Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 8.406
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1374238, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38774627

RESUMO

Gallbladder cancer (GBC) is the most common malignant tumor of the biliary system with the worst prognosis. Even after radical surgery, the majority of patients with GBC have difficulty achieving a clinical cure. The risk of tumor recurrence remains more than 65%, and the overall 5-year survival rate is less than 5%. The gut microbiota refers to a variety of microorganisms living in the human intestine, including bacteria, viruses and fungi, which profoundly affect the host state of general health, disease and even cancer. Over the past few decades, substantial evidence has supported that gut microbiota plays a critical role in promoting the progression of GBC. In this review, we summarize the functions, molecular mechanisms and recent advances of the intestinal microbiota in GBC. We focus on the driving role of bacteria in pivotal pathways, such as virulence factors, metabolites derived from intestinal bacteria, chronic inflammatory responses and ecological niche remodeling. Additionally, we emphasize the high level of correlation between viruses and fungi, especially EBV and Candida spp., with GBC. In general, this review not only provides a solid theoretical basis for the close relationship between gut microbiota and GBC but also highlights more potential research directions for further research in the future.


Assuntos
Bactérias , Neoplasias da Vesícula Biliar , Microbioma Gastrointestinal , Humanos , Neoplasias da Vesícula Biliar/microbiologia , Bactérias/classificação , Bactérias/isolamento & purificação , Animais , Disbiose/microbiologia , Fatores de Virulência , Fungos/patogenicidade , Fungos/classificação
2.
Front Cell Infect Microbiol ; 14: 1352267, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38774629

RESUMO

Hypertensive disorders of pregnancy, including pre-eclampsia, are a leading cause of serious and debilitating complications that affect both the mother and the fetus. Despite the occurrence and the health implications of these disorders there is still relatively limited evidence on the molecular underpinnings of the pathophysiology. An area that has come to the fore with regard to its influence on health and disease is the microbiome. While there are several microbiome niches on and within the body, the distal end of the gut harbors the largest of these impacting on many different systems of the body including the central nervous system, the immune system, and the reproductive system. While the role of the microbiome in hypertensive disorders, including pre-eclampsia, has not been fully elucidated some studies have indicated that several of the symptoms of these disorders are linked to an altered gut microbiome. In this review, we examine both pre-eclampsia and microbiome literature to summarize the current knowledge on whether the microbiome drives the symptoms of pre-eclampsia or if the aberrant microbiome is a consequence of this condition. Despite the paucity of studies, obvious gut microbiome changes have been noted in women with pre-eclampsia and the individual symptoms associated with the condition. Yet further research is required to fully elucidate the role of the microbiome and the significance it plays in the development of the symptoms. Regardless of this, the literature highlights the potential for a microbiome targeted intervention such as dietary changes or prebiotic and probiotics to reduce the impact of some aspects of these disorders.


Assuntos
Microbioma Gastrointestinal , Pré-Eclâmpsia , Pré-Eclâmpsia/microbiologia , Humanos , Gravidez , Feminino , Disbiose/microbiologia , Probióticos , Animais
3.
Sci Rep ; 14(1): 11532, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773342

RESUMO

The presence of dysbiotic cervicovaginal microbiota has been observed to be linked to the persistent development of cervical carcinogenesis mediated by the human papillomavirus (HPV). Nevertheless, the characteristics of the cervical microbiome in individuals diagnosed with cervical cancer (CC) are still not well understood. Comprehensive analysis was conducted by re-analyzing the cervical 16S rRNA sequencing datasets of a total of 507 samples from six previously published studies. We observed significant alpha and beta diversity differences in between CC, cervical intraepithelial neoplasia (CIN) and normal controls (NC), but not between HPV and NC in the combined dataset. Meta-analysis revealed that opportunistic pernicious microbes Streptococcus, Fusobacterium, Pseudomonas and Anaerococcus were enriched in CC, while Lactobacillus was depleted compared to NC. Members of Gardnerella, Sneathia, Pseudomonas, and Fannyhessea have significantly increased relative abundance compared to other bacteria in the CIN group. Five newly identified bacterial genera were found to differentiate CC from NC, with an area under the curve (AUC) of 0.8947. Moreover, co-occurrence network analysis showed that the most commonly encountered Lactobacillus was strongly negatively correlated with Prevotella. Overall, our study identified a set of potential biomarkers for CC from samples across different geographic regions. Our meta-analysis provided significant insights into the characteristics of dysbiotic cervicovaginal microbiota undergoing CC, which may lead to the development of noninvasive CC diagnostic tools and therapeutic interventions.


Assuntos
Disbiose , Microbiota , RNA Ribossômico 16S , Neoplasias do Colo do Útero , Humanos , Feminino , Neoplasias do Colo do Útero/microbiologia , Neoplasias do Colo do Útero/virologia , RNA Ribossômico 16S/genética , Disbiose/microbiologia , Microbiota/genética , Bactérias/genética , Bactérias/classificação , Bactérias/isolamento & purificação , Carcinogênese , Displasia do Colo do Útero/microbiologia , Displasia do Colo do Útero/virologia , Vagina/microbiologia , Colo do Útero/microbiologia , Colo do Útero/patologia
4.
Nature ; 629(8012): 652-659, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38693261

RESUMO

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.


Assuntos
Disbiose , Pai , Microbioma Gastrointestinal , Masculino , Animais , Feminino , Camundongos , Gravidez , Disbiose/microbiologia , Espermatozoides/metabolismo , Testículo/metabolismo , Testículo/microbiologia , Aptidão Genética , Leptina/metabolismo , Camundongos Endogâmicos C57BL , Placenta/microbiologia , Placenta/metabolismo
5.
Food Res Int ; 187: 114345, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38763637

RESUMO

Long-term consumption of Western-style diet (WSD) can lead to metabolic disorders and dysbiosis of gut microbiota, presenting a critical risk factor for various chronic conditions such as fatty liver disease. In the present study, we investigated the beneficial role of co-fermented whole grain quinoa and black barley with Lactobacillus kisonensis on rats fed a WSD. Male Sprague-Dawley (SD) rats, aged six weeks and weighing 180 ± 10 g, were randomly assigned to one of three groups: the normal control group (NC, n = 7), the WSD group (HF, n = 7), and the WSD supplemented with a co-fermented whole grain quinoa with black barley (FQB) intervention group (HFF, n = 7). The findings indicated that FQB was effective in suppressing body weight gain, mitigating hepatic steatosis, reducing perirenal fat accumulation, and ameliorating pathological damage in the livers and testicular tissues of rats. Additionally, FQB intervention led to decreased levels of serum uric acid (UA), aspartate aminotransferase (AST), and alanine aminotransferase (ALT). These advantageous effects can be ascribed to the regulation of FQB on gut microbiota dysbiosis, which includes the restoration of intestinal flora diversity, reduction of the F/B ratio, and promotion of probiotics abundance, such as Akkermansia and [Ruminococcus] at the genus level. The study employed the UPLC-Q-TOF-MSE technique to analyze metabolites in fecal and hepatic samples. The findings revealed that FQB intervention led to a regression in the levels of specific metabolites in feces, including oxoadipic acid and 20a, 22b-dihydroxycholesterol, as well as in the liver, such as pyridoxamine, xanthine and xanthosine. The transcriptome sequencing of liver tissues revealed that FQB intervention modulated the mRNA expression of specific genes, including Cxcl12, Cidea, and Gck, known for their roles in anti-inflammatory and anti-insulin resistance mechanisms in the context of WSD. Our findings indicate that co-fermented whole-grain quinoa with black barley has the potential to alleviate metabolic disorders and chronic inflammation resulting from the consumption of WSD.


Assuntos
Chenopodium quinoa , Dieta Ocidental , Fermentação , Microbioma Gastrointestinal , Hordeum , Lactobacillus , Ratos Sprague-Dawley , Animais , Hordeum/química , Masculino , Lactobacillus/metabolismo , Chenopodium quinoa/química , Ratos , Fígado/metabolismo , Disbiose , Metabolômica , Alimentos Fermentados , Multiômica
6.
BMC Gastroenterol ; 24(1): 158, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38720308

RESUMO

BACKGROUND: Primary sclerosing cholangitis (PSC) is a complex disease with pathogenic mechanisms that remain to be elucidated. Previous observational studies with small sample sizes have reported associations between PSC, dyslipidemia, and gut microbiota dysbiosis. However, the causality of these associations is uncertain, and there has been no systematic analysis to date. METHODS: The datasets comprise data on PSC, 179 lipid species, and 412 gut microbiota species. PSC data (n = 14,890) were sourced from the International PSC Study Group, while the dataset pertaining to plasma lipidomics originated from a study involving 7174 Finnish individuals. Data on gut microbiota species were derived from the Dutch Microbiome Project study, which conducted a genome-wide association study involving 7738 participants. Furthermore, we employed a two-step Mendelian randomization (MR) analysis to quantify the proportion of the effect of gut microbiota-mediated lipidomics on PSC. RESULTS: Following a rigorous screening process, our MR analysis revealed a causal relationship between higher levels of gene-predicted Phosphatidylcholine (O-16:1_18:1) (PC O-16:1_18:1) and an increased risk of developing PSC (inverse variance-weighted method, odds ratio (OR) 1.30, 95% confidence interval (CI) 1.03-1.63). There is insufficient evidence to suggest that gene-predicted PSC impacts the levels of PC O-16:1_18:1 (OR 1.01, 95% CI 0.98-1.05). When incorporating gut microbiota data into the analysis, we found that Eubacterium rectale-mediated genetic prediction explains 17.59% of the variance in PC O-16:1_18:1 levels. CONCLUSION: Our study revealed a causal association between PC O-16:1_18:1 levels and PSC, with a minor portion of the effect mediated by Eubacterium rectale. This study aims to further explore the pathogenesis of PSC and identify promising therapeutic targets. For patients with PSC who lack effective treatment options, the results are encouraging.


Assuntos
Colangite Esclerosante , Microbioma Gastrointestinal , Lipidômica , Análise da Randomização Mendeliana , Humanos , Colangite Esclerosante/sangue , Colangite Esclerosante/microbiologia , Colangite Esclerosante/genética , Microbioma Gastrointestinal/genética , Masculino , Estudo de Associação Genômica Ampla , Feminino , Fosfatidilcolinas/sangue , Disbiose/sangue , Pessoa de Meia-Idade , Adulto
7.
J Transl Med ; 22(1): 433, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38720361

RESUMO

Doxorubicin (DOX) is a broad-spectrum and highly efficient anticancer agent, but its clinical implication is limited by lethal cardiotoxicity. Growing evidences have shown that alterations in intestinal microbial composition and function, namely dysbiosis, are closely linked to the progression of DOX-induced cardiotoxicity (DIC) through regulating the gut-microbiota-heart (GMH) axis. The role of gut microbiota and its metabolites in DIC, however, is largely unelucidated. Our review will focus on the potential mechanism between gut microbiota dysbiosis and DIC, so as to provide novel insights into the pathophysiology of DIC. Furthermore, we summarize the underlying interventions of microbial-targeted therapeutics in DIC, encompassing dietary interventions, fecal microbiota transplantation (FMT), probiotics, antibiotics, and natural phytochemicals. Given the emergence of microbial investigation in DIC, finally we aim to point out a novel direction for future research and clinical intervention of DIC, which may be helpful for the DIC patients.


Assuntos
Cardiotoxicidade , Doxorrubicina , Microbioma Gastrointestinal , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Doxorrubicina/efeitos adversos , Cardiotoxicidade/etiologia , Animais , Disbiose , Transplante de Microbiota Fecal
8.
Food Res Int ; 186: 114404, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38729686

RESUMO

Autism spectrum disorder (ASD) is a neurodevelopmental disorder with an unknown etiology. It is associated with various factors and causes great inconvenience to the patient's life. The gut-brain axis (GBA), which serves as a bidirectional information channel for exchanging information between the gut microbiota and the brain, is vital in studying many neurodegenerative diseases. Dietary flavonoids provide anti-inflammatory and antioxidant benefits, as well as regulating the structure and function of the gut microbiota. The occurrence and development of ASD are associated with dysbiosis of the gut microbiota. Modulation of gut microbiota can effectively improve the severity of ASD. This paper reviews the links between gut microbiota, flavonoids, and ASD, focusing on the mechanism of dietary flavonoids in regulating ASD through the GBA.


Assuntos
Transtorno do Espectro Autista , Eixo Encéfalo-Intestino , Flavonoides , Microbioma Gastrointestinal , Microbioma Gastrointestinal/efeitos dos fármacos , Humanos , Transtorno do Espectro Autista/microbiologia , Transtorno do Espectro Autista/metabolismo , Transtorno do Espectro Autista/dietoterapia , Flavonoides/farmacologia , Dieta , Disbiose , Encéfalo/metabolismo , Animais , Antioxidantes/farmacologia
9.
Sci Rep ; 14(1): 10394, 2024 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710815

RESUMO

Tobacco use significantly influences the oral microbiome. However, less is known about how different tobacco products specifically impact the oral microbiome over time. To address this knowledge gap, we characterized the oral microbiome of cigarette users, smokeless tobacco users, and non-users over 4 months (four time points). Buccal swab and saliva samples (n = 611) were collected from 85 participants. DNA was extracted from all samples and sequencing was carried out on an Illumina MiSeq, targeting the V3-V4 region of the 16S rRNA gene. Cigarette and smokeless tobacco users had more diverse oral bacterial communities, including a higher relative abundance of Firmicutes and a lower relative abundance of Proteobacteria, when compared to non-users. Non-users had a higher relative abundance of Actinomyces, Granulicatella, Haemophilus, Neisseria, Oribacterium, Prevotella, Pseudomonas, Rothia, and Veillonella in buccal swab samples, compared to tobacco users. While the most abundant bacterial genera were relatively constant over time, some species demonstrated significant shifts in relative abundance between the first and last time points. In addition, some opportunistic pathogens were detected among tobacco users including Neisseria subflava, Bulleidia moorei and Porphyromonas endodontalis. Overall, our results provide a more holistic understanding of the structure of oral bacterial communities in tobacco users compared to non-users.


Assuntos
Disbiose , Microbiota , Boca , RNA Ribossômico 16S , Tabaco sem Fumaça , Humanos , Tabaco sem Fumaça/efeitos adversos , Masculino , Feminino , Disbiose/microbiologia , Adulto , RNA Ribossômico 16S/genética , Boca/microbiologia , Saliva/microbiologia , Pessoa de Meia-Idade , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Fumantes , Adulto Jovem , Fumar Cigarros/efeitos adversos , Mucosa Bucal/microbiologia
10.
BMC Microbiol ; 24(1): 161, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730357

RESUMO

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.


Assuntos
Diabetes Gestacional , Disbiose , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Diabetes Gestacional/microbiologia , Diabetes Gestacional/metabolismo , Humanos , Gravidez , Feminino , Disbiose/microbiologia , Ácidos Graxos Voláteis/metabolismo , Bactérias/classificação , Bactérias/genética , Bactérias/metabolismo , Bactérias/isolamento & purificação , Interações entre Hospedeiro e Microrganismos , Lipopolissacarídeos/metabolismo
11.
Respir Res ; 25(1): 204, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730440

RESUMO

BACKGROUND: The impact of cigarette smoke (CS) on lung diseases and the role of microbiome dysbiosis in chronic obstructive pulmonary disease (COPD) have been previously reported; however, the relationships remain unclear. METHODS: Our research examined the effects of 20-week cigarette smoke (CS) exposure on the lung and intestinal microbiomes in C57BL/6JNarl mice, alongside a comparison with COPD patients' intestinal microbiome data from a public dataset. RESULTS: The study found that CS exposure significantly decreased forced vital capacity (FVC), thickened airway walls, and induced emphysema. Increased lung damage was observed along with higher lung keratinocyte chemoattractant (KC) levels by CS exposure. Lung microbiome analysis revealed a rise in Actinobacteriota, while intestinal microbiome showed significant diversity changes, indicating dysbiosis. Principal coordinate analysis highlighted distinct intestinal microbiome compositions between control and CS-exposed groups. In the intestinal microbiome, notable decreases in Patescibacteria, Campilobacterota, Defferibacterota, Actinobacteriota, and Desulfobacterota were observed. We also identified correlations between lung function and dysbiosis in both lung and intestinal microbiomes. Lung interleukins, interferon-É£, KC, and 8-isoprostane levels were linked to lung microbiome dysbiosis. Notably, dysbiosis patterns in CS-exposed mice were similar to those in COPD patients, particularly of Global Initiative for Chronic Obstructive Lung Disease (GOLD) stage 4 patients. This suggests a systemic impact of CS exposure. CONCLUSION: In summary, CS exposure induces significant dysbiosis in lung and intestinal microbiomes, correlating with lung function decline and injury. These results align with changes in COPD patients, underscoring the important role of microbiome in smoke-related lung diseases.


Assuntos
Disbiose , Microbioma Gastrointestinal , Pulmão , Camundongos Endogâmicos C57BL , Doença Pulmonar Obstrutiva Crônica , Animais , Doença Pulmonar Obstrutiva Crônica/microbiologia , Microbioma Gastrointestinal/fisiologia , Camundongos , Humanos , Masculino , Pulmão/microbiologia , Feminino , Pessoa de Meia-Idade , Idoso , Fumaça/efeitos adversos
12.
Front Cell Infect Microbiol ; 14: 1363276, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38707511

RESUMO

Introduction: Chronic kidney disease (CKD) is worldwide healthcare burden with growing incidence and death rate. Emerging evidence demonstrated the compositional and functional differences of gut microbiota in patients with CKD. As such, gut microbial features can be developed as diagnostic biomarkers and potential therapeutic target for CKD. Methods: To eliminate the outcome bias arising from factors such as geographical distribution, sequencing platform, and data analysis techniques, we conducted a comprehensive analysis of the microbial differences between patients with CKD and healthy individuals based on multiple samples worldwide. A total of 980 samples from six references across three nations were incorporated from the PubMed, Web of Science, and GMrepo databases. The obtained 16S rRNA microbiome data were subjected to DADA2 processing, QIIME2 and PICRUSt2 analyses. Results: The gut microbiota of patients with CKD differs significantly from that of healthy controls (HC), with a substantial decrease in the microbial diversity among the CKD group. Moreover, a significantly reduced abundance of bacteria Faecalibacterium prausnitzii (F. prausnitzii) was detected in the CKD group through linear discriminant analysis effect size (LEfSe) analysis, which may be associated with the alleviating effects against CKD. Notably, we identified CKD-depleted F. prausnitzii demonstrated a significant negative correlation with three pathways based on predictive functional analysis, suggesting its potential role in regulating systemic acidbase disturbance and pro-oxidant metabolism. Discussion: Our findings demonstrated notable alterations of gut microbiota in CKD patients. Specific gut-beneficial microbiota, especially F. prausnitzii, may be developed as a preventive and therapeutic tool for CKD clinical management.


Assuntos
Microbioma Gastrointestinal , RNA Ribossômico 16S , Insuficiência Renal Crônica , Microbioma Gastrointestinal/genética , Humanos , RNA Ribossômico 16S/genética , Insuficiência Renal Crônica/microbiologia , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/microbiologia , Filogenia , Faecalibacterium prausnitzii/genética , Biodiversidade , Disbiose/microbiologia
13.
World J Gastroenterol ; 30(16): 2191-2194, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38690026

RESUMO

This editorial explores the intricate relationship between microplastics (MPs) and gut microbiota, emphasizing the complexity and environmental health implications. The gut microbiota, a crucial component of gastrointestinal health, is examined in the context of potential microbial degradation of MPs. Furthermore, dysbiosis induced by MPs emerges as a consensus, disrupting the balance of gut microbiota and decreasing diversity. The mechanisms triggering dysbiosis, including physical interactions and chemical composition, are under investigation. Ongoing research addresses the consequences of MPs on immune fun-ction, nutrient metabolism, and overall host health. The bidirectional relationship between MPs and gut microbiota has significant implications for environmental and human health. Despite uncertainties, MPs negatively impact gut microbiota and health. Further research is essential to unravel the complex interactions and assess the long-term consequences of MPs on both environmental and human well-being.


Assuntos
Disbiose , Microbioma Gastrointestinal , Microplásticos , Microplásticos/efeitos adversos , Humanos , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Saúde Ambiental , Exposição Ambiental/efeitos adversos , Animais
14.
J Clin Invest ; 134(9)2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38690730

RESUMO

The gut microbiota is an integral part of the human metaorganism that is required to shape physiologic host immune responses including host defense against pathogens. Disease-associated gut dysbiosis has been characterized by blooms of pathobionts, which are bacterial species that can drive disease under certain conditions. Pathobionts like Enterobacteriaceae often bloom during flares of inflammatory bowel disease (IBD) and are causally linked with IBD in murine models. In this issue of the JCI, Hecht and colleagues investigated how simple carbohydrates are causally linked to the bloom of the gut pathobiont Klebsiella pneumoniae, which belong to the Enterobacteriaceae family. Notably, the presence of fiber reduced the dissemination of K. pneumoniae into the blood and liver in a colitis model. Their findings provide a diet-related mechanism for gut dysbiosis, which has implications in the management of IBD and other conditions in which gut dysbiosis is an underlying factor.


Assuntos
Disbiose , Microbioma Gastrointestinal , Doenças Inflamatórias Intestinais , Klebsiella pneumoniae , Humanos , Animais , Doenças Inflamatórias Intestinais/microbiologia , Camundongos , Carboidratos da Dieta/efeitos adversos , Infecções por Klebsiella , Colite/induzido quimicamente , Colite/microbiologia , Fibras na Dieta
15.
Elife ; 122024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38690990

RESUMO

Caesarean section scar diverticulum (CSD) is a significant cause of infertility among women who have previously had a Caesarean section, primarily due to persistent inflammatory exudation associated with this condition. Even though abnormal bacterial composition is identified as a critical factor leading to this chronic inflammation, clinical data suggest that a long-term cure is often unattainable with antibiotic treatment alone. In our study, we employed metagenomic analysis and mass spectrometry techniques to investigate the fungal composition in CSD and its interaction with bacteria. We discovered that local fungal abnormalities in CSD can disrupt the stability of the bacterial population and the entire microbial community by altering bacterial abundance via specific metabolites. For instance, Lachnellula suecica reduces the abundance of several Lactobacillus spp., such as Lactobacillus jensenii, by diminishing the production of metabolites like Goyaglycoside A and Janthitrem E. Concurrently, Clavispora lusitaniae and Ophiocordyceps australis can synergistically impact the abundance of Lactobacillus spp. by modulating metabolite abundance. Our findings underscore that abnormal fungal composition and activity are key drivers of local bacterial dysbiosis in CSD.


Assuntos
Bactérias , Cesárea , Cicatriz , Divertículo , Feminino , Cesárea/efeitos adversos , Humanos , Divertículo/microbiologia , Divertículo/metabolismo , Bactérias/metabolismo , Bactérias/genética , Cicatriz/microbiologia , Cicatriz/metabolismo , Disbiose/microbiologia , Fungos/metabolismo , Fungos/genética , Fungos/fisiologia , Interações Microbianas , Microbiota
16.
EBioMedicine ; 103: 105137, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38703606

RESUMO

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).


Assuntos
Doença da Artéria Coronariana , Microbioma Gastrointestinal , Micobioma , Humanos , Doença da Artéria Coronariana/microbiologia , Masculino , Feminino , Pessoa de Meia-Idade , Idoso , Estudos Transversais , Fezes/microbiologia , Metagenômica/métodos , Fungos/genética , Fungos/classificação , Fungos/isolamento & purificação , Índice de Gravidade de Doença , Disbiose/microbiologia , Estudos de Casos e Controles , RNA Ribossômico 16S/genética , Adulto
17.
Gut Microbes ; 16(1): 2348441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38706224

RESUMO

Colorectal cancer (CRC), a malignant tumor worldwide, is associated with gut microbiota. The influence of gut microbe-derived metabolites on CRC has attracted a lot of attention. However, the role of immunity mediated by commensal microbiota-derived metabolites in tumorigenesis of CRC is not intensively explored. Here we monitored the gut microbial dysbiosis in CRC mouse model (ApcMin/+ model) without dietary and pharmacological intervention, followed by characterized of metabolites enriched in CRC model mice. Profound changes of gut microbiome (bacteriome) were observed during intestinal disorders. Metabolomic profiling indicated that agmatine, derived from the gut bacteria i.e. Blautia, Odoribacter, Alistipes and Paraprevotella, could interact with Rnf128 to suppress the Rnf128-mediated ubiquitination of ß-catenin to further upregulate the downstream targets of ß-catenin including Cyclin D1, Lgr5, CD44 and C-myc, thus activating Wnt signaling. The activated Wnt signaling pathway promoted dysplasia of intestinal cells and inflammatory infiltration of lymphocytes via inducing the upregulation of pro-inflammatory cytokines (IL-6 and TNF-α) and downregulation of anti-inflammatory cytokine (IL-10), thereby contributing to colorectal carcinogenesis. Therefore, our study presented novel insights into the roles and mechanisms of gut microbiota in pathogenesis of CRC.


Assuntos
Carcinogênese , Neoplasias Colorretais , Microbioma Gastrointestinal , Inflamação , Via de Sinalização Wnt , Animais , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/metabolismo , Camundongos , Inflamação/metabolismo , Inflamação/microbiologia , Bactérias/classificação , Bactérias/metabolismo , Bactérias/isolamento & purificação , Bactérias/genética , Camundongos Endogâmicos C57BL , beta Catenina/metabolismo , Disbiose/microbiologia , Humanos , Modelos Animais de Doenças , Citocinas/metabolismo , Simbiose , Masculino
18.
Pharm Biol ; 62(1): 356-366, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38720666

RESUMO

CONTEXT: Yi-Shen-Hua-Shi (YSHS) is a traditional Chinese medicine that treats chronic kidney disease (CKD). However, its efficacy in reducing proteinuria and underlying mechanisms is unknown. OBJECTIVE: This single-center randomized controlled trial explored whether YSHS could improve proteinuria and modulate the gut microbiota. MATERIALS AND METHODS: 120 CKD patients were enrolled and randomized to receive the renin-angiotensin-aldosterone system (RAAS) inhibitor plus YSHS (n = 56) or RAAS inhibitor (n = 47) alone for 4 months, and 103 patients completed the study. We collected baseline and follow-up fecal samples and clinical outcomes from participants. Total bacterial DNA was extracted, and the fecal microbiome was analyzed using bioinformatics. RESULTS: Patients in the intervention group had a significantly higher decrease in 24-h proteinuria. After 4 months of the YSHS intervention, the relative abundance of bacteria that have beneficial effects on the body, such as Faecalibacterium, Lachnospiraceae, Lachnoclostridium, and Sutterella increased significantly, while pathogenic bacteria such as the Eggerthella and Clostridium innocuum group decreased. However, we could not find these changes in the control group. Redundancy analysis showed that the decline in 24-h proteinuria during follow-up was significantly correlated with various taxa of gut bacteria, such as Lachnospiraceae and the Lachnoclostridium genus in the YSHS group. KEGG analysis also showed the potential role of YSHS in regulating glycan, lipid, and vitamin metabolism. DISCUSSION AND CONCLUSION: The YSHS granule reduced proteinuria associated with mitigating intestinal microbiota dysbiosis in CKD patients. The definite mechanisms of YSHS to improve proteinuria need to be further explored. TRIAL REGISTRATION: ChiCTR2300076136, retrospectively registered.


Assuntos
Medicamentos de Ervas Chinesas , Disbiose , Microbioma Gastrointestinal , Proteinúria , Insuficiência Renal Crônica , Humanos , Microbioma Gastrointestinal/efeitos dos fármacos , Masculino , Feminino , Insuficiência Renal Crônica/microbiologia , Insuficiência Renal Crônica/tratamento farmacológico , Proteinúria/tratamento farmacológico , Proteinúria/microbiologia , Pessoa de Meia-Idade , Medicamentos de Ervas Chinesas/farmacologia , Fezes/microbiologia , Idoso , Adulto , Medicina Tradicional Chinesa/métodos
19.
Microbiome ; 12(1): 85, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38725043

RESUMO

BACKGROUND: Left ventricular diastolic dysfunction (LVDD) is an important precursor of heart failure (HF), but little is known about its relationship with gut dysbiosis and microbial-related metabolites. By leveraging the multi-omics data from the Hispanic Community Health Study/Study of Latinos (HCHS/SOL), a study with population at high burden of LVDD, we aimed to characterize gut microbiota associated with LVDD and identify metabolite signatures of gut dysbiosis and incident LVDD. RESULTS: We included up to 1996 Hispanic/Latino adults (mean age: 59.4 years; 67.1% female) with comprehensive echocardiography assessments, gut microbiome, and blood metabolome data. LVDD was defined through a composite criterion involving tissue Doppler assessment and left atrial volume index measurements. Among 1996 participants, 916 (45.9%) had prevalent LVDD, and 212 out of 594 participants without LVDD at baseline developed incident LVDD over a median 4.3 years of follow-up. Using multivariable-adjusted analysis of compositions of microbiomes (ANCOM-II) method, we identified 7 out of 512 dominant gut bacterial species (prevalence > 20%) associated with prevalent LVDD (FDR-q < 0.1), with inverse associations being found for Intestinimonas_massiliensis, Clostridium_phoceensis, and Bacteroide_coprocola and positive associations for Gardnerella_vaginali, Acidaminococcus_fermentans, Pseudomonas_aeruginosa, and Necropsobacter_massiliensis. Using multivariable adjusted linear regression, 220 out of 669 circulating metabolites with detection rate > 75% were associated with the identified LVDD-related bacterial species (FDR-q < 0.1), with the majority being linked to Intestinimonas_massiliensis, Clostridium_phoceensis, and Acidaminococcus_fermentans. Furthermore, 46 of these bacteria-associated metabolites, mostly glycerophospholipids, secondary bile acids, and amino acids, were associated with prevalent LVDD (FDR-q < 0.1), 21 of which were associated with incident LVDD (relative risk ranging from 0.81 [p = 0.001, for guanidinoacetate] to 1.25 [p = 9 × 10-5, for 1-stearoyl-2-arachidonoyl-GPE (18:0/20:4)]). The inclusion of these 21 bacterial-related metabolites significantly improved the prediction of incident LVDD compared with a traditional risk factor model (the area under the receiver operating characteristic curve [AUC] = 0.73 vs 0.70, p = 0.001). Metabolite-based proxy association analyses revealed the inverse associations of Intestinimonas_massilliensis and Clostridium_phoceensis and the positive association of Acidaminococcus_fermentans with incident LVDD. CONCLUSION: In this study of US Hispanics/Latinos, we identified multiple gut bacteria and related metabolites linked to LVDD, suggesting their potential roles in this preclinical HF entity. Video Abstract.


Assuntos
Microbioma Gastrointestinal , Hispânico ou Latino , Disfunção Ventricular Esquerda , Humanos , Feminino , Pessoa de Meia-Idade , Masculino , Disfunção Ventricular Esquerda/microbiologia , Disfunção Ventricular Esquerda/sangue , Estados Unidos , Disbiose/microbiologia , Idoso , Bactérias/classificação , Bactérias/isolamento & purificação , Metaboloma , Ecocardiografia
20.
Nutr Diabetes ; 14(1): 31, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773069

RESUMO

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.


Assuntos
Diabetes Gestacional , Disbiose , Microbioma Gastrointestinal , Obesidade , Humanos , Diabetes Gestacional/microbiologia , Gravidez , Feminino , Obesidade/microbiologia , Disbiose/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA