tryptophan metabolism pathways under IMI exposure. Several genes encoding rate-limiting enzymes in serotonin and melatonin synthesis were all inhibited in both IMI and LL groups. Meanwhile, significant decrease (P < 0.5) of serotonin and melatonin levels was observed. However, there's remarkable improvement of biochemical and transcriptional status in prolonged darkness group. In addition, microbiome analysis showed great alteration of gut bacterial community structure and inhibition of tryptophan metabolism pathway. Similarly, the gut microbiota dysbiosis induced by IMI was alleviated in prolonged darkness. In summary, sub-chronic IMI exposure induced neurotoxicity and gut toxicity in zebrafish by oxidative stress and impaired the brain-gut-axis through tryptophan metabolism perturbation. Prolonged darkness could effectively attenuate the IMI toxicity probably through maintaining a normal tryptophan metabolism.


Subject(s)
Brain-Gut Axis , Melatonin , Animals , Zebrafish/physiology , Serotonin/metabolism , Darkness , Melatonin/metabolism , Tryptophan
10.
Eur J Neurol ; 30(11): 3529-3539, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36905309

ABSTRACT

BACKGROUND AND PURPOSE: Growing evidence suggests that abnormalities in brain-gut-microbiome (BGM) interactions are involved in the pathogenesis of irritable bowel syndrome (IBS). Our study aimed to explore alterations in dynamic functional connectivity (DFC), the gut microbiome and the bidirectional interaction in the BGM. METHODS: Resting-state functional magnetic resonance imaging (rs-fMRI), fecal samples and clinical chacteristics were collected from 33 IBS patients and 32 healthy controls. We performed a systematic DFC analysis on rs-fMRI. The gut microbiome was analyzed by 16S rRNA gene sequencing. Associations between DFC characteristics and microbial alterations were explored. RESULTS: In the DFC analysis, four dynamic functional states were identified. IBS patients exhibited increased mean dwell and fraction time in State 4, and reduced transitions from State 3 to State 1. Aberrant temporal properties in State 4 were only evident when choosing a short window (36 s or 44 s). Decreased functional connectivity (FC) variability was found in State 1 and State 3 in IBS patients, two of which (independent component [IC]51-IC91, IC46-IC11) showed significant correlations with clinical characteristics. Additionally, we identified nine significantly differential abundances in microbial composition. We also found that IBS-related microbiota were associated with aberrant FC variability, although these exploratory results were obtained at an uncorrected threshold of significance. CONCLUSIONS: Although future studies are needed to confirm our results, the findings not only provide a new insight into the dysconnectivity hypothesis in IBS from a dynamic perspective, but also establish a possible link between DFC and the gut microbiome, which lays the foundation for future research on disrupted BGM interactions.

11.
Chinese Journal of Geriatrics ; (12): 484-488, 2023.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-993840

ABSTRACT

Recent studies have found that the brain-gut-microbiome axis(BGMA)is closely related to the occurrence and development of Alzheimer's disease(AD). BGMA can affect AD in various aspects such as neuro-immune regulation and intestinal microflora, and is a potential new target for the treatment of AD.The "Sanjiao" acupuncture method is proposed by professor Han Jingxian, a famous Chinese medicine practitioner, based on his theory of "dysfunction of Qi activity of Sanjiao leads to aging" , and has been widely used in the treatment of AD and other age-related diseases in clinical practice.This article reviews the theory of "dysfunction of Qi activity of Sanjiao leads to aging" and the relationship between the "Sanjiao" acupuncture method and BGMA, with the hope that the "San Jiao" acupuncture method can become a new target for treatment of AD in the future.

12.
Front Cell Infect Microbiol ; 13: 1269548, 2023.
Article in English | MEDLINE | ID: mdl-38173792

ABSTRACT

Objective: Intermittent energy restriction (IER) is an effective weight loss strategy. However, little is known about the dynamic effects of IER on the brain-gut-microbiome axis. Methods: In this study, a total of 25 obese individuals successfully lost weight after a 2-month IER intervention. FMRI was used to determine the activity of brain regions. Metagenomic sequencing was performed to identify differentially abundant gut microbes and pathways in from fecal samples. Results: Our results showed that IER longitudinally reduced the activity of obese-related brain regions at different timepoints, including the inferior frontal orbital gyrus in the cognitive control circuit, the putamen in the emotion and learning circuit, and the anterior cingulate cortex in the sensory circuit. IER longitudinally reduced E. coli abundance across multiple timepoints while elevating the abundance of obesity-related Faecalibacterium prausnitzii, Parabacteroides distasonis, and Bacterokles uniformis. Correlation analysis revealed longitudinally correlations between gut bacteria abundance alterations and brain activity changes. Conclusions: There was dynamical alteration of BGM axis (the communication of E. coli with specific brain regions) during the weight loss under the IER.


Subject(s)
Gastrointestinal Microbiome , Humans , Caloric Restriction/methods , Escherichia coli , Obesity , Weight Loss
13.
Pharmacol Rep ; 74(5): 871-889, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35945482

ABSTRACT

BACKGROUND: Complex interactions between the brain, gut and adipose tissue allow to recognize obesity as a neurometabolic disorder. The recent data have shown that gut microbiota can play a potential role in obesity development. Transcranial direct current stimulation (tDCS) is a safe and non-invasive technique to modulate the activity of cerebral cortex and other connected brain areas also in context of appetite control. The objective of this study was to evaluate the effects of repetitive anodal tDCS (AtDCS) of prefrontal cortex on feeding behavior, metabolic status and selected phyla of gut microbiota in rats with obesity induced by high-calorie diet (HCD). METHODS: 32 female Wistar rats were equally divided into 4 subgroups depending on diet effect (lean versus obese) and type of stimulation (active versus sham tDCS versus no stimulation). Feed intake, body weight, blood lipoproteins and leptin levels as well as Firmicutes and Bacteroidetes in intestines and stool were examined. RESULTS: HCD changed feeding behavior and metabolic parameters typically for obesity-related ranges and resulted in an abundance of Firmicutes at the expanse of Bacteroidetes in the large intestine and stool. AtDCS decreased appetite, body weight, and cholesterol levels. In addition, AtDCS reduced ratio of the average number of Firmicutes to average number of Bacteroidetes in all examined tissues. CONCLUSIONS: Repetitive AtDCS is not only effective for appetite restriction but can also modulate gut microbiome composition which demonstrates the existence of the brain-gut-microbiome axis and points at this technique as a promising complementary treatment for obesity. However, the effects should be further replicated in human studies.


Subject(s)
Transcranial Direct Current Stimulation , Humans , Animals , Female , Rats , Transcranial Direct Current Stimulation/methods , Leptin , Rodentia , Brain-Gut Axis , Rats, Wistar , Obesity/therapy , Obesity/metabolism , Body Weight , Cholesterol
14.
Int J Mol Sci ; 23(9)2022 May 05.
Article in English | MEDLINE | ID: mdl-35563564

ABSTRACT

The intestinal microbiome composition and dietary supplementation with psychobiotics can result in neurochemical alterations in the brain, which are possible due to the presence of the brain-gut-microbiome axis. In the present study, magnetic resonance spectroscopy (MRS) and behavioural testing were used to evaluate whether treatment with Lacticaseibacillus rhamnosus JB-1 (JB­1) bacteria alters brain metabolites' levels and behaviour during continuous exposure to chronic stress. Twenty Wistar rats were subjected to eight weeks of a chronic unpredictable mild stress protocol. Simultaneously, half of them were fed with JB-1 bacteria, and the second half was given a daily placebo. Animals were examined at three-time points: before starting the stress protocol and after five and eight weeks of stress onset. In the elevated plus maze behavioural test the placebo group displayed increased anxiety expressed by almost complete avoidance of exploration, while the JB-1 dietary supplementation mitigated anxiety which resulted in a longer exploration time. Hippocampal MRS measurements demonstrated a significant decrease in glutamine + glutathione concentration in the placebo group compared to the JB-1 bacteria-supplemented group after five weeks of stress. With the progression of stress the decrease of glutamate, glutathione, taurine, and macromolecular concentrations were observed in the placebo group as compared to baseline. The level of brain metabolites in the JB-1-supplemented rats were stable throughout the experiment, with only the taurine level decreasing between weeks five and eight of stress. These data indicated that the JB-1 bacteria diet might stabilize levels of stress-related neurometabolites in rat brain and could prevent the development of anxiety/depressive-like behaviour.


Subject(s)
Lacticaseibacillus rhamnosus , Animals , Behavior, Animal , Eating , Glutathione/metabolism , Lacticaseibacillus rhamnosus/metabolism , Rats , Rats, Wistar , Stress, Psychological , Taurine/metabolism
15.
Microbiome ; 9(1): 236, 2021 11 30.
Article in English | MEDLINE | ID: mdl-34847963

ABSTRACT

BACKGROUND: There is growing recognition that bidirectional signaling between the digestive tract and the brain contributes to irritable bowel syndrome (IBS). We recently showed in a large randomized controlled trial that cognitive behavioral therapy (CBT) reduces IBS symptom severity. This study investigated whether baseline brain and gut microbiome parameters predict CBT response and whether response is associated with changes in the brain-gut-microbiome (BGM) axis. METHODS: Eighty-four Rome III-diagnosed IBS patients receiving CBT were drawn from the Irritable Bowel Syndrome Outcome Study (IBSOS; ClinicalTrials.gov NCT00738920) for multimodal brain imaging and psychological assessments at baseline and after study completion. Fecal samples were collected at baseline and post-treatment from 34 CBT recipients for 16S rRNA gene sequencing, untargeted metabolomics, and measurement of short-chain fatty acids. Clinical measures, brain functional connectivity and microstructure, and microbiome features associated with CBT response were identified by multivariate linear and negative binomial models. RESULTS: At baseline, CBT responders had increased fecal serotonin levels, and increased Clostridiales and decreased Bacteroides compared to non-responders. A random forests classifier containing 11 microbial genera predicted CBT response with high accuracy (AUROC 0.96). Following treatment, CBT responders demonstrated reduced functional connectivity in regions of the sensorimotor, brainstem, salience, and default mode networks and changes in white matter in the basal ganglia and other structures. Brain changes correlated with microbiome shifts including Bacteroides expansion in responders. CONCLUSIONS: Pre-treatment intestinal microbiota and serotonin levels were associated with CBT response, suggesting that peripheral signals from the microbiota can modulate central processes affected by CBT that generate abdominal symptoms in IBS. CBT response is characterized by co-correlated shifts in brain networks and gut microbiome that may reflect top-down effects of the brain on the microbiome during CBT. Video abstract.


Subject(s)
Cognitive Behavioral Therapy , Gastrointestinal Microbiome , Irritable Bowel Syndrome , Brain-Gut Axis , Humans , Irritable Bowel Syndrome/therapy , RNA, Ribosomal, 16S/genetics
16.
Cell ; 184(24): 5916-5931.e17, 2021 11 24.
Article in English | MEDLINE | ID: mdl-34767757

ABSTRACT

There is increasing interest in the potential contribution of the gut microbiome to autism spectrum disorder (ASD). However, previous studies have been underpowered and have not been designed to address potential confounding factors in a comprehensive way. We performed a large autism stool metagenomics study (n = 247) based on participants from the Australian Autism Biobank and the Queensland Twin Adolescent Brain project. We found negligible direct associations between ASD diagnosis and the gut microbiome. Instead, our data support a model whereby ASD-related restricted interests are associated with less-diverse diet, and in turn reduced microbial taxonomic diversity and looser stool consistency. In contrast to ASD diagnosis, our dataset was well powered to detect microbiome associations with traits such as age, dietary intake, and stool consistency. Overall, microbiome differences in ASD may reflect dietary preferences that relate to diagnostic features, and we caution against claims that the microbiome has a driving role in ASD.


Subject(s)
Autistic Disorder/microbiology , Feeding Behavior , Gastrointestinal Microbiome , Adolescent , Age Factors , Autistic Disorder/diagnosis , Behavior , Child , Child, Preschool , Feces/microbiology , Female , Humans , Male , Phenotype , Phylogeny , Species Specificity
17.
Biomolecules ; 11(7)2021 07 07.
Article in English | MEDLINE | ID: mdl-34356624

ABSTRACT

The brain-gut-microbiome axis is a bidirectional communication pathway between the gut microbiota and the central nervous system. The growing interest in the gut microbiota and mechanisms of its interaction with the brain has contributed to the considerable attention given to the potential use of probiotics, prebiotics and postbiotics in the prevention and treatment of depressive disorders. This review discusses the up-to-date findings in preclinical and clinical trials regarding the use of pro-, pre- and postbiotics in depressive disorders. Studies in rodent models of depression show that some of them inhibit inflammation, decrease corticosterone level and change the level of neurometabolites, which consequently lead to mitigation of the symptoms of depression. Moreover, certain clinical studies have indicated improvement in mood as well as changes in biochemical parameters in patients suffering from depressive disorders.


Subject(s)
Brain/metabolism , Depressive Disorder , Gastrointestinal Microbiome , Prebiotics , Probiotics/therapeutic use , Brain/microbiology , Depressive Disorder/metabolism , Depressive Disorder/microbiology , Depressive Disorder/therapy , Humans , Inflammation/metabolism , Inflammation/microbiology
18.
Curr Med Res Opin ; 37(10): 1739-1744, 2021 10.
Article in English | MEDLINE | ID: mdl-34289768

ABSTRACT

After fecal microbiota transplantation (FMT) to treat Clostridioides difficile infection (CDI), cognitive improvement is noticeable, suggesting an essential association between the gut microbiome and neural function. Although it is known that the gut microbiome is linked with cognitive function, whether FMT may lead to cognitive improvement in patients with neurodegenerative disorders remains to be elucidated. We present the case of a 90-year-old woman with Alzheimer's dementia and severe CDI who underwent FMT. Cognitive function testing (Mini-Mental State Examination, Montreal Cognitive Assessment, and Clinical Dementia Rating assessment) was performed one month before FMT and one week and one month after FMT. We collected the patients' fecal samples before FMT and 3 weeks after FMT to compare the microbiota composition. The 16S rRNA gene amplicons were analyzed using the QIIME2 platform (version 2020.2) and the Phyloseq R package. The linear discriminant analysis effect size was performed to determine the taxonomic difference between pre- and post-FMT. Functional biomarker analysis using the Kruskal-Wallis H test was performed between the pre- and post-FMT. The cognitive function tests after FMT showed an improvement compared to the tests before the procedure. FMT changed the microbiota composition in recipient feces. We found that the genera were reported to be associated with cognitive function. In addition, short-chain fatty acids were found to be significantly different between before and after FMT. This finding suggests the presence of an association between the gut microbiome and cognitive function. Further, it emphasizes the need for clinical awareness regarding the effect of FMT on the brain-gut-microbiome axis and its potential as a therapy for patients with dementia.


Subject(s)
Alzheimer Disease , Clostridioides difficile , Clostridium Infections , Aged, 80 and over , Alzheimer Disease/therapy , Clostridium Infections/therapy , Cognition , Fecal Microbiota Transplantation , Feces , Female , Humans , RNA, Ribosomal, 16S/genetics , Treatment Outcome
19.
Nutrients ; 13(2)2021 Feb 13.
Article in English | MEDLINE | ID: mdl-33668627

ABSTRACT

In the past few decades, obesity has reached pandemic proportions. Obesity is among the main risk factors for cardiovascular diseases, since chronic fat accumulation leads to dysfunction in vascular endothelium and to a precocious arterial stiffness. So far, not all the mechanisms linking adipose tissue and vascular reactivity have been explained. Recently, novel findings reported interesting pathological link between endothelial dysfunction with gut hormones and gut microbiota and energy homeostasis. These findings suggest an active role of gut secretome in regulating the mediators of vascular function, such as nitric oxide (NO) and endothelin-1 (ET-1) that need to be further investigated. Moreover, a central role of brain has been suggested as a main player in the regulation of the different factors and hormones beyond these complex mechanisms. The aim of the present review is to discuss the state of the art in this field, by focusing on the processes leading to endothelial dysfunction mediated by obesity and metabolic diseases, such as insulin resistance. The role of perivascular adipose tissue (PVAT), gut hormones, gut microbiota dysbiosis, and the CNS function in controlling satiety have been considered. Further understanding the crosstalk between these complex mechanisms will allow us to better design novel strategies for the prevention of obesity and its complications.


Subject(s)
Dysbiosis/physiopathology , Endothelium, Vascular/physiopathology , Gastrointestinal Hormones/metabolism , Gastrointestinal Microbiome/physiology , Obesity/physiopathology , Adipose Tissue/physiopathology , Animals , Brain/physiopathology , Cardiovascular Diseases/etiology , Cardiovascular Diseases/microbiology , Cardiovascular Diseases/physiopathology , Dysbiosis/complications , Energy Metabolism/physiology , Humans , Obesity/complications , Obesity/microbiology , Satiation/physiology , Vascular Stiffness/physiology
20.
Article in English | MEDLINE | ID: mdl-33326819

ABSTRACT

The high comorbidity of psychological disorders in both functional and organic gastrointestinal diseases suggests the intimate and complex link between the brain and the gut. Termed the brain-gut axis, this bidirectional communication between the central nervous system and enteric nervous system relies on immune, endocrine, neural, and metabolic pathways. There is increasing evidence that the gut microbiome is a key part of this system, and dysregulation of the brain-gut-microbiome axis (BGMA) has been implicated in disorders of brain-gut interaction, including irritable bowel syndrome, and in neuropsychiatric disorders, including depression, Alzheimer's disease, and autism spectrum disorder. Further, alterations in the gut microbiome have been implicated in the pathogenesis of organic gastrointestinal diseases, including inflammatory bowel disease. The BGMA is an attractive therapeutic target, as using prebiotics, probiotics, or postbiotics to modify the gut microbiome or mimic gut microbial signals could provide novel treatment options to address these debilitating diseases. However, despite significant advancements in our understanding of the BGMA, clinical data is lacking. In this article, we will review current understanding of the comorbidity of gastrointestinal diseases and psychological disorders. We will also review the current evidence supporting the key role of the BGMA in this pathology. Finally, we will discuss the clinical implications of the BGMA in the evaluation and management of psychological and gastrointestinal disorders.


Subject(s)
Brain-Gut Axis/physiology , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/psychology , Gastrointestinal Microbiome/physiology , Mental Disorders/epidemiology , Mental Disorders/psychology , Brain/physiology , Comorbidity , Enteric Nervous System/physiology , Gastrointestinal Diseases/diet therapy , Humans , Irritable Bowel Syndrome/diet therapy , Irritable Bowel Syndrome/epidemiology , Irritable Bowel Syndrome/psychology , Mental Disorders/diet therapy , Prebiotics/administration & dosage , Probiotics/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...