Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.351
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(37): e2314337121, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39226363

RESUMEN

Epidemiological studies have revealed an inverse relationship between the incidence of Alzheimer's disease (AD) and various cancers, including colorectal cancer (CRC). We aimed to determine whether the incidence of CRC is reduced in AD-like mice and whether gut microbiota confers resistance to tumorigenesis through inducing inflammatory tolerance using 16S ribosomal RNA gene sequencing and fecal microbiota transplantation (FMT). AD-like mice experienced a significantly decreased incidence of CRC tumorigenesis induced by azoxymethane-dextran sodium sulfate as evidenced by suppressed intestinal inflammation compared with control mice. However, FMT from age-matched control mice reversed the inhibitory effects on the tumorigenesis of CRC and inflammatory response in AD-like mice. The key bacterial genera in gut microbiota, including Prevotella, were increased in both the AD-like mice and in patients with amnestic mild cognitive impairment (aMCI) but were decreased in patients with CRC. Pretreatment with low-dose Prevotella-derived lipopolysaccharides (LPS) induced inflammatory tolerance both in vivo and in vitro and inhibited CRC tumorigenesis in mice. Imbalanced gut microbiota increased intestinal barrier permeability, which facilitated LPS absorption from the gut into the blood, causing cognitive decline in AD-like mice and patients with aMCI. These data reveal that intestinal Prevotella-derived LPS exerts a resistant effect to CRC tumorigenesis via inducing inflammatory tolerance in the presence of AD. These findings provide biological evidence demonstrating the inverse relationship between the incidence of AD and CRC.


Asunto(s)
Enfermedad de Alzheimer , Neoplasias Colorrectales , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Animales , Enfermedad de Alzheimer/microbiología , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/patología , Ratones , Humanos , Masculino , Inflamación , Disfunción Cognitiva , Femenino , Prevotella , Modelos Animales de Enfermedad , Lipopolisacáridos , Carcinogénesis , Sulfato de Dextran
2.
Carbohydr Polym ; 344: 122535, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39218555

RESUMEN

Oat ß-(1 â†’ 3, 1 â†’ 4)-d-glucan (OBG), a linear polysaccharide primarily found in oat bran, has been demonstrated to possess immunomodulatory properties and regulate gut microbiota. This study aimed to investigate the impact of low molecular weight (Mw) OBG (155.2 kDa) on colonic injury and allergic symptoms induced by food allergy (FA), and to explore its potential mechanism. In Experiment 1, results indicated that oral OBG improved colonic inflammation and epithelial barrier, and significantly relieved allergy symptoms. Importantly, the OBG supplement altered the gut microbiota composition, particularly increasing the abundance of Lachnospiraceae and its genera, and promoted the production of short-chain fatty acids, especially butyrate. However, in Experiment 2, the gut microbial depletion eliminated these protective effects of OBG on the colon in allergic mice. Further, in Experiment 3, fecal microbiota transplantation and sterile fecal filtrate transfer directly validated the role of OBG-mediated gut microbiota and its metabolites in relieving FA and its induced colonic injury. Our findings suggest that low Mw OBG can alleviate FA-induced colonic damage by increasing Lachnospiraceae abundance and butyrate production, and provide novel insights into the health benefits and mechanisms of dietary polysaccharide intervention for FA.


Asunto(s)
Avena , Butiratos , Colon , Hipersensibilidad a los Alimentos , Microbioma Gastrointestinal , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Colon/patología , Colon/efectos de los fármacos , Colon/metabolismo , Butiratos/metabolismo , Avena/química , Clostridiales , beta-Glucanos/farmacología , beta-Glucanos/química , Ratones Endogámicos BALB C , Masculino , Glucanos/farmacología , Glucanos/química , Ácidos Grasos Volátiles/metabolismo , Trasplante de Microbiota Fecal
3.
Theranostics ; 14(12): 4622-4642, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39239516

RESUMEN

Rationale: Consumption of a high-fat diet (HFD) has been implicated in cognitive deficits and gastrointestinal dysfunction in humans, with the gut microbiota emerging as a pivotal mediator of these diet-associated pathologies. The introduction of plant-based polysaccharides into the diet as a therapeutic strategy to alleviate such conditions is gaining attention. Nevertheless, the mechanistic paradigm by which polysaccharides modulate the gut microbiota remains largely undefined. This study investigated the mechanisms of action of Eucommiae cortex polysaccharides (EPs) in mitigating gut dysbiosis and examined their contribution to rectifying diet-related cognitive decline. Methods: Initially, we employed fecal microbiota transplantation (FMT) and gut microbiota depletion to verify the causative role of changes in the gut microbiota induced by HFD in synapse engulfment-dependent cognitive impairments. Subsequently, colonization of the gut of chow-fed mice with Escherichia coli (E. coli) from HFD mice confirmed that inhibition of Proteobacteria by EPs was a necessary prerequisite for alleviating HFD-induced cognitive impairments. Finally, supplementation of HFD mice with butyrate and treatment of EPs mice with GW9662 demonstrated that EPs inhibited the expansion of Proteobacteria in the colon of HFD mice by reshaping the interactions between the gut microbiota and colonocytes. Results: Findings from FMT and antibiotic treatments demonstrated that HFD-induced cognitive impairments pertaining to neuronal spine loss were contingent on gut microbial composition. Association analysis revealed strong associations between bacterial taxa belonging to the phylum Proteobacteria and cognitive performance in mice. Further, introducing E. coli from HFD-fed mice into standard diet-fed mice underscored the integral role of Proteobacteria proliferation in triggering excessive synaptic engulfment-related cognitive deficits in HFD mice. Crucially, EPs effectively counteracted the bloom of Proteobacteria and subsequent neuroinflammatory responses mediated by microglia, essential for cognitive improvement in HFD-fed mice. Mechanistic insights revealed that EPs promoted the production of bacteria-derived butyrate, thereby ameliorating HFD-induced colonic mitochondrial dysfunction and reshaping colonocyte metabolism. This adjustment curtailed the availability of growth substrates for facultative anaerobes, which in turn limited the uncontrolled expansion of Proteobacteria. Conclusions: Our study elucidates that colonocyte metabolic disturbances, which promote Proteobacteria overgrowth, are a likely cause of HFD-induced cognitive deficits. Furthermore, dietary supplementation with EPs can rectify behavioral dysfunctions associated with HFD by modifying gut microbiota-colonocyte interactions. These insights contribute to the broader understanding of the modulatory effects of plant prebiotics on the microbiota-gut-brain axis and suggest a potential therapeutic avenue for diet-associated cognitive dysfunction.


Asunto(s)
Disfunción Cognitiva , Dieta Alta en Grasa , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Polisacáridos , Microbioma Gastrointestinal/efectos de los fármacos , Animales , Dieta Alta en Grasa/efectos adversos , Ratones , Disfunción Cognitiva/terapia , Polisacáridos/farmacología , Masculino , Disbiosis/terapia , Colon/microbiología , Escherichia coli , Butiratos/metabolismo , Proteobacteria/aislamiento & purificación , Proteobacteria/efectos de los fármacos , Modelos Animales de Enfermedad
4.
Gut Microbes ; 16(1): 2393766, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39224076

RESUMEN

Clostridioides difficile is a major nosocomial pathogen, causing significant morbidity and mortality worldwide. Antibiotic usage, a major risk factor for Clostridioides difficile infection (CDI), disrupts the gut microbiota, allowing C. difficile to proliferate and cause infection, and can often lead to recurrent CDI (rCDI). Fecal microbiota transplantation (FMT) and live biotherapeutic products (LBPs) have emerged as effective treatments for rCDI and aim to restore colonization resistance provided by a healthy gut microbiota. However, much is still unknown about the mechanisms mediating their success. Bile acids, extensively modified by gut microbes, affect C. difficile's germination, growth, and toxin production while also shaping the gut microbiota and influencing host immune responses. Additionally, microbial interactions, such as nutrient competition and cross-feeding, contribute to colonization resistance against C. difficile and may contribute to the success of microbiota-focused therapeutics. Bile acids as well as other microbial mediated interactions could have implications for other diseases being treated with microbiota-focused therapeutics. This review focuses on the intricate interplay between bile acid modifications, microbial ecology, and host responses with a focus on C. difficile, hoping to shed light on how to move forward with the development of new microbiota mediated therapeutic strategies to combat rCDI and other intestinal diseases.


Asunto(s)
Ácidos y Sales Biliares , Clostridioides difficile , Infecciones por Clostridium , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Ácidos y Sales Biliares/metabolismo , Humanos , Clostridioides difficile/fisiología , Infecciones por Clostridium/terapia , Infecciones por Clostridium/microbiología , Animales
5.
Age Ageing ; 53(8)2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-39141079

RESUMEN

BACKGROUND: Advanced age has been widely identified as a risk factor for recurrent Clostridioides difficile infection (CDI), but most related studies were performed before the introduction of novel therapies. The aim of this study was to compare CDI characteristics and outcomes in patients over and under 80 years old with CDI and their outcomes in the era of new treatments. METHODS: This was a retrospective cohort study of patients diagnosed with CDI from January 2021 to December 2022 in an academic hospital. We compared recurrence and mortality at 12 weeks after the end of treatment. An extension of the Fine and Grey model adjusted for competing events was used to assess the effect of age on recurrence. RESULTS: Four hundred seventy-six patients were considered to have CDI (320 in patients <80 years and 156 in ≥80 years). CDI in older patients was more frequently healthcare-associated and was more severe. Although the Charlson index was almost identical between populations, comorbidities clearly differed. New treatments (bezlotoxumab, fidaxomicin and faecal microbiota transplantation) were more frequently used in older patients without statistical significance (41.3% vs. 33.4%, P = .053). There were 69 (14.5%) recurrences, with no differences by age group after adjusting for competing events. Mortality was greater in the oldest (35.3%) than in the youngest (13.1%); P < .001. CONCLUSIONS: No differences in CDI recurrence rates were found between age groups. However, there was a high mortality rate in patients ≥80 years old, which emphasises the urgent need to improve the prevention and treatment of CDI in this group.


Asunto(s)
Infecciones por Clostridium , Recurrencia , Humanos , Masculino , Anciano de 80 o más Años , Infecciones por Clostridium/epidemiología , Infecciones por Clostridium/mortalidad , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/terapia , Infecciones por Clostridium/diagnóstico , Infecciones por Clostridium/tratamiento farmacológico , Femenino , Estudios Retrospectivos , Factores de Riesgo , Anciano , Factores de Edad , Trasplante de Microbiota Fecal , Antibacterianos/uso terapéutico , Clostridioides difficile , Persona de Mediana Edad , Fidaxomicina/uso terapéutico , Anticuerpos ampliamente neutralizantes/uso terapéutico , Anticuerpos Monoclonales
6.
Sci Rep ; 14(1): 18188, 2024 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-39107366

RESUMEN

Fecal Microbiota Transplant (FMT) has shown some success in treating inflammatory bowel diseases (IBD). There is emerging evidence that host engraftment of donor taxa is a tenet of successful FMT. We undertook a double-blind, randomized, placebo-controlled pilot study to characterize the response to FMT in children and young adults with mild to moderate active Crohn's disease (CD) and ulcerative colitis (UC). Subjects with CD or UC were randomized to receive antibiotics and weekly FMT or placebo in addition to baseline medications. We enrolled 15 subjects aged 14-29 years. Four subjects had CD, and 11 had UC. Subjects exhibited a wide range of microbial diversity and donor engraftment. Specifically, engraftment ranged from 26 to 90% at week 2 and 3-92% at 2 months. Consistent with the current literature, increases over time of both alpha diversity (p < 0.05) and donor engraftment (p < 0.05) correlated with improved clinical response. We discovered that the post-antibiotic but pre-FMT time point was rich in microbial correlates of eventual engraftment. Greater residual alpha diversity after antibiotic treatment was positively correlated with engraftment and subsequent clinical response. Interestingly, a transient rise in the relative abundance of Lactobacillus was also positively correlated with engraftment, a finding that we recapitulated with our analysis of another FMT trial.


Asunto(s)
Trasplante de Microbiota Fecal , Lactobacillus , Humanos , Trasplante de Microbiota Fecal/métodos , Adulto , Adolescente , Femenino , Masculino , Adulto Joven , Método Doble Ciego , Enfermedades Inflamatorias del Intestino/terapia , Enfermedades Inflamatorias del Intestino/microbiología , Microbioma Gastrointestinal , Proyectos Piloto , Heces/microbiología , Resultado del Tratamiento , Enfermedad de Crohn/terapia , Enfermedad de Crohn/microbiología , Antibacterianos/uso terapéutico , Antibacterianos/farmacología , Colitis Ulcerosa/terapia , Colitis Ulcerosa/microbiología
7.
Front Cell Infect Microbiol ; 14: 1430586, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39170985

RESUMEN

Background: Psoriasis is one of the most common autoimmune skin diseases. Increasing evidence shows that alterations in the diversity and function of microbiota can participate in the pathogenesis of psoriasis through various pathways and mechanisms. Objective: To review the connection between microbial changes and psoriasis, how microbial-targeted therapy can be used to treat psoriasis, as well as the potential of prebiotics, probiotics, synbiotics, fecal microbiota transplantation, diet, and Traditional Chinese Medicine as supplementary and adjunctive therapies. Methods: Literature related to the relationship between psoriasis and gut microbiota was searched in PubMed and CNKI. Results: Adjunct therapies such as dietary interventions, traditional Chinese medicine, and probiotics can enhance gut microbiota abundance and diversity in patients with psoriasis. These therapies stimulate immune mediators including IL-23, IL-17, IL-22, and modulate gamma interferon (IFN-γ) along with the NF-kB pathway, thereby suppressing the release of pro-inflammatory cytokines and ameliorating systemic inflammatory conditions. Conclusion: This article discusses the direction of future research and clinical treatment of psoriasis from the perspective of intestinal microbiota and the mechanism of traditional Chinese medicine, so as to provide clinicians with more comprehensive diagnosis and treatment options and bring greater hope to patients with psoriasis.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Medicina Tradicional China , Probióticos , Psoriasis , Psoriasis/terapia , Psoriasis/microbiología , Psoriasis/tratamiento farmacológico , Humanos , Probióticos/uso terapéutico , Prebióticos , Citocinas/metabolismo , Interleucina-17/metabolismo
8.
Gut Microbes ; 16(1): 2392009, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39161102

RESUMEN

Here, we explored the vast potential of microbiome-based interventions in preventing and managing non-communicable diseases including obesity, diabetes, allergies, celiac disease, inflammatory bowel diseases, malnutrition, and cardiovascular diseases across different life stages. We discuss the intricate relationship between microbiome and non-communicable diseases, emphasizing on the "window of opportunity" for microbe-host interactions during the first years after birth. Specific biotics and also live biotherapeutics including fecal microbiota transplantation emerge as pivotal tools for precision medicine, acknowledging the "one size doesn't' fit all" aspect. Challenges in implementation underscore the need for advanced technologies, scientific transparency, and public engagement. Future perspectives advocate for understanding maternal-neonatal microbiome, exploring the maternal exposome and delving into human milk's role in the establishment and restoration of the infant microbiome and its influence over health and disease. An integrated scientific approach, employing multi-omics and accounting for inter-individual variance in microbiome composition and function appears central to unleash the full potential of early-life microbiome interventions in revolutionizing healthcare.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Embarazo , Femenino , Recién Nacido , Leche Humana/microbiología , Trasplante de Microbiota Fecal , Lactante , Interacciones Microbiota-Huesped
9.
Gut Microbes ; 16(1): 2390136, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39163273

RESUMEN

Abdominal obesity-related metabolic syndrome (MetS) has emerged as a significant global public health issue that affects human health. Flavonoids, such as quercetin, have been reported to exert obvious anti-obesity and lipid-lowering effects in both humans and animal models. However, the precise underlying mechanism remains elusive. In this study, we investigated the potential roles of gut microbiota-bile acids (BAs) interactions in quercetin-induced anti-obesity effects and metabolic benefits. Oral administration of quercetin significantly enhanced energy metabolism through activating thermogenesis of brown adipose tissues (BAT) and browning of white adipose tissues (WAT), thus mitigating metabolic dysfunctions in an abdominal obesity-related MetS mouse model. Further mechanistic studies demonstrated that quercetin treatment substantially promoted the generation of non-12α-hydroxylated BAs (non-12OH BAs), particularly ursodeoxycholic acid (UDCA) and lithocholic acid (LCA), in serum via regulating the overall structure of gut microbiota and enriching Lactobacillus. High level of non-12OH BAs bind to Takeda G protein-coupled receptor 5 (TGR5) on adipocytes to stimulate thermogenesis. Remarkably, fecal microbiota transplantation (FMT) from quercetin-treated mice replicated the effects of quercetin on non-12OH BAs generation and energy expenditure, which suggested gut microbiota reshape and concomitant BAs regulation were responsible for the benefits on energy metabolism of quercetin in the MetS mouse model. Our findings not only highlighted the critical role of gut microbiota-BAs crosstalk in mediating quercetin-induced energy expenditure, but also enriched the pharmacological mechanisms of quercetin in ameliorating MetS-related diseases.


Asunto(s)
Tejido Adiposo Pardo , Ácidos y Sales Biliares , Metabolismo Energético , Microbioma Gastrointestinal , Síndrome Metabólico , Ratones Endogámicos C57BL , Quercetina , Termogénesis , Quercetina/farmacología , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Ratones , Metabolismo Energético/efectos de los fármacos , Síndrome Metabólico/metabolismo , Síndrome Metabólico/microbiología , Síndrome Metabólico/tratamiento farmacológico , Masculino , Ácidos y Sales Biliares/metabolismo , Termogénesis/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/efectos de los fármacos , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Modelos Animales de Enfermedad , Trasplante de Microbiota Fecal
10.
Gut Microbes ; 16(1): 2388295, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39163526

RESUMEN

Fecal Microbiota Transplantation (FMT) has emerged as a potential modality for mitigating microbiome-associated diseases. Despite this potential, the precise causal pathways by which specific gut microbiota strains induce remission remain inadequately elucidated. In this study, we aimed to discern the impact of engraftment of donor-infused strains on alterations in plasma metabolites, subsequently contributing to the amelioration of clinical parameters involved in subjects with metabolic syndrome (MetSyn) receiving an FMT. We observed that a higher fraction of donor strains engrafted in the recipient is correlated to a reduction in diastolic blood pressure and found specific strain associations through canonical correlation analysis. Integrating the metabolomics profile shows that engraftment of Collinsella aerofaciens and Fusocatenibacter saccharovorans was related to a reduction in 2-oxoarginine in plasma, which was subsequently correlated to a reduction in diastolic blood pressure. In conclusion, we applied a novel framework to elucidate on the complex and heterogenous FMT intervention, establishing a connection between engrafted microbiota and clinical outcome parameters. Our findings underscore the potential therapeutic efficacy of FMT in ameliorating MetSyn, demonstrating a potential contribution of microbial strain engraftment to the improvement of MetSyn via modulation of circulating metabolites.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Síndrome Metabólico , Humanos , Masculino , Persona de Mediana Edad , Síndrome Metabólico/terapia , Síndrome Metabólico/microbiología , Femenino , Heces/microbiología , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Bacterias/genética , Adulto , Presión Sanguínea , Resultado del Tratamiento
11.
Cell Rep Med ; 5(8): 101678, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39096912

RESUMEN

Chemotherapy-induced premature ovarian insufficiency (CIPOI) triggers gonadotoxicity in women undergoing cancer treatment, leading to loss of ovarian reserves and subfertility, with no effective therapies available. In our study, fecal microbiota transplantation in a cisplatin-induced POI mouse model reveals that a dysbiotic gut microbiome negatively impacts ovarian health in CIPOI. Multi-omics analyses show a significant decrease in Limosilactobacillus reuteri and its catabolite, ß-resorcylic acid , in the CIPOI group in comparison to healthy controls. Supplementation with L. reuteri or ß-RA mitigates cisplatin-induced hormonal disruptions, morphological damages, and reductions in follicular reserve. Most importantly, ß-RA pre-treatment effectively preserves oocyte function, embryonic development, and fetus health, thereby protecting against chemotherapy-induced subfertility. Our results provide evidence that ß-RA suppresses the nuclear accumulation of sex-determining region Y-box 7, which in turn reduces Bcl-2-associated X activation and inhibits granulosa cell apoptosis. These findings highlight the therapeutic potential of targeting the gut-ovary axis for fertility preservation in CIPOI.


Asunto(s)
Cisplatino , Limosilactobacillus reuteri , Ovario , Insuficiencia Ovárica Primaria , Femenino , Animales , Cisplatino/efectos adversos , Cisplatino/toxicidad , Ratones , Insuficiencia Ovárica Primaria/inducido químicamente , Insuficiencia Ovárica Primaria/patología , Ovario/efectos de los fármacos , Ovario/patología , Ovario/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Trasplante de Microbiota Fecal , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Ratones Endogámicos C57BL , Antineoplásicos/toxicidad , Antineoplásicos/efectos adversos , Células de la Granulosa/efectos de los fármacos , Células de la Granulosa/metabolismo , Modelos Animales de Enfermedad , Infertilidad
12.
Commun Biol ; 7(1): 1006, 2024 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-39152200

RESUMEN

Antibiotic-induced dysbiosis in the fish gut causes significant adverse effects. We use fecal microbiota transplantation (FMT) to accelerate the restoration of florfenicol-perturbed intestinal microbiota in koi carp, identifying key bacterial populations and metabolites involved in the recovery process through microbiome and metabolome analyses. We demonstrate that florfenicol disrupts intestinal microbiota, reducing beneficial genera such as Lactobacillus, Bifidobacterium, Bacteroides, Romboutsia, and Faecalibacterium, and causing mucosal injuries. Key metabolites, including aromatic amino acids and glutathione-related compounds, are diminished. We show that FMT effectively restores microbial populations, repairs intestinal damage, and normalizes critical metabolites, while natural recovery is less effective. Spearman correlation analyses reveal strong associations between the identified bacterial genera and the levels of aromatic amino acids and glutathione-related metabolites. This study underscores the potential of FMT to counteract antibiotic-induced dysbiosis and maintain fish intestinal health. The restored microbiota and normalized metabolites provide a basis for developing personalized probiotic therapies for fish.


Asunto(s)
Antibacterianos , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Tianfenicol , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Tianfenicol/análogos & derivados , Tianfenicol/farmacología , Disbiosis/terapia , Disbiosis/microbiología , Antibacterianos/farmacología , Antibacterianos/efectos adversos , Carpas/microbiología , Bacterias/metabolismo , Bacterias/efectos de los fármacos
13.
Microbiome ; 12(1): 147, 2024 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-39113097

RESUMEN

BACKGROUND: Understanding the cause vs consequence relationship of gut inflammation and microbial dysbiosis in inflammatory bowel diseases (IBD) requires a reproducible mouse model of human-microbiota-driven experimental colitis. RESULTS: Our study demonstrated that human fecal microbiota transplant (FMT) transfer efficiency is an underappreciated source of experimental variability in human microbiota-associated (HMA) mice. Pooled human IBD patient fecal microbiota engrafted germ-free (GF) mice with low amplicon sequence variant (ASV)-level transfer efficiency, resulting in high recipient-to-recipient variation of microbiota composition and colitis severity in HMA Il-10-/- mice. In contrast, mouse-to-mouse transfer of mouse-adapted human IBD patient microbiota transferred with high efficiency and low compositional variability resulting in highly consistent and reproducible colitis phenotypes in recipient Il-10-/- mice. Engraftment of human-to-mouse FMT stochastically varied with individual transplantation events more than mouse-adapted FMT. Human-to-mouse FMT caused a population bottleneck with reassembly of microbiota composition that was host inflammatory environment specific. Mouse-adaptation in the inflamed Il-10-/- host reassembled a more aggressive microbiota that induced more severe colitis in serial transplant to Il-10-/- mice than the distinct microbiota reassembled in non-inflamed WT hosts. CONCLUSIONS: Our findings support a model of IBD pathogenesis in which host inflammation promotes aggressive resident bacteria, which further drives a feed-forward process of dysbiosis exacerbated by gut inflammation. This model implies that effective management of IBD requires treating both the dysregulated host immune response and aggressive inflammation-driven microbiota. We propose that our mouse-adapted human microbiota model is an optimized, reproducible, and rigorous system to study human microbiome-driven disease phenotypes, which may be generalized to mouse models of other human microbiota-modulated diseases, including metabolic syndrome/obesity, diabetes, autoimmune diseases, and cancer. Video Abstract.


Asunto(s)
Modelos Animales de Enfermedad , Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Interleucina-10 , Animales , Humanos , Ratones , Enfermedades Inflamatorias del Intestino/microbiología , Disbiosis/microbiología , Interleucina-10/genética , Colitis/microbiología , Heces/microbiología , Colon/microbiología , Ratones Noqueados , Ratones Endogámicos C57BL , Femenino , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Inflamación , Masculino
14.
Gut Microbes ; 16(1): 2390176, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39205654

RESUMEN

Gut microbiota dysbiosis is involved in cholestatic liver diseases. However, the mechanisms remain to be elucidated. The purpose of this study was to examine the effects and mechanisms of Lactobacillus acidophilus (L. acidophilus) on cholestatic liver injury in both animals and humans. Bile duct ligation (BDL) was performed to mimic cholestatic liver injury in mice and serum liver function was tested. Gut microbiota were analyzed by 16S rRNA sequencing. Fecal bacteria transplantation (FMT) was used to evaluate the role of gut microbiota in cholestasis. Bile acids (BAs) profiles were analyzed by targeted metabolomics. Effects of L. acidophilus in cholestatic patients were evaluated by a randomized controlled clinical trial (NO: ChiCTR2200063330). BDL induced different severity of liver injury, which was associated with gut microbiota. 16S rRNA sequencing of feces confirmed the gut flora differences between groups, of which L. acidophilus was the most distinguished genus. Administration of L. acidophilus after BDL significantly attenuated hepatic injury in mice, decreased liver total BAs and increased fecal total BAs. Furthermore, after L. acidophilus treatment, inhibition of hepatic Cholesterol 7α-hydroxylase (CYP7α1), restored ileum Fibroblast growth factor 15 (FGF15) and Small heterodimer partner (SHP) accounted for BAs synthesis decrease, whereas enhanced BAs excretion was attributed to the increase of unconjugated BAs by enriched bile salt hydrolase (BSH) enzymes in feces. Similarly, in cholestasis patients, supplementation of L. acidophilus promoted the recovery of liver function and negatively correlated with liver function indicators, possibly in relationship with the changes in BAs profiles and gut microbiota composition. L. acidophilus treatment ameliorates cholestatic liver injury through inhibited hepatic BAs synthesis and enhances fecal BAs excretion.


Asunto(s)
Ácidos y Sales Biliares , Colestasis , Microbioma Gastrointestinal , Lactobacillus acidophilus , Hígado , Ratones Endogámicos C57BL , Probióticos , Ácidos y Sales Biliares/metabolismo , Animales , Colestasis/metabolismo , Colestasis/microbiología , Ratones , Humanos , Masculino , Probióticos/farmacología , Probióticos/administración & dosificación , Hígado/metabolismo , Heces/microbiología , Colesterol 7-alfa-Hidroxilasa/metabolismo , Colesterol 7-alfa-Hidroxilasa/genética , Femenino , Factores de Crecimiento de Fibroblastos/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Trasplante de Microbiota Fecal , Disbiosis/microbiología , Disbiosis/terapia , ARN Ribosómico 16S/genética , Persona de Mediana Edad , Adulto , Modelos Animales de Enfermedad , Íleon/microbiología , Íleon/metabolismo
15.
Iran J Med Sci ; 49(8): 472-486, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39205822

RESUMEN

Pouchitis, as the most common complication after ileal pouch-anal anastomosis (IPAA), has an incidence from 7% to 46%. Pouchitis treatment still represents one of the biggest gaps of knowledge in the treatment of diseases. This review has focused on achievements and challenges in the treatment of pouchitis. A combined assessment of symptoms, endoscopic findings, histologic results, quick biomarkers, and fecal calprotectin test were determined to be valuable diagnostic criteria. Conventional therapy was described as a modification of bacterial flora, mainly with antibiotics and more recently with probiotics such as bifidobacteria, lactobacilli, and streptococci. Other therapeutic approaches such as anti-tumor necrosis factor, infliximab, adalimumab, vedolizumab, ustekinumab, tacrolimus, tofacitinib, thiopurines, corticosteroids, prolyl hydroxylase-containing enzymes, povidone-iodine, dextrose spray, fecal microbiota transplantation, herbal medicines, and leukocyte apheresis have been discussed. Changes in dietary components, and administration of complementary and alternative medicine, probiotics, and fecal transplantation in addition to conventional therapies were also shown to affect the outcome of disease. Due to the potential significant impairment in quality of life caused by pouchitis, it is essential to address the gaps in knowledge for both patients and physicians in its treatment. Therefore, well-designed and adequately powered studies should assess the optimal treatment for pouchitis.


Asunto(s)
Reservoritis , Humanos , Reservoritis/terapia , Reservoritis/etiología , Reservoritis/tratamiento farmacológico , Trasplante de Microbiota Fecal/métodos , Probióticos/uso terapéutico , Antibacterianos/uso terapéutico
17.
Cell Host Microbe ; 32(8): 1219-1224, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-39146793

RESUMEN

Fecal microbial transplantation (FMT) for inflammatory diseases or refractory immune checkpoint inhibitor therapy is less effective than for preventing recurrent Clostridioides difficile infection. This commentary outlines strategies to use biomarkers of successful FMT to guide newer approaches to restore microbial homeostasis in individuals with dysbiosis-mediated inflammation.


Asunto(s)
Disbiosis , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Medicina de Precisión , Humanos , Trasplante de Microbiota Fecal/métodos , Disbiosis/terapia , Medicina de Precisión/métodos , Infecciones por Clostridium/terapia , Infecciones por Clostridium/microbiología , Inflamación , Clostridioides difficile , Biomarcadores
18.
CNS Neurosci Ther ; 30(8): e70003, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39161161

RESUMEN

AIMS: We evaluated the potential of Parkinson's disease (PD) fecal microbiota transplantation to initiate or exacerbate PD pathologies and investigated the underlying mechanisms. METHODS: We transplanted the fecal microbiota from PD patients into mice by oral gavage and assessed the motor and intestinal functions, as well as the inflammatory and pathological changes in the colon and brain. Furthermore, 16S rRNA gene sequencing combined with metabolomics analysis was conducted to assess the impacts of fecal delivery on the fecal microbiota and metabolism in recipient mice. RESULTS: The fecal microbiota from PD patients increased intestinal inflammation, deteriorated intestinal barrier function, intensified microglia and astrocyte activation, abnormal deposition of α-Synuclein, and dopaminergic neuronal loss in the brains of A53T mice. A mechanistic study revealed that the fecal microbiota of PD patients stimulated the TLR4/NF-κB/NLRP3 pathway in both the brain and colon. Additionally, multiomics analysis found that transplantation of fecal microbiota from PD patients not only altered the composition of the gut microbiota but also influenced the fecal metabolic profile of the recipient mice. CONCLUSION: The fecal microbiota from PD patients intensifies inflammation and neurodegeneration in A53T mice. Our findings demonstrate that imbalance and dysfunction in the gut microbiome play significant roles in the development and advancement of PD.


Asunto(s)
Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/microbiología , Enfermedad de Parkinson/metabolismo , Humanos , Microbioma Gastrointestinal/fisiología , Masculino , Inflamación/metabolismo , Inflamación/microbiología , Heces/microbiología , Ratones Transgénicos , Ratones Endogámicos C57BL , Femenino , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Encéfalo/patología
19.
Gut Microbes ; 16(1): 2392876, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39172643

RESUMEN

Fecal filtrate transfer (FFT) is emerging as a safer alternative to traditional fecal microbiota transplantation (FMT) - particularly in the context of necrotizing enterocolitis (NEC), a severe gastrointestinal condition affecting preterm infants. Using a preterm piglet model, FFT has demonstrated superiority over FMT in safety and NEC prevention. Since FFT is virtually devoid of bacteria, prokaryotic viruses (bacteriophages) are assumed to mediate the beneficial effects. However, this assumption remains unproven. To address this gap, we separated virus-like particles (30 kDa to 0.45 µm) of donor feces from the residual postbiotic fluid. We then compared clinical and gut microbiota responses to these fractions with the parent FFT solution after transferring them to NEC-susceptible preterm piglets. Virome transfer was equally effective as FFT in reducing the severity of NEC-like pathology. The bacterial compositional data corroborated clinical findings as virome transfer reduced the relative abundance of several NEC-associated pathogens e.g. Klebsiella pneumoniae and Clostridium perfringens. Virome transfer diversified gut viral communities with concomitant constraining effects on the bacterial composition. Unexpectedly, virome transfer, but not residual postbiotic fluid, led to earlier diarrhea. While diarrhea may be a minor concern in human infants, future work should identify ways of eliminating this side effect without losing treatment efficacy.


Asunto(s)
Enterocolitis Necrotizante , Trasplante de Microbiota Fecal , Heces , Microbioma Gastrointestinal , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/terapia , Animales , Heces/virología , Heces/microbiología , Trasplante de Microbiota Fecal/métodos , Porcinos , Humanos , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Animales Recién Nacidos , Modelos Animales de Enfermedad , Viroma , Clostridium perfringens , Bacteriófagos/genética , Bacteriófagos/fisiología , Diarrea/terapia , Diarrea/virología , Diarrea/prevención & control , Diarrea/microbiología
20.
Gut Microbes ; 16(1): 2391535, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39182245

RESUMEN

BACKGROUND: Chronic infection with the neurotropic parasite Toxoplasma gondii (T. gondii) can cause anxiety and gut microbiota dysbiosis in hosts. However, the potential role of gut microbiota in anxiety induced by the parasite remains unclear. METHODS: C57BL/6J mice were infected with 10 cysts of T. gondii. Antibiotic depletion of gut microbiota and fecal microbiota transplantation experiments were utilized to investigate the causal relationship between gut microbiota and anxiety. Anxiety-like behaviors were examined by the elevated plus maze test and the open field test; blood, feces, colon and amygdala were collected to evaluate the profiles of serum endotoxin (Lipopolysaccharide, LPS) and serotonin (5-hydroxytryptamine, 5-HT), gut microbiota composition, metabolomics, global transcriptome and neuroinflammation in the amygdala. Furthermore, the effects of Diethyl butylmalonate (DBM, an inhibitor of mitochondrial succinate transporter, which causes the accumulation of endogenous succinate) on the disorders of the gut-brain axis were evaluated. RESULTS: Here, we found that T. gondii chronic infection induced anxiety-like behaviors and disturbed the composition of the gut microbiota in mice. In the amygdala, T. gondii infection triggered the microglial activation and neuroinflammation. In the colon, T. gondii infection caused the intestinal dyshomeostasis including elevated colonic inflammation, enhanced bacterial endotoxin translocation to blood and compromised intestinal barrier. In the serum, T. gondii infection increased the LPS levels and decreased the 5-HT levels. Interestingly, antibiotics ablation of gut microbiota alleviated the anxiety-like behaviors induced by T. gondii infection. More importantly, transplantation of the fecal microbiota from T. gondii-infected mice resulted in anxiety and the transcriptomic alteration in the amygdala of the antibiotic-pretreated mice. Notably, the decreased abundance of succinate-producing bacteria and the decreased production of succinate were observed in the feces of the T. gondii-infected mice. Moreover, DBM administration ameliorated the anxiety and gut barrier impairment induced by T. gondii infection. CONCLUSIONS: The present study uncovers a novel role of gut microbiota in mediating the anxiety-like behaviors induced by chronic T. gondii infection. Moreover, we show that DBM supplementation has a beneficial effect on anxiety. Overall, these findings provide new insights into the treatment of T. gondii-related mental disorders.


Asunto(s)
Ansiedad , Microbioma Gastrointestinal , Ratones Endogámicos C57BL , Toxoplasma , Animales , Ratones , Ansiedad/microbiología , Toxoplasma/fisiología , Masculino , Trasplante de Microbiota Fecal , Disbiosis/microbiología , Amígdala del Cerebelo/metabolismo , Conducta Animal , Toxoplasmosis/fisiopatología , Toxoplasmosis/psicología , Toxoplasmosis/parasitología , Toxoplasmosis/microbiología , Enfermedad Crónica , Eje Cerebro-Intestino/fisiología , Modelos Animales de Enfermedad , Colon/microbiología , Colon/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA