Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 7.498
Filtrer
1.
PLoS Comput Biol ; 20(9): e1012359, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39288161

RÉSUMÉ

Considering biological systems as information processing entities and analyzing their organizational structure via information-theoretic measures has become an established approach in life sciences. We transfer this framework to a field of broad general interest, the human gut microbiome. We use BacArena, a software combining agent-based modelling and flux-balance analysis, to simulate a simplified human intestinal microbiome (SIHUMI). In a first step, we derive information theoretic measures from the simulated abundance data, and, in a second step, relate them to the metabolic processes underlying the abundance data. Our study provides further evidence on the role of active information storage as an indicator of unexpected structural change in the observed system. Besides, we show that information transfer reflects coherent behavior in the microbial community, both as a reaction to environmental changes and as a result of direct effective interaction. In this sense, purely abundance-based information theoretic measures can provide meaningful insight on metabolic interactions within bacterial communities. Furthermore, we shed light on the important however little noticed technical aspect of distinguishing immediate and delayed effects in the interpretation of local information theoretical measures.


Sujet(s)
Simulation numérique , Microbiome gastro-intestinal , Humains , Microbiome gastro-intestinal/physiologie , Biologie informatique , Modèles biologiques , Logiciel , Mémorisation et recherche des informations/méthodes
3.
Gut Microbes ; 16(1): 2401939, 2024.
Article de Anglais | MEDLINE | ID: mdl-39259834

RÉSUMÉ

Early life stress alters gut microbiota and increases the risk of neuropsychiatric disorders, including social deficits and anxiety, in the host. However, the role of gut commensal bacteria in early life stress-induced neurobehavioral abnormalities remains unclear. Using the maternally separated (MS) mice, our research has unveiled a novel aspect of this complex relationship. We discovered that the reduced levels of amino acid transporters in the intestine of MS mice led to low glutamine (Gln) levels in the blood and synaptic dysfunction in the medial prefrontal cortex (mPFC). Abnormally low blood Gln levels limit the brain's availability of Gln, which is required for presynaptic glutamate (Glu) and γ-aminobutyric acid (GABA) replenishment. Furthermore, MS resulted in gut microbiota dysbiosis characterized by a reduction in the relative abundance of Lactobacillus reuteri (L. reuteri). Notably, supplementation with L. reuteri ameliorates neurobehavioral abnormalities in MS mice by increasing intestinal amino acid transport and restoring synaptic transmission in the mPFC. In conclusion, our findings on the role of L. reuteri in regulating intestinal amino acid transport and buffering early life stress-induced behavioral abnormalities provide a novel insight into the microbiota-gut-brain signaling basis for emotional behaviors.


Sujet(s)
Anxiété , Microbiome gastro-intestinal , Stress psychologique , Animaux , Microbiome gastro-intestinal/physiologie , Souris , Anxiété/microbiologie , Anxiété/métabolisme , Stress psychologique/microbiologie , Stress psychologique/métabolisme , Acides aminés/métabolisme , Mâle , Souris de lignée C57BL , Systèmes de transport d'acides aminés/métabolisme , Cortex préfrontal/métabolisme , Comportement animal , Dysbiose/microbiologie , Séparation d'avec la mère , Glutamine/métabolisme , Axe cerveau-intestin/physiologie , Transmission synaptique , Femelle , Acide glutamique/métabolisme
4.
Nutrients ; 16(17)2024 Aug 29.
Article de Anglais | MEDLINE | ID: mdl-39275207

RÉSUMÉ

The regulation of neurogenesis, the complex process of producing and differentiating new brain tissue cells, is influenced by a complex interaction of internal and external factors. Over the past decade, extensive research has been conducted on neurotrophins and their key role in adult neurogenesis, as well as their impact on diseases such as depression. Among neurotrophins, the brain-derived neurotrophic factor (BDNF) has been the subject of comprehensive studies on adult neurogenesis, and scientific evidence supports its necessity for neurogenesis in the subventricular zone of the hippocampus. A novel area of research is the emerging role of gut microbiota as a significant contributor to neurogenesis and neurotrophin production. Studies have shown that reduced BDNF levels can lead to mood disorders, which are observed in intestinal dysbiosis, characterized by an imbalance in the composition and quantity of the intestinal microbiota. There is evidence in the literature that there is a link between brain function and gut microbiota. Physical activity, and especially the regularity and intensity of exercise, is important in relation to the level of BDNF and the intestinal microbiota. Probiotics, prebiotics and physical activity may have a positive effect on the intestinal microbiota, and therefore also on the level of the brain-derived neurotrophic factor.


Sujet(s)
Facteur neurotrophique dérivé du cerveau , Exercice physique , Microbiome gastro-intestinal , Probiotiques , Facteur neurotrophique dérivé du cerveau/métabolisme , Microbiome gastro-intestinal/physiologie , Humains , Exercice physique/physiologie , Neurogenèse , Animaux , Dysbiose , Prébiotiques , Encéphale/métabolisme , Axe cerveau-intestin/physiologie
5.
Cells ; 13(17)2024 Aug 27.
Article de Anglais | MEDLINE | ID: mdl-39273008

RÉSUMÉ

The influence of gut microbiome, metabolites, omics, hormones, and stress on general and mental health is increasingly being recognized. Ancient cultures recognized the importance of diet and gut health on the overall health of an individual. Western science and modern scientific methods are beginning to unravel the foundations and mechanisms behind some of the ancient beliefs and customs. The gut microbiome, an organ itself, is now thought to influence almost all other organs, ranging from the brain to the reproductive systems. Gut microbiome, metabolites, hormones, and biological sex also influence a myriad of health conditions that range from mental health disorders, obesity, gastrointestinal disorders, and cardiovascular diseases to reproductive health. Here, we review the history and current understanding of the gut-brain axis bidirectional talk in various mental health disorders with special emphasis on anxiety and depressive disorders, whose prevalence has increased by over 50% in the past three decades with COVID-19 pandemic being the biggest risk factor in the last few years. The vagal nerve is an important contributor to this bidirectional talk, but other pathways also contribute, and most remain understudied. Probiotics containing Lactobacillus and Bifidobacterium species seem to have the most impact on improvement in mental health symptoms, but the challenge appears to be maintaining sustained levels, especially since neither Lactobacillus nor Bifidobacterium can permanently colonize the gut. Ancient endogenous retroviral DNA in the human genome is also linked to several psychiatric disorders, including depression. These discoveries reveal the complex and intricately intertwined nature of gut health with mental health disorders.


Sujet(s)
Axe cerveau-intestin , Microbiome gastro-intestinal , Troubles mentaux , Humains , Microbiome gastro-intestinal/physiologie , Axe cerveau-intestin/physiologie , Troubles mentaux/métabolisme , Troubles mentaux/microbiologie , Métabolomique , Hormones/métabolisme , Stress psychologique/métabolisme , Stress psychologique/microbiologie , Santé mentale , COVID-19/métabolisme , COVID-19/virologie , Encéphale/métabolisme , Animaux
6.
Nutrients ; 16(17)2024 Aug 24.
Article de Anglais | MEDLINE | ID: mdl-39275156

RÉSUMÉ

The gut microbiota constitutes a complex ecosystem, comprising trillions of microbes that have co-evolved with their host over hundreds of millions of years. Over the past decade, a growing body of knowledge has underscored the intricate connections among diet, gut microbiota, and human health. Bioactive polysaccharides (BPs) from natural sources like medicinal plants, seaweeds, and fungi have diverse biological functions including antioxidant, immunoregulatory, and metabolic activities. Their effects are closely tied to the gut microbiota, which metabolizes BPs into health-influencing compounds. Understanding how BPs and gut microbiota interact is critical for harnessing their potential health benefits. This review provides an overview of the human gut microbiota, focusing on its role in metabolic diseases like obesity, type II diabetes mellitus, non-alcoholic fatty liver disease, and cardiovascular diseases. It explores the basic characteristics of several BPs and their impact on gut microbiota. Given their significance for human health, we summarize the biological functions of these BPs, particularly in terms of immunoregulatory activities, blood sugar, and hypolipidemic effect, thus providing a valuable reference for understanding the potential benefits of natural BPs in treating metabolic diseases. These properties make BPs promising agents for preventing and treating metabolic diseases. The comprehensive understanding of the mechanisms by which BPs exert their effects through gut microbiota opens new avenues for developing targeted therapies to improve metabolic health.


Sujet(s)
Microbiome gastro-intestinal , Maladies métaboliques , Polyosides , Humains , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Polyosides/pharmacologie , Maladies métaboliques/microbiologie , Maladies métaboliques/traitement médicamenteux , Stéatose hépatique non alcoolique/microbiologie , Stéatose hépatique non alcoolique/traitement médicamenteux , Stéatose hépatique non alcoolique/métabolisme , Diabète de type 2/microbiologie , Diabète de type 2/traitement médicamenteux , Animaux , Obésité/microbiologie , Obésité/traitement médicamenteux , Obésité/métabolisme
7.
Nutrients ; 16(17)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39275251

RÉSUMÉ

Inulin is a plant polysaccharide which, due to its chemical structure, is not digestible by human gut enzymes but by some bacteria of the human microbiota, acting as a prebiotic. Consequently, inulin consumption has been associated with changes in the composition of the intestinal microbiota related to an improvement of the metabolic state, counteracting different obesity-related disturbances. However, the specific mechanisms of action, including bacterial changes, are not exactly known. Here, a bibliographic review was carried out to study the main effects of inulin on human metabolic health, with a special focus on the mechanisms of action of this prebiotic. Inulin supplementation contributes to body weight and BMI control, reduces blood glucose levels, improves insulin sensitivity, and reduces inflammation markers, mainly through the selective favoring of short-chain fatty acid (SCFA)-producer species from the genera Bifidobacterium and Anaerostipes. These SCFAs have been shown to ameliorate glucose metabolism and decrease hepatic lipogenesis, reduce inflammation, modulate immune activity, and improve anthropometric parameters such as body weight or BMI. In conclusion, the studies collected suggest that inulin intake produces positive metabolic effects through the improvement of the intestinal microbiota and through the metabolites produced by its fermentation.


Sujet(s)
Microbiome gastro-intestinal , Inuline , Prébiotiques , Humains , Inuline/pharmacologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Acides gras volatils/métabolisme , Obésité/métabolisme , Obésité/microbiologie , Indice de masse corporelle , Glycémie/métabolisme , Poids/effets des médicaments et des substances chimiques , Insulinorésistance
8.
Nutrients ; 16(17)2024 Sep 04.
Article de Anglais | MEDLINE | ID: mdl-39275296

RÉSUMÉ

BACKGROUND: The causes of functional constipation (FC) in adults are unclear, but changes in the gut microbiome may play an important role. The present study aimed to assess the relationship between urinary metabolites of dopamine and serotonin and some dysbiosis indicators in patients with FC. The study included 40 healthy women and 40 women with FC aged 21-46 years. METHODS: Urinary levels of homovanillic acid (HVA), 5-hydoxyindoleacetic acid (5-HIAA), p-hydroxyphenylacetic acid (PhAc), and 3-indoxyl sulfate, as final metabolites of dopamine, serotonin, and indole pathway, respectively, were determined using the LC-Ms/Ms method. However, hydrogen-methane and ammonia breath tests were performed. The GA-map Dysbiosis Test was used to identify and characterize the dysbiosis index (DI). RESULTS: In patients with FC, the DI was significantly higher than in the control group: 4.05 ± 0.53 vs. 1.52 ± 0.81 points (p < 0.001), but the number of many types of bacteria varied among individuals. The levels of HVA were higher, while 5-HIAA levels were lower in patients. Moreover, the HVA/5-HIAA ratio had a positive correlation with DI as well as with the severity of symptoms. CONCLUSIONS: In patients with functional constipation, the balance in dopamine and serotonin secretion is disturbed, which is associated with changes in the gut microbiome.


Sujet(s)
Constipation , Dopamine , Dysbiose , Microbiome gastro-intestinal , Sérotonine , Humains , Constipation/urine , Constipation/microbiologie , Femelle , Adulte , Dysbiose/urine , Sérotonine/urine , Adulte d'âge moyen , Dopamine/urine , Dopamine/métabolisme , Microbiome gastro-intestinal/physiologie , Jeune adulte , Études cas-témoins , Acide homovanillique/urine
9.
Adv Exp Med Biol ; 1460: 357-372, 2024.
Article de Anglais | MEDLINE | ID: mdl-39287858

RÉSUMÉ

Gut microbiota is an indispensable commensal partner of human superorganism. The wealth of genetic repertoire provided by these microorganisms extends host's substrate processing capability. Energy and nutrient harvesting machinery primarily depends on the proper function of these organisms. However, the dynamic composition of microbiota changes with age, lifestyle, stress factors, infections, medications, and host pathophysiological conditions. Host immune system is primarily responsible for shaping up the microbial community and sustaining the symbiotic state. This involves controlling the delicate balance between agility toward pathobionts and tolerance toward symbionts. When things go wrong with this crosstalk, dysbiosis may arise.Metabolic syndrome is a multisystemic, low-grade chronic inflammatory disease that involves dyslipidemia, glucose intolerance, insulin resistance, and central obesity. Excess caloric intake with high-sugar and high-fat diet promote high energy harvesting and lipogenesis. The secretion of adipokines accompanies lipid spillover from fat cells, which contribute to insulin resistance and the expansion of adipose tissue in ectopic sites. Proinflammatory cytokines from adipose tissue macrophages increase the extent of adipose dysfunction.The inflammatory nature of obesity and metabolic syndrome recall the connection between dysbiosis and immune dysfunction. A remarkable association exits between obesity, inflammatory bowel disease, gluten-sensitive enteropathy, and dysbiosis. These conditions compromise the gut mucosa barrier and allow lipopolysaccharide to enter circulation. Unresolved chronic inflammation caused by one condition may overlap or trigger the other(s). Experimental studies and therapeutic trials of fecal microbiota transplantation promise limited improvement in some of these conditions.Typically, metabolic syndrome is considered as a consequence of overnutrition and the vicious cycle of lipogenesis, lipid accumulation, and chronic low-level inflammation. Because of the complex nature of this disorder, it remains inconclusive whether dysbiosis is a cause or consequence of obesity and metabolic syndrome.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Syndrome métabolique X , Obésité , Humains , Microbiome gastro-intestinal/physiologie , Syndrome métabolique X/microbiologie , Syndrome métabolique X/métabolisme , Obésité/microbiologie , Obésité/métabolisme , Animaux , Métabolisme lipidique , Inflammation/métabolisme , Inflammation/microbiologie , Tissu adipeux/métabolisme , Tissu adipeux/immunologie
11.
J Psychiatry Neurosci ; 49(5): E289-E300, 2024.
Article de Anglais | MEDLINE | ID: mdl-39299780

RÉSUMÉ

BACKGROUND: Increasing evidence suggests an important role of the gut microbiome in the pathogenesis of mental disorders, including depression, along the microbiota-gut-brain axis. We sought to explore the interactions between gut microbe composition and neural circuits in late-life depression (LLD). METHODS: We performed fecal 16S ribosomal RNA (rRNA) sequencing and resting-state functional magnetic resonance imaging in a case-control cohort of older adults with LLD and healthy controls to characterize the association between gut microbiota and brain functional connectivity (FC). We used the Hamilton Depression Rating Scale (HAMD) to assess depressive symptoms. RESULTS: We included 32 adults with LLD and 16 healthy controls. At the genus level, the relative abundance of Enterobacter, Akkermansiaceae, Hemophilus, Burkholderia, and Rothia was significantly higher among patients with LDD than controls. Reduced FC within mood regulation circuits was mainly found in the frontal cortex (e.g., the right superior and inferior frontal gyrus, right lateral occipital cortex, left middle frontal gyrus, and left caudate) among patients with MDD. Group-characterized gut microbes among controls and patients showed opposite correlations with seed-based FC, which may account for the aberrant emotion regulation among patients with LDD. The abundance of Enterobacter (dominant genus among patients with LLD) was positively correlated with both HAMD scores (r = 0.49, p = 0.0004) and group-characterized FC (r = -0.37, p < 0.05), while Odoribacter (dominant genus among controls) was negatively correlated with both HAMD scores (r = -0.30, p = 0.04) and group-characterized FC. LIMITATIONS: The study's cross-sectional design and small sample size limit causal inferences; larger longitudinal studies are required for detailed subgroup analyses. CONCLUSION: We identified significant correlations between LDD-characterized gut microbes and brain FC, as well as depression severity, which may contribute to the pathophysiology of depression development among patients with LLD. Specific microbes were linked to altered brain connectivity, suggesting potential targets for treating LLD.


Sujet(s)
Microbiome gastro-intestinal , Imagerie par résonance magnétique , Humains , Microbiome gastro-intestinal/physiologie , Mâle , Femelle , Sujet âgé , Études cas-témoins , Axe cerveau-intestin/physiologie , Encéphale/imagerie diagnostique , Encéphale/physiopathologie , Encéphale/microbiologie , Fèces/microbiologie , ARN ribosomique 16S/génétique , Trouble dépressif majeur/microbiologie , Trouble dépressif majeur/physiopathologie , Trouble dépressif majeur/imagerie diagnostique , Adulte d'âge moyen
12.
Integr Cancer Ther ; 23: 15347354241280390, 2024.
Article de Anglais | MEDLINE | ID: mdl-39313964

RÉSUMÉ

BACKGROUND: In patients with metastatic colorectal cancer (mCRC), Quxie Cap-sule (QX)-a combination of conventional therapy (including chemotherapy, targeted therapy or supportive care)-has shown a significant overall survival benefit compared with placebo and might have the property of dual effects of antitumor and immunity enhancement, both mediated by the microbiome. In preclinical models, QX has also shown activity against colorectal cancer. This study aimed to describe how the aforementioned effects of QX look after when focusing on the patients in third or above line setting. METHODS: A Simon's Minimax two-stage phase II design was used in this study, which enrolled mCRC patients who progressed after second-line treatment. Patients received conventional therapy plus QX until disease progression or unacceptable toxicity. Before and after 1-month intervention, we collected patients' stool samples for microbiome analysis by 16s rRNA sequencing approaches. And the microbiome analysis before and after 1-month intervention was done through bioinformation analysis platform. RESULTS: Fifteen patients were enrolled and gut microbiome were analyzed from 7 of 10 patients that with PFS over 3.7 months. Microbiome community analysis on genus level showed that the proportion of Lachnospiraceae_UCG-001 (0.04% vs 1.06%, P = .02249) significantly increased after conventional therapy plus QX while the proportion of Alistipes (2.96% vs 1.35%, P = .03461), Flavonifractor (0.04% vs 0.02%, P = .02249), Bifidobacterium (6.11% vs 1.14%, P = .02249) and Butyricimonas (0.24% vs 0.11%, P = .03603) significantly decreased after intervention . LEfSe analysis showed that after intervention, samples were highly related with unclassified-f-lachnospiraceae, Eubacterium and Lachnospiraceae_UCG-001. CONCLUSIONS: Decrease of gut bacteria with potential roles in carcinogenesis of colorectal cancer and increase in the abundance of gut anticancer bacteria such as Lachnospiraceae may partly explain how conventional therapy combined with QX can influence carcinogenesis and tumor progression in colon cancer. TRIAL REGISTRATION: Chinese Clinical Trial Registry (ChiCTR2100053874).


Sujet(s)
Tumeurs colorectales , Microbiome gastro-intestinal , Humains , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Mâle , Femelle , Adulte d'âge moyen , Sujet âgé , Adulte , Métastase tumorale , Capsules , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Médicaments issus de plantes chinoises/usage thérapeutique , Médicaments issus de plantes chinoises/pharmacologie , Fèces/microbiologie
13.
Microbiologyopen ; 13(5): e1434, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39311537

RÉSUMÉ

The role of gut microbiota in health and disease is being thoroughly examined in various contexts, with a specific focus on the bacterial fraction due to its significant abundance. However, despite their lower abundance, viruses within the gut microbiota are gaining recognition for their crucial role in shaping the structure and function of the intestinal microbiota, with significant effects on the host as a whole, particularly the immune system. Similarly, environmental factors such as stress are key in modulating the host immune system, which in turn influences the composition of the gut virome and neurological functions through the bidirectional communication of the gut-brain axis. In this context, alterations in the host immune system due to stress and/or dysbiosis of the gut virome are critical factors in the development of both infectious and noninfectious diseases. The molecular mechanisms and correlation patterns between microbial species are not yet fully understood. This literature review seeks to explore the interconnected relationship between stress and the gut virome, with a focus on how this interaction is influenced by the host's immune system. We also discuss how disturbances in this finely balanced system can lead to the onset and/or progression of diseases.


Sujet(s)
Dysbiose , Microbiome gastro-intestinal , Stress physiologique , Virome , Humains , Microbiome gastro-intestinal/physiologie , Maladies transmissibles/virologie , Maladies transmissibles/microbiologie , Animaux , Virus/classification
14.
Diabetes Metab J ; 48(5): 821-836, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39313228

RÉSUMÉ

The human gut microbiota is increasingly recognized as a pivotal factor in diabetes management, playing a significant role in the body's response to treatment. However, it is important to understand that long-term usage of medicines like metformin and other diabetic treatments can result in problems, gastrointestinal discomfort, and dysbiosis of the gut flora. Advanced sequencing technologies have improved our understanding of the gut microbiome's role in diabetes, uncovering complex interactions between microbial composition and metabolic health. We explore how the gut microbiota affects glucose metabolism and insulin sensitivity by examining a variety of -omics data, including genomics, transcriptomics, epigenomics, proteomics, metabolomics, and metagenomics. Machine learning algorithms and genome-scale modeling are now being applied to find microbiological biomarkers associated with diabetes risk, predicted disease progression, and guide customized therapy. This study holds promise for specialized diabetic therapy. Despite significant advances, some concerns remain unanswered, including understanding the complex relationship between diabetes etiology and gut microbiota, as well as developing user-friendly technological innovations. This mini-review explores the relationship between multiomics, precision medicine, and machine learning to improve our understanding of the gut microbiome's function in diabetes. In the era of precision medicine, the ultimate goal is to improve patient outcomes through personalized treatments.


Sujet(s)
Microbiome gastro-intestinal , Médecine de précision , Biologie des systèmes , Humains , Microbiome gastro-intestinal/physiologie , Médecine de précision/méthodes , Biologie des systèmes/méthodes , Apprentissage machine , Dysbiose , Glycémie/analyse , Diabète/microbiologie , Diabète de type 2/microbiologie , Hypoglycémiants/usage thérapeutique
16.
Brain Behav ; 14(9): e70014, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39295072

RÉSUMÉ

OBJECTIVE: Anorexia nervosa has the highest mortality rate among psychiatric illnesses. Current treatments remain ineffective for a large fraction of patients. This may be due to unclear mechanisms behind its development and maintenance. Studies exploring the role of the gut microbiome have revealed inconsistent evidence of dysbiosis. This article aims to investigate changes in the gut microbiome, particularly, mean differences in the Firmicutes to Bacteroidetes ratio, in adolescent and adult individuals with anorexia nervosa following inpatient treatment. METHODS: Longitudinal studies investigating gut microbiome composition in inpatient populations of anorexia nervosa before and after treatment were systematically reviewed. Additionally, gut microbiome compositions were characterized in three acute anorexia nervosa inpatients early after admission and after 4-12 weeks of treatment. RESULTS: Review results indicated an increase in the Firmicutes to Bacteroidetes ratio in individuals with anorexia nervosa after treatment. These however did not match values of their healthy counterparts. In the case-series samples, the reverse occurred with samples taken 4 weeks after treatment. In the patient who provided an extra sample 12 weeks after treatment, similar results to the studies included in the review were observed. Furthermore, Firmicutes to Bacteroidetes ratio values in the case-series samples were notably higher in the two patients who had chronic anorexia nervosa. DISCUSSION: Differences in methodologies, small sample sizes, and insufficient data limited the generalizability of the outcomes of the reviewed studies. Results suggest a potentially unique microbiome signature in individuals with chronic anorexia nervosa, which may explain different outcomes in this group of patients.


Sujet(s)
Anorexie mentale , Bacteroidetes , Firmicutes , Microbiome gastro-intestinal , Patients hospitalisés , Anorexie mentale/microbiologie , Anorexie mentale/thérapie , Humains , Microbiome gastro-intestinal/physiologie , Bacteroidetes/isolement et purification , Firmicutes/isolement et purification , Femelle , Adulte , Adolescent , Jeune adulte , Dysbiose/microbiologie
17.
J Cell Mol Med ; 28(18): e70099, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39300699

RÉSUMÉ

Along with mounting evidence that gut microbiota and their metabolites migrate endogenously to distal organs, the 'gut-lung axis,' 'gut-brain axis,' 'gut-liver axis' and 'gut-renal axis' have been established. Multiple animal recent studies have demonstrated gut microbiota may also be a key susceptibility factor for neurological disorders such as Alzheimer's disease, Parkinson's disease and autism. The gastrointestinal tract is innervated by the extrinsic sympathetic and vagal nerves and the intrinsic enteric nervous system, and the gut microbiota interacts with the nervous system to maintain homeostatic balance in the host gut. A total of 1507 publications on the interactions between the gut microbiota, the gut-brain axis and neurological disorders are retrieved from the Web of Science to investigate the interactions between the gut microbiota and the nervous system and the underlying mechanisms involved in normal and disease states. We provide a comprehensive overview of the effects of the gut microbiota and its metabolites on nervous system function and neurotransmitter secretion, as well as alterations in the gut microbiota in neurological disorders, to provide a basis for the possibility of targeting the gut microbiota as a therapeutic agent for neurological disorders.


Sujet(s)
Axe cerveau-intestin , Microbiome gastro-intestinal , Maladies du système nerveux , Humains , Microbiome gastro-intestinal/physiologie , Animaux , Maladies du système nerveux/microbiologie , Maladies du système nerveux/métabolisme , Axe cerveau-intestin/physiologie , Système nerveux entérique/métabolisme , Encéphale/métabolisme , Système nerveux/métabolisme , Système nerveux/microbiologie , Tube digestif/microbiologie , Tube digestif/métabolisme
18.
Med Sci (Paris) ; 40(8-9): 654-660, 2024.
Article de Français | MEDLINE | ID: mdl-39303118

RÉSUMÉ

For 350 years, we have known that the human body hosts microbes, then called "animalcules". For over a century, following the demonstration of the role of some of these microbes in diseases, questions have arisen about the role of the largely predominant ones colonizing human skin and mucous surfaces, particularly the rich microbial ecosystem of the intestine, the gut microbiota. From the invention of germ-free life - axenism - which experimentally validated the human-microbe symbiosis, resulting from a long coevolution, to the development of anaerobic culture methods, then to the invention of molecular diagnosis, deep sequencing opening up metagenomic and omics approaches in general, a remarkable race has taken place between technological innovations and conceptual advances. This race, beyond the exhaustive description of the microbiota in its intra- and inter-human diversity, and the essential symbiotic functions of the microbiome, has paved the way for a new field of medicine: microbial medicine.


Title: Le microbiome humain : 340 ans d'histoire, 140 ans d'interrogations, d'innovations technologiques et émergence de la « médecine microbienne ¼. Abstract: On sait depuis 340ans que le corps humain héberge des microbes, alors appelés « animalcules ¼. Depuis plus d'un siècle, après la démonstration de la responsabilité de certains de ces microbes dans les maladies, on s'interroge sur le rôle de ceux ­ largement majoritaires ­ qui colonisent les surfaces cutanées et muqueuses humaines, particulièrement le riche écosystème microbien de l'intestin, le microbiote intestinal. De l'invention de la vie sans germe (axénie), qui a permis de valider expérimentalement la symbiose entre êtres humains et microbes, fruit d'une longue coévolution, à la mise au point des méthodes de culture anaérobies, puis à l'invention du diagnostic moléculaire, du séquençage profond ouvrant les approches métagénomiques et omiques en général, une formidable course s'est déroulée entre innovations technologiques et avancées conceptuelles. Cette course, au-delà de la description exhaustive du microbiote dans sa diversité intraet interhumaine, des fonctions symbiotiques essentielles du microbiome, a ouvert la voie d'un nouveau domaine de la médecine : la médecine microbienne.


Sujet(s)
Microbiote , Humains , Microbiote/physiologie , Histoire du 20ème siècle , Histoire du 21ème siècle , Histoire du 19ème siècle , Symbiose , Inventions/histoire , Inventions/tendances , Histoire du 18ème siècle , Microbiome gastro-intestinal/physiologie
19.
J Infect Dis ; 230(Supplement_2): S141-S149, 2024 Sep 10.
Article de Anglais | MEDLINE | ID: mdl-39255394

RÉSUMÉ

Alzheimer's disease (AD) is a neurodegenerative disorder with limited therapeutic options. Accordingly, new approaches for prevention and treatment are needed. One focus is the human microbiome, the consortium of microorganisms that live in and on us, which contributes to human immune, metabolic, and cognitive development and that may have mechanistic roles in neurodegeneration. AD and Alzheimer's disease-related dementias (ADRD) are recognized as spectrum disorders with complex pathobiology. AD/ADRD onset begins before overt clinical signs, but initiation triggers remain undefined. We posit that disruption of the normal gut microbiome in early life leads to a pathological cascade within septohippocampal and cortical brain circuits. We propose investigation to understand how early-life microbiota changes may lead to hallmark AD pathology in established AD/ADRD models. Specifically, we hypothesize that antibiotic exposure in early life leads to exacerbated AD-like disease endophenotypes that may be amenable to specific microbiological interventions. We propose suitable models for testing these hypotheses.


Sujet(s)
Maladie d'Alzheimer , Microbiome gastro-intestinal , Animaux , Humains , Maladie d'Alzheimer/microbiologie , Maladie d'Alzheimer/physiopathologie , Antibactériens/administration et posologie , Antibactériens/effets indésirables , Encéphale/microbiologie , Encéphale/anatomopathologie , Encéphale/physiopathologie , Microbiome gastro-intestinal/effets des médicaments et des substances chimiques , Microbiome gastro-intestinal/physiologie , Modèles animaux de maladie humaine , Axe cerveau-intestin/effets des médicaments et des substances chimiques , Axe cerveau-intestin/physiologie
20.
Medicine (Baltimore) ; 103(22): e37825, 2024 May 31.
Article de Anglais | MEDLINE | ID: mdl-39259062

RÉSUMÉ

BACKGROUND: Gut microbial metabolites such as trimethylamine N-oxide (TMAO) and its precursors, namely betaine, L-carnitine, and choline, have been implicated as risk factors for cardiovascular events and mortality development. Therefore, we aim to perform a systematic review and meta-analysis to assess the validity of these associations. METHODS: MEDLINE and Scopus were queried from their inception to August 2023 to identify studies that quantified estimates of the associations of TMAO with the development of major adverse cardiovascular events (MACE) or death. A random-effects meta-analysis was conducted to pool unadjusted or multivariable-adjusted hazard ratios (HR) and their 95% confidence intervals. The primary endpoint was the risk of MACE and all-cause death. RESULTS: 30 prospective observational studies (n = 48 968) were included in the analysis. Elevated TMAO levels were associated with a significantly greater risk of MACE and all-cause death compared to low TMAO levels (HR: 1.41, 95% CI 1.2-1.54, P < .00001, I2 = 43%) and (HR: 1.55, 95% CI 1.37-1.75, P < .00001, I2 = 46%), respectively. Furthermore, high levels of either L-carnitine or choline were found to significantly increase the risk of MACE. However, no significant difference was seen in MACE in either high or low levels of betaine. CONCLUSION: Elevated concentrations of TMAO were associated with increased risks of MACE and all-cause mortality. High levels of L-carnitine/choline were also significantly associated with an increased risk of MACE. However, no significant difference was found between high or low levels of betaine for the outcome of MACE.


Sujet(s)
Maladies cardiovasculaires , Carnitine , Choline , Microbiome gastro-intestinal , Méthylamines , Humains , Bétaïne/métabolisme , Maladies cardiovasculaires/métabolisme , Maladies cardiovasculaires/mortalité , Maladies cardiovasculaires/physiopathologie , Carnitine/métabolisme , Choline/métabolisme , Microbiome gastro-intestinal/physiologie , Méthylamines/métabolisme , Facteurs de risque
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE