Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.394
Filtrar
1.
BMC Microbiol ; 24(1): 169, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760705

RESUMEN

BACKGROUND: Polycystic ovary syndrome (PCOS) is an endocrinopathy in childbearing-age females which can cause many complications, such as diabetes, obesity, and dyslipidemia. The metabolic disorders in patients with PCOS were linked to gut microbial dysbiosis. However, the correlation between the gut microbial community and dyslipidemia in PCOS remains unillustrated. Our study elucidated the different gut microbiota in patients with PCOS and dyslipidemia (PCOS.D) compared to those with only PCOS and healthy women. RESULTS: In total, 18 patients with PCOS, 16 healthy females, and 18 patients with PCOS.D were enrolled. The 16 S rRNA sequencing in V3-V4 region was utilized for identifying the gut microbiota, which analyzes species annotation, community diversity, and community functions. Our results showed that the ß diversity of gut microbiota did not differ significantly among the three groups. Regarding gut microbiota dysbiosis, patients with PCOS showed a decreased abundance of Proteobacteria, and patients with PCOS.D showed an increased abundance of Bacteroidota compared to other groups. With respect to the gut microbial imbalance at genus level, the PCOS.D group showed a higher abundance of Clostridium_sensu_stricto_1 compared to other two groups. Furthermore, the abundances of Faecalibacterium and Holdemanella were lower in the PCOS.D than those in the PCOS group. Several genera, including Faecalibacterium and Holdemanella, were negatively correlated with the lipid profiles. Pseudomonas was negatively correlated with luteinizing hormone levels. Using PICRUSt analysis, the gut microbiota community functions suggested that certain metabolic pathways (e.g., amino acids, glycolysis, and lipid) were altered in PCOS.D patients as compared to those in PCOS patients. CONCLUSIONS: The gut microbiota characterizations in patients with PCOS.D differ from those in patients with PCOS and controls, and those might also be related to clinical parameters. This may have the potential to become an alternative therapy to regulate the clinical lipid levels of patients with PCOS in the future.


Asunto(s)
Bacterias , Disbiosis , Dislipidemias , Microbioma Gastrointestinal , Síndrome del Ovario Poliquístico , ARN Ribosómico 16S , Humanos , Síndrome del Ovario Poliquístico/microbiología , Femenino , Dislipidemias/microbiología , Adulto , Disbiosis/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , ARN Ribosómico 16S/genética , Adulto Joven , Heces/microbiología
2.
Gut Microbes ; 16(1): 2351503, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38748594

RESUMEN

C-section is crucial in reducing maternal and neonatal mortality when medically indicated, but one of its side effects could be the disruption of vertical transmission of maternal-infant microbiota during delivery, potentially leading to gut dysbiosis and increased disease risks in C-section infants. To address such dysbiosis, it seems reasonable to supplement "what is missing" during C-section procedure. This idea has prompted several clinical trials, including proof-of-concept, investigating interventions like vaginal microbial seeding, oral administration of maternal vaginal microbes and even oral administration of maternal fecal materials. Hereby, we have summarized these trials to help understand the current state of these researches, highlighting the predominantly pilot nature of most of these studies and emphasizing the need for well-designed studies with larger sample to guide evidence-based medicine in the future.


C-section is associated with gut dysbiosis in CS infants and increased disease risks from childhood to adulthood.Apart from using traditional probiotics to restore CS-related dysbiosis, a new research direction is to investigate the potential of mimicking natural inoculation process would alleviate infant gut dysbiosis.Several small-scale studies have shown that transplanting maternal vaginal or even fecal microbiota might restore CS-related infant dysbiosis. Controversy remains regarding the clinical applicability, safety, efficacy and mechanisms of these approaches.


Asunto(s)
Cesárea , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Humanos , Disbiosis/microbiología , Femenino , Cesárea/efectos adversos , Embarazo , Recién Nacido , Vagina/microbiología , Lactante
3.
Nutr Diabetes ; 14(1): 31, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38773069

RESUMEN

OBJECTIVES: The purpose of this review is to investigate the relationship between gastrointestinal microbiome, obesity, and gestational diabetes mellitus (GDM) in an objective manner. METHODS: We conducted a thorough and comprehensive search of the English language literatures published in PubMed, Web of Science, and the Cochrane Library from the establishment of the library until 12 December 2023. Our search strategy included both keywords and free words searches, and we strictly applied inclusion and exclusion criteria. Meta-analyses and systematic reviews were prepared. RESULTS: Six high-quality literature sources were identified for meta-analysis. However, after detailed study and analysis, a certain degree of heterogeneity was found, and the credibility of the combined analysis results was limited. Therefore, descriptive analyses were conducted. The dysbiosis of intestinal microbiome, specifically the ratio of Firmicutes/Bacteroides, is a significant factor in the development of metabolic diseases such as obesity and gestational diabetes. Patients with intestinal dysbiosis and obesity are at a higher risk of developing GDM. CONCLUSIONS: During pregnancy, gastrointestinal microbiome disorders and obesity may contribute to the development of GDM, with all three factors influencing each other. This finding could aid in the diagnosis and management of patients with GDM through further research on their gastrointestinal microbiome.


Asunto(s)
Diabetes Gestacional , Disbiosis , Microbioma Gastrointestinal , Obesidad , Humanos , Diabetes Gestacional/microbiología , Embarazo , Femenino , Obesidad/microbiología , Disbiosis/microbiología
4.
BMC Oral Health ; 24(1): 608, 2024 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38796419

RESUMEN

BACKGROUND: The oral microbiome plays an essential role in maintaining oral homeostasis and health; smoking significantly affects it, leading to microbial dysbiosis. The study aims to investigate changes in the oral microbiome composition of smokers in the Qatari population and establish a correlation with lipid biomarkers. METHODS: The oral microbiota was profiled from saliva samples of 200 smokers and 100 non-smokers in the Qatari population, and 16s rRNA V3-V4 region were sequenced using the Illumina MiSeq platform. The operational taxonomic units (OTUs) were clustered using QIIME and the statistical analysis was performed by R. RESULTS: Non-smokers exhibited a more diverse microbiome, with significant alpha and beta diversity differences between the non-smoker and smoker groups. Smokers had a higher abundance of Firmicutes, Bacteroidota, Actinobacteriota, Patescibacteria, and Proteobacteria at the phylum level and of Streptococcus, Prevotella, Veillonella, TM7x, and Porphyromonas at the genus level. In contrast, non-smokers had more Bacteroidota, Firmicutes, Proteobacteria, Fusobacteriota, and Patescibacteria at the phylum level, and Prevotella, Streptococcus, Veillonella, Porphromonas, and Neisseria at the genus level. Notably, Streptococcus was significantly positively correlated with LDL and negatively correlated with HDL. Additionally, Streptococcus salivarius, within the genus Streptococcus, was substantially more abundant in smokers. CONCLUSION: This study highlights the significant influence of smoking on the composition of the oral microbiome by enriching anaerobic microbes and depleting aerobic microbes. Moreover, the observed correlation between Streptococcus abundance and the lipid biomarkers suggests a potential link between smokers-induced salivary microbiome dysbiosis and lipid metabolism. Understanding the impact of smoking on altering the oral microbiome composition and its correlation with chemistry tests is essential for developing targeted interventions and strategies to improve oral health and reduce the risk of diseases.


Asunto(s)
Biomarcadores , Disbiosis , Microbiota , Saliva , Fumar , Humanos , Saliva/microbiología , Saliva/química , Disbiosis/microbiología , Masculino , Femenino , Biomarcadores/análisis , Adulto , Lípidos/análisis , Persona de Mediana Edad , ARN Ribosómico 16S/análisis
5.
Int J Mol Sci ; 25(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38791443

RESUMEN

Broad-spectrum antibiotics are frequently used to treat bacteria-induced infections, but the overuse of antibiotics may induce the gut microbiota dysbiosis and disrupt gastrointestinal tract function. Probiotics can be applied to restore disturbed gut microbiota and repair abnormal intestinal metabolism. In the present study, two strains of Enterococcus faecium (named DC-K7 and DC-K9) were isolated and characterized from the fecal samples of infant dogs. The genomic features of E. faecium DC-K7 and DC-K9 were analyzed, the carbohydrate-active enzyme (CAZyme)-encoding genes were predicted, and their abilities to produce short-chain fatty acids (SCFAs) were investigated. The bacteriocin-encoding genes in the genome sequences of E. faecium DC-K7 and DC-K9 were analyzed, and the gene cluster of Enterolysin-A, which encoded a 401-amino-acid peptide, was predicted. Moreover, the modulating effects of E. faecium DC-K7 and DC-K9 on the gut microbiota dysbiosis induced by antibiotics were analyzed. The current results demonstrated that oral administrations of E. faecium DC-K7 and DC-K9 could enhance the relative abundances of beneficial microbes and decrease the relative abundances of harmful microbes. Therefore, the isolated E. faecium DC-K7 and DC-K9 were proven to be able to alter the gut microbiota dysbiosis induced by antibiotic treatment.


Asunto(s)
Antibacterianos , Disbiosis , Enterococcus faecium , Microbioma Gastrointestinal , Animales , Disbiosis/microbiología , Microbioma Gastrointestinal/efectos de los fármacos , Antibacterianos/farmacología , Ratones , Heces/microbiología , Ácidos Grasos Volátiles/metabolismo , Probióticos/farmacología , Perros , Bacteriocinas/farmacología
6.
EBioMedicine ; 103: 105137, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38703606

RESUMEN

BACKGROUND: Coronary artery disease (CAD) is a prevalent cardiovascular condition, and numerous studies have linked gut bacterial imbalance to CAD. However, the relationship of gut fungi, another essential component of the intestinal microbiota, with CAD remains poorly understood. METHODS: In this cross-sectional study, we analyzed fecal samples from 132 participants, split into 31 healthy controls and 101 CAD patients, further categorized into stable CAD (38), unstable angina (41), and acute myocardial infarction (22) groups. We conducted internal transcribed spacer 1 (ITS1) and 16S sequencing to examine gut fungal and bacterial communities. FINDINGS: Based on ITS1 analyses, Ascomycota and Basidiomycota were the dominant fungal phyla in all the groups. The α diversity of gut mycobiome remained unaltered among the control group and CAD subgroups; however, the structure and composition of the mycobiota differed significantly with the progression of CAD. The abundances of 15 taxa gradually changed with the occurrence and progression of the disease and were significantly correlated with major CAD risk factor indicators. The mycobiome changes were closely linked to gut microbiome dysbiosis in patients with CAD. Furthermore, disease classifiers based on gut fungi effectively identified subgroups with different degrees of CAD. Finally, the FUNGuild analysis further categorized these fungi into distinct ecological guilds. INTERPRETATION: In conclusion, the structure and composition of the gut fungal community differed from healthy controls to various subtypes of CAD, revealing key fungi taxa alterations linked to the onset and progression of CAD. Our study highlights the potential role of gut fungi in CAD and may facilitate the development of novel biomarkers and therapeutic targets for CAD. FUNDING: This work was supported by the grants from the National Natural Science Foundation of China (No. 82170302, 92168117, 82370432), National clinical key specialty construction project- Cardiovascular Surgery, the Reform and Development Program of Beijing Institute of Respiratory Medicine (No. Ggyfz202417, Ggyfz202308), the Beijing Natural Science Foundation (No. 7222068); and the Clinical Research Incubation Program of Beijing Chaoyang Hospital Affiliated to Capital Medical University (No. CYFH202209).


Asunto(s)
Enfermedad de la Arteria Coronaria , Microbioma Gastrointestinal , Micobioma , Humanos , Enfermedad de la Arteria Coronaria/microbiología , Masculino , Femenino , Persona de Mediana Edad , Anciano , Estudios Transversales , Heces/microbiología , Metagenómica/métodos , Hongos/genética , Hongos/clasificación , Hongos/aislamiento & purificación , Índice de Severidad de la Enfermedad , Disbiosis/microbiología , Estudios de Casos y Controles , ARN Ribosómico 16S/genética , Adulto
7.
BMC Microbiol ; 24(1): 161, 2024 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-38730357

RESUMEN

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


Asunto(s)
Diabetes Gestacional , Disbiosis , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Diabetes Gestacional/microbiología , Diabetes Gestacional/metabolismo , Humanos , Embarazo , Femenino , Disbiosis/microbiología , Ácidos Grasos Volátiles/metabolismo , Bacterias/clasificación , Bacterias/genética , Bacterias/metabolismo , Bacterias/aislamiento & purificación , Interacciones Microbiota-Huesped , Lipopolisacáridos/metabolismo
8.
Int J Mol Sci ; 25(9)2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38732048

RESUMEN

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


Asunto(s)
Heces , Microbioma Gastrointestinal , Infecciones por VIH , ARN Ribosómico 16S , Humanos , Infecciones por VIH/microbiología , Infecciones por VIH/tratamiento farmacológico , México , Femenino , Masculino , Adulto , Persona de Mediana Edad , Heces/microbiología , ARN Ribosómico 16S/genética , Disbiosis/microbiología , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/análisis , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Butiratos/metabolismo
9.
PLoS One ; 19(5): e0302726, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743706

RESUMEN

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


Asunto(s)
Bacteroides , Diabetes Gestacional , Microbioma Gastrointestinal , ARN Ribosómico 16S , Veillonella , Humanos , Diabetes Gestacional/microbiología , Femenino , Embarazo , Bacteroides/genética , ARN Ribosómico 16S/genética , Veillonella/genética , Lactante , Adulto , Masculino , Disbiosis/microbiología , Heces/microbiología , Preescolar , Secuenciación de Nucleótidos de Alto Rendimiento
10.
J Appl Microbiol ; 135(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38755019

RESUMEN

AIM: The present study is a single-centre, randomized, controlled clinical trial aimed to evaluate the effectiveness of the probiotic Lacticaseibacillus rhamnosus TOM 22.8 (DSM 33500) strain, orally administrated, to treat vaginal dysbiosis. METHODS AND RESULTS: Overall, 80 women, with signs and symptoms of vaginal dysbiosis, were enrolled and allocated to the treatment group (A, n=60), who took 1 capsule of the probiotic strain for 10 consecutive days, or the non-treatment group (B, n=20), who did not receive any treatment. Clinical (vaginal signs and symptoms; pH of the vaginal fluid; Amsel criteria; Nugent score; Lactobacillary grade) and microbiological examinations were performed at baseline (T0), 10 days (T1), and 30 (T2) days after the oral administration of the probiotic TOM 22.8 strain. The latter resulted in a restoration of the physiological pH, accompanied by remission or attenuation of clinical signs and symptoms as well as the improvement of the quality of life (QoL). Microbiological data revealed a significant reduction of potentially pathogenic bacteria. CONCLUSION: The administration of the L. rhamnosus TOM 22.8 probiotic strain could be proposed as an effective strategy for the treatment of vaginal dysbiosis.


Asunto(s)
Disbiosis , Lacticaseibacillus rhamnosus , Probióticos , Vagina , Femenino , Humanos , Probióticos/administración & dosificación , Probióticos/uso terapéutico , Disbiosis/microbiología , Vagina/microbiología , Adulto , Persona de Mediana Edad , Adulto Joven , Calidad de Vida , Lactobacillus , Vaginosis Bacteriana/microbiología , Vaginosis Bacteriana/tratamiento farmacológico
11.
J Nanobiotechnology ; 22(1): 269, 2024 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-38764018

RESUMEN

Symbiotic microbial communities are crucial for human health, and dysbiosis is associated with various diseases. Plant-derived nanovesicles (PDNVs) have a lipid bilayer structure and contain lipids, metabolites, proteins, and RNA. They offer unique advantages in regulating microbial community homeostasis and treating diseases related to dysbiosis compared to traditional drugs. On the one hand, lipids on PDNVs serve as the primary substances that mediate specific recognition and uptake by bacteria. On the other hand, due to the multifactorial nature of PDNVs, they have the potential to enhance growth and survival of beneficial bacterial while simultaneously reducing the pathogenicity of harmful bacteria. In addition, PDNVs have the capacity to modulate bacterial metabolism, thus facilitating the establishment of a harmonious microbial equilibrium and promoting stability within the microbiota. These remarkable attributes make PDNVs a promising therapeutic approach for various conditions, including periodontitis, inflammatory bowel disease, and skin infection diseases. However, challenges such as consistency, isolation methods, and storage need to be addressed before clinical application. This review aims to explore the value of PDNVs in regulating microbial community homeostasis and provide recommendations for their use as novel therapeutic agents for health protection.


Asunto(s)
Microbiota , Humanos , Plantas , Bacterias/metabolismo , Disbiosis/microbiología , Animales , Nanopartículas/química , Nanoestructuras/química , Periodontitis/microbiología
12.
Int J Mol Sci ; 25(10)2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38791599

RESUMEN

This review scrutinizes the intricate interplay between the microbiome and the human body, exploring its multifaceted dimensions and far-reaching implications. The human microbiome, comprising diverse microbial communities inhabiting various anatomical niches, is increasingly recognized as a critical determinant of human health and disease. Through an extensive examination of current research, this review elucidates the dynamic interactions between the microbiome and host physiology across multiple organ systems. Key topics include the establishment and maintenance of microbiota diversity, the influence of host factors on microbial composition, and the bidirectional communication pathways between microbiota and host cells. Furthermore, we delve into the functional implications of microbiome dysbiosis in disease states, emphasizing its role in shaping immune responses, metabolic processes, and neurological functions. Additionally, this review discusses emerging therapeutic strategies aimed at modulating the microbiome to restore host-microbe homeostasis and promote health. Microbiota fecal transplantation represents a groundbreaking therapeutic approach in the management of dysbiosis-related diseases, offering a promising avenue for restoring microbial balance within the gut ecosystem. This innovative therapy involves the transfer of fecal microbiota from a healthy donor to an individual suffering from dysbiosis, aiming to replenish beneficial microbial populations and mitigate pathological imbalances. By synthesizing findings from diverse fields, this review offers valuable insights into the complex relationship between the microbiome and the human body, highlighting avenues for future research and clinical interventions.


Asunto(s)
Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Humanos , Disbiosis/microbiología , Disbiosis/terapia , Microbiota , Animales , Cuerpo Humano , Interacciones Microbiota-Huesped/fisiología
13.
Nature ; 629(8012): 652-659, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693261

RESUMEN

The gut microbiota operates at the interface of host-environment interactions to influence human homoeostasis and metabolic networks1-4. Environmental factors that unbalance gut microbial ecosystems can therefore shape physiological and disease-associated responses across somatic tissues5-9. However, the systemic impact of the gut microbiome on the germline-and consequently on the F1 offspring it gives rise to-is unexplored10. Here we show that the gut microbiota act as a key interface between paternal preconception environment and intergenerational health in mice. Perturbations to the gut microbiota of prospective fathers increase the probability of their offspring presenting with low birth weight, severe growth restriction and premature mortality. Transmission of disease risk occurs via the germline and is provoked by pervasive gut microbiome perturbations, including non-absorbable antibiotics or osmotic laxatives, but is rescued by restoring the paternal microbiota before conception. This effect is linked with a dynamic response to induced dysbiosis in the male reproductive system, including impaired leptin signalling, altered testicular metabolite profiles and remapped small RNA payloads in sperm. As a result, dysbiotic fathers trigger an elevated risk of in utero placental insufficiency, revealing a placental origin of mammalian intergenerational effects. Our study defines a regulatory 'gut-germline axis' in males, which is sensitive to environmental exposures and programmes offspring fitness through impacting placenta function.


Asunto(s)
Disbiosis , Padre , Microbioma Gastrointestinal , Masculino , Animales , Femenino , Ratones , Embarazo , Disbiosis/microbiología , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/microbiología , Aptitud Genética , Leptina/metabolismo , Ratones Endogámicos C57BL , Placenta/microbiología , Placenta/metabolismo
14.
Nat Commun ; 15(1): 4232, 2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38762479

RESUMEN

Toll-like receptor 9 (TLR9) recognizes bacterial, viral and self DNA and play an important role in immunity and inflammation. However, the role of TLR9 in obesity is less well-studied. Here, we generate B-cell-specific Tlr9-deficient (Tlr9fl/fl/Cd19Cre+/-, KO) B6 mice and model obesity using a high-fat diet. Compared with control mice, B-cell-specific-Tlr9-deficient mice exhibited increased fat tissue inflammation, weight gain, and impaired glucose and insulin tolerance. Furthermore, the frequencies of IL-10-producing-B cells and marginal zone B cells were reduced, and those of follicular and germinal center B cells were increased. This was associated with increased frequencies of IFNγ-producing-T cells and increased follicular helper cells. In addition, gut microbiota from the KO mice induced a pro-inflammatory state leading to immunological and metabolic dysregulation when transferred to germ-free mice. Using 16 S rRNA gene sequencing, we identify altered gut microbial communities including reduced Lachnospiraceae, which may play a role in altered metabolism in KO mice. We identify an important network involving Tlr9, Irf4 and Il-10 interconnecting metabolic homeostasis, with the function of B and T cells, and gut microbiota in obesity.


Asunto(s)
Linfocitos B , Dieta Alta en Grasa , Disbiosis , Microbioma Gastrointestinal , Inflamación , Interleucina-10 , Ratones Noqueados , Obesidad , Receptor Toll-Like 9 , Animales , Obesidad/inmunología , Obesidad/microbiología , Obesidad/metabolismo , Disbiosis/inmunología , Disbiosis/microbiología , Receptor Toll-Like 9/metabolismo , Receptor Toll-Like 9/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Inflamación/metabolismo , Ratones , Dieta Alta en Grasa/efectos adversos , Interleucina-10/metabolismo , Masculino , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad , Factores Reguladores del Interferón
15.
Gut Microbes ; 16(1): 2356284, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38769683

RESUMEN

Inflammatory bowel disease (IBD) is a chronic and recurrent condition affecting the gastrointestinal tract. Disturbed gut microbiota and abnormal bile acid (BA) metabolism are notable in IBD, suggesting a bidirectional relationship. Specifically, the diversity of the gut microbiota influences BA composition, whereas altered BA profiles can disrupt the microbiota. IBD patients often exhibit increased primary bile acid and reduced secondary bile acid concentrations due to a diminished bacteria population essential for BA metabolism. This imbalance activates BA receptors, undermining intestinal integrity and immune function. Consequently, targeting the microbiota-BA axis may rectify these disturbances, offering symptomatic relief in IBD. Here, the interplay between gut microbiota and bile acids (BAs) is reviewed, with a particular focus on the role of gut microbiota in mediating bile acid biotransformation, and contributions of the gut microbiota-BA axis to IBD pathology to unveil potential novel therapeutic avenues for IBD.


Asunto(s)
Bacterias , Ácidos y Sales Biliares , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/metabolismo , Humanos , Ácidos y Sales Biliares/metabolismo , Animales , Bacterias/metabolismo , Bacterias/clasificación , Bacterias/genética , Disbiosis/microbiología , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/metabolismo
16.
Gut Microbes ; 16(1): 2351478, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38780485

RESUMEN

For many years, it has been hypothesized that pathological changes to the gut microbiome in critical illness is a driver of infections, organ dysfunction, and other adverse outcomes in the intensive care unit (ICU). The advent of contemporary microbiome methodologies and multi-omics tools have allowed researchers to test this hypothesis by dissecting host-microbe interactions in the gut to better define its contribution to critical illness pathogenesis. Observational studies of patients in ICUs have revealed that gut microbial communities are profoundly altered in critical illness, characterized by markedly reduced alpha diversity, loss of commensal taxa, and expansion of potential pathogens. These key features of ICU gut dysbiosis have been associated with adverse outcomes including life-threatening hospital-acquired (nosocomial) infections. Current research strives to define cellular and molecular mechanisms connecting gut dysbiosis with infections and other outcomes, and to identify opportunities for therapeutic modulation of host-microbe interactions. This review synthesizes evidence from studies of critically ill patients that have informed our understanding of intestinal dysbiosis in the ICU, mechanisms linking dysbiosis to infections and other adverse outcomes, as well as clinical trials of microbiota-modifying therapies. Additionally, we discuss novel avenues for precision microbial therapeutics to combat nosocomial infections and other life-threatening complications of critical illness.


Asunto(s)
Enfermedad Crítica , Infección Hospitalaria , Disbiosis , Microbioma Gastrointestinal , Disbiosis/microbiología , Humanos , Infección Hospitalaria/microbiología , Infección Hospitalaria/tratamiento farmacológico , Unidades de Cuidados Intensivos , Animales , Interacciones Microbiota-Huesped , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación
17.
Gut Microbes ; 16(1): 2357177, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38781112

RESUMEN

The prevalence of eating disorders has been increasing over the last 50 years. Binge eating disorder (BED) and bulimia nervosa (BN) are two typical disabling, costly and life-threatening eating disorders that substantially compromise the physical well-being of individuals while undermining their psychological functioning. The distressing and recurrent episodes of binge eating are commonly observed in both BED and BN; however, they diverge as BN often involves the adoption of inappropriate compensatory behaviors aimed at averting weight gain. Normal eating behavior is coordinated by a well-regulated trade-off between intestinal and central ingestive mechanism. Conversely, despite the fact that the etiology of BED and BN remains incompletely resolved, emerging evidence corroborates the notion that dysbiosis of gastrointestinal microbiome and its metabolites, alteration of gut-brain axis, as well as malfunctioning central circuitry regulating motivation, execution and reward all contribute to the pathology of binge eating. In this review, we aim to outline the current state of knowledge pertaining to the potential mechanisms through which each component of the gut-brain axis participates in binge eating behaviors, and provide insight for the development of microbiome-based therapeutic interventions that hold promise in ameliorating patients afflicted with binge eating disorders.


Asunto(s)
Trastorno por Atracón , Eje Cerebro-Intestino , Encéfalo , Disbiosis , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiología , Humanos , Trastorno por Atracón/microbiología , Trastorno por Atracón/fisiopatología , Trastorno por Atracón/metabolismo , Eje Cerebro-Intestino/fisiología , Encéfalo/microbiología , Encéfalo/fisiopatología , Animales , Disbiosis/microbiología , Conducta Alimentaria
18.
Adv Protein Chem Struct Biol ; 140: 199-248, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38762270

RESUMEN

The human gut microbiota is a complex and dynamic community of microorganisms, that influence metabolic, neurodevelopmental, and immune pathways. Microbial dysbiosis, characterized by changes in microbial diversity and relative abundances, is implicated in the development of various chronic neurological and neurodegenerative disorders. These disorders are marked by the accumulation of pathological protein aggregates, leading to the progressive loss of neurons and behavioural functions. Dysregulations in protein-protein interaction networks and signalling complexes, critical for normal brain function, are common in neurological disorders but challenging to unravel, particularly at the neuron and synapse-specific levels. To advance therapeutic strategies, a deeper understanding of neuropathogenesis, especially during the progressive disease phase, is needed. Biomarkers play a crucial role in identifying disease pathophysiology and monitoring disease progression. Proteomics, a powerful technology, shows promise in accelerating biomarker discovery and aiding in the development of novel treatments. In this chapter, we provide an in-depth overview of how proteomic techniques, utilizing various biofluid samples from patients with neurological conditions and diverse animal models, have contributed valuable insights into the pathogenesis of numerous neurological disorders. We also discuss the current state of research, potential challenges, and future directions in proteomic approaches to unravel neuro-pathological conditions.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Proteómica , Humanos , Disbiosis/metabolismo , Disbiosis/microbiología , Enfermedades del Sistema Nervioso/metabolismo , Enfermedades del Sistema Nervioso/microbiología , Animales , Eje Cerebro-Intestino , Biomarcadores/metabolismo
19.
Front Cell Infect Microbiol ; 14: 1366192, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779566

RESUMEN

Background: Ulcerative colitis (UC) is a multifactorial chronic inflammatory bowel disease (IBD) that affects the large intestine with superficial mucosal inflammation. A dysbiotic gut microbial profile has been associated with UC. Our study aimed to characterize the UC gut bacterial, fungal, and metabolic fingerprints by omic approaches. Methods: The 16S rRNA- and ITS2-based metataxonomics and gas chromatography-mass spectrometry/solid phase microextraction (GC-MS/SPME) metabolomic analysis were performed on stool samples of 53 UC patients and 37 healthy subjects (CTRL). Univariate and multivariate approaches were applied to separated and integrated omic data, to define microbiota, mycobiota, and metabolic signatures in UC. The interaction between gut bacteria and fungi was investigated by network analysis. Results: In the UC cohort, we reported the increase of Streptococcus, Bifidobacterium, Enterobacteriaceae, TM7-3, Granulicatella, Peptostreptococcus, Lactobacillus, Veillonella, Enterococcus, Peptoniphilus, Gemellaceae, and phenylethyl alcohol; and we also reported the decrease of Akkermansia; Ruminococcaceae; Ruminococcus; Gemmiger; Methanobrevibacter; Oscillospira; Coprococus; Christensenellaceae; Clavispora; Vishniacozyma; Quambalaria; hexadecane; cyclopentadecane; 5-hepten-2-ol, 6 methyl; 3-carene; caryophyllene; p-Cresol; 2-butenal; indole, 3-methyl-; 6-methyl-3,5-heptadiene-2-one; 5-octadecene; and 5-hepten-2-one, 6 methyl. The integration of the multi-omic data confirmed the presence of a distinctive bacterial, fungal, and metabolic fingerprint in UC gut microbiota. Moreover, the network analysis highlighted bacterial and fungal synergistic and/or divergent interkingdom interactions. Conclusion: In this study, we identified intestinal bacterial, fungal, and metabolic UC-associated biomarkers. Furthermore, evidence on the relationships between bacterial and fungal ecosystems provides a comprehensive perspective on intestinal dysbiosis and ecological interactions between microorganisms in the framework of UC.


Asunto(s)
Bacterias , Colitis Ulcerosa , Heces , Hongos , Cromatografía de Gases y Espectrometría de Masas , Microbioma Gastrointestinal , Metabolómica , ARN Ribosómico 16S , Humanos , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/metabolismo , Masculino , Adulto , Femenino , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Bacterias/genética , Persona de Mediana Edad , Metabolómica/métodos , ARN Ribosómico 16S/genética , Heces/microbiología , Hongos/clasificación , Hongos/aislamiento & purificación , Hongos/metabolismo , Disbiosis/microbiología , Metaboloma , Anciano , Adulto Joven , Microextracción en Fase Sólida , Micobioma , Multiómica
20.
Mol Med Rep ; 30(1)2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38785152

RESUMEN

The gut­microbiota­brain axis is a complex bidirectional communication system linking the gastrointestinal tract to the brain. Changes in the balance, composition and diversity of the gut­microbiota (gut dysbiosis) have been found to be associated with the development of psychosis. Early­life stress, along with various stressors encountered in different developmental phases, have been shown to be associated with the abnormal composition of the gut microbiota, leading to irregular immunological and neuroendocrine functions, which are potentially responsible for the occurrence of first­episode psychosis (FEP). The aim of the present narrative review was to summarize the significant differences of the altered microbiome composition in patients suffering from FEP vs. healthy controls, and to discuss its effects on the occurrence and intensity of symptoms in FEP.


Asunto(s)
Disbiosis , Microbioma Gastrointestinal , Trastornos Psicóticos , Humanos , Disbiosis/microbiología , Trastornos Psicóticos/microbiología , Eje Cerebro-Intestino/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA