Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.448
Filtrar
1.
Methods Mol Biol ; 2855: 195-207, 2025.
Artículo en Inglés | MEDLINE | ID: mdl-39354310

RESUMEN

Short- and medium-chain fatty acids (SMCFA) are monocarboxylic acids with a carbon chain length of 1-12 carbon atoms. They are mainly produced in humans by the gut microbiota, play crucial metabolic roles, are vital for intestinal health, and have multifaceted impact on immune and neurological functions. Accurate detection and quantification of SMCFA in different human biofluids is achieved using 3-nitro phenylhydrazine (3-NPH) derivatization of the free fatty acids followed by reverse phase liquid chromatography (RPLC) separation and detection by tandem mass spectrometry (MS/MS). Here, we describe the simultaneous measurement of 14 SMCFA and lactate in detail. All 3-NPH-SMCFA-hydrazones are separated in less than 5 min with an 8-min total run time (injection-to-injection). Linear dynamic range over 0.1-500 µM is achieved for most SCFAs, while it is 0.05-100 µM for MCFAs. Validation of the procedure depicts good linearity (R2 > 0.98) and repeatability (CV ≤ 20%). The lower limit of detection (LLOD) is 10-30 nM. The lower limit of quantification (LLOQ) is 50-100 nM for most analytes, while it is 0.5 µM for acetate. In conclusion, the method offers several benefits compared to alternative methods regarding throughput, selectivity, sensitivity, and robustness.


Asunto(s)
Cromatografía de Fase Inversa , Espectrometría de Masas en Tándem , Espectrometría de Masas en Tándem/métodos , Humanos , Cromatografía de Fase Inversa/métodos , Ácidos Grasos Volátiles/análisis , Ensayos Analíticos de Alto Rendimiento/métodos , Límite de Detección , Ácidos Grasos/análisis , Ácidos Grasos/química , Reproducibilidad de los Resultados
2.
Front Microbiol ; 15: 1473441, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39351297

RESUMEN

Background: We aim to establish a gestational diabetes mellitus (GDM) mouse model with mice fed with a high-fat diet (HFD) in comparison with pregnant mice with normal blood glucose levels to investigate the role of intestinal microbiota in the development of HFD-induced GDM. Methods: We divided healthy 6-week-old female C57BL mice into an HFD-induced GDM group and a normal diet group. Their bacterial flora and metabolites in intestinal fecal exosomes were co-analyzed using 16 s multi-region sequencing and compared. Findings: Alpha (α) diversity was lower within the model group compared to the control group. Beta (ß) diversity was significantly different between the two groups. The relative abundances of Lactobacillus, Actinomyces, Rothia, and Bacteroidetes were significantly different between the two groups. Fermentation and nitrate consumption were significantly higher in the GDM group. Multiple bacteria were associated with glycerophosphocholine, S-methyl-5'-thioadenosine, quinolinate, galactinol, deoxyadenosine, DL-arginine, and 2-oxoadenic acid. Interpretation: Imbalances in the production of Lactobacillus, Bacteroidetes, Actinomyces, and Rothia and their related metabolites may lead to metabolic disturbances in GDM. These indicators may be used to assess changes affecting the intestinal microbiota during pregnancy and thus help modulate diet and alter blood glucose.

3.
Trends Mol Med ; 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-39353744

RESUMEN

Although the field of psychiatry has made gains in biomarker discovery, our ability to change long-term outcomes remains inadequate. Matching individuals to the best treatment for them is a persistent clinical challenge. Moreover, the development of novel treatments has been hampered in part due to a limited understanding of the biological mechanisms underlying individual differences that contribute to clinical heterogeneity. The gut microbiome has become an area of intensive research in conditions ranging from metabolic disorders to cancer. Innovation in these spaces has led to translational breakthroughs, offering novel microbiome-informed approaches that may improve patient outcomes. In this review we examine how translational microbiome research is poised to advance biomarker discovery in mental health, with a focus on depression.

4.
Front Cell Infect Microbiol ; 14: 1436477, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39355267

RESUMEN

Aims: Hyperglycemia is one of the adverse effects of tacrolimus (TAC), but the underlying mechanism is not fully identified. We used multi-omics analysis to evaluate the changes in the gut microbiota and metabolic profile of rats with TAC-induced diabetes. Methods: To establish a diabetic animal model, Sprague Dawley rats were divided randomly into two groups. Those in the TAC group received intraperitoneal injections of TAC (3 mg/kg) for 8 weeks, and those in the CON group served as the control. 16S rRNA sequencing was used to analyze fecal microbiota. The metabolites of the two groups were detected and analyzed by nontargeted and targeted metabolomics, including amino acids (AAs), bile acids (BAs), and short-chain fatty acids (SCFAs). Results: The rats treated with TAC exhibited hyperglycemia as well as changes in the gut microbiota and metabolites. Specifically, their gut microbiota had significantly higher abundances of Escherichia-Shigella, Enterococcus, and Allobaculum, and significantly lower abundances of Ruminococcus, Akkermansia, and Roseburia. In addition, they had significantly reduced serum levels of AAs including asparagine, aspartic acid, glutamic acid, and methionine. With respect to BAs, they had significantly higher serum levels of taurocholic acid (TCA), and glycochenodeoxycholic acid (GCDCA), but significantly lower levels of taurodeoxycholic acid (TDCA) and tauroursodeoxycholic acid (TUDCA). There were no differences in the levels of SCFAs between the two groups. Correlations existed among glucose metabolism indexes (fasting blood glucose and fasting insulin), gut microbiota (Ruminococcus and Akkermansia), and metabolites (glutamic acid, hydroxyproline, GCDCA, TDCA, and TUDCA). Conclusions: Both AAs and BAs may play crucial roles as signaling molecules in the regulation of TAC-induced diabetes.


Asunto(s)
Aminoácidos , Heces , Microbioma Gastrointestinal , Metabolómica , ARN Ribosómico 16S , Ratas Sprague-Dawley , Tacrolimus , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Tacrolimus/farmacología , Ratas , Masculino , Heces/microbiología , ARN Ribosómico 16S/genética , Aminoácidos/metabolismo , Aminoácidos/sangre , Diabetes Mellitus Experimental/metabolismo , Ácidos y Sales Biliares/metabolismo , Ácidos Grasos Volátiles/metabolismo , Metaboloma/efectos de los fármacos , Modelos Animales de Enfermedad , Hiperglucemia/metabolismo , Bacterias/clasificación , Bacterias/efectos de los fármacos , Bacterias/metabolismo , Bacterias/genética , Glucemia/metabolismo , Inmunosupresores
5.
Bioresour Technol ; : 131580, 2024 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-39384049

RESUMEN

Challenges in obtaining stable nitrite have impeded the use of anammox in municipal wastewater treatment. This study explored the feasibility of using sludge fermentation products as carbon source and selective nitrification inhibitor to supply nitrite via partial nitrification (PN) and partial denitrification (PD). PD was initiated within 15 days, achieving nitrite transformation rate of over 90 % with a C/N ratio of 3 and a reaction time of 0.75 h. The dominant genus, Romboutsia, increased in relative abundance from 4.1 to 35 %. Organic acids in sludge fermentation products, like acetate (200 mg/L) and propionate (400 mg/L), selectively suppressed nitrite-oxidizing bacteria (NOB) more than ammonia-oxidizing bacteria (AOB), leading to PN. Combining anaerobic exposure with sludge fermentation products addition achieved PN with over 80.0 % nitrite accumulation. AOB increased tenfold in the long term, significantly outpacing NOB growth. This strategy simplifies difficulty of anammox application and shows broad application potential in municipal wastewater treatment.

6.
Front Cell Infect Microbiol ; 14: 1434687, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39372498

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic lung disease that affects premature infants and leads to long-term pulmonary complications. The pathogenesis of BPD has not been fully elucidated yet. In recent years, the microbiome and its metabolites, especially short-chain fatty acids (SCFAs), in the gut and lungs have been demonstrated to be involved in the development and progression of the disease. This review aims to summarize the current knowledge on the potential involvement of the microbiome and SCFAs, especially the latter, in the development and progression of BPD. First, we introduce the gut-lung axis, the production and functions of SCFAs, and the role of SCFAs in lung health and diseases. We then discuss the evidence supporting the involvement of the microbiome and SCFAs in BPD. Finally, we elaborate on the potential mechanisms of the microbiome and SCFAs in BPD, including immune modulation, epigenetic regulation, enhancement of barrier function, and modulation of surfactant production and the gut microbiome. This review could advance our understanding of the microbiome and SCFAs in the pathogenesis of BPD, which also helps identify new therapeutic targets and facilitate new drug development.


Asunto(s)
Displasia Broncopulmonar , Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Pulmón , Displasia Broncopulmonar/metabolismo , Displasia Broncopulmonar/microbiología , Humanos , Ácidos Grasos Volátiles/metabolismo , Microbioma Gastrointestinal/fisiología , Pulmón/microbiología , Pulmón/metabolismo , Pulmón/patología , Microbiota , Recien Nacido Prematuro , Recién Nacido , Animales , Epigénesis Genética
7.
Heliyon ; 10(19): e38302, 2024 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-39386817

RESUMEN

Background: The gut microbiota thrives in a complex ecological environment and its dynamic balance is closely related to host health. Recent studies have shown that the occurrence of various diseases including prostate inflammation is related to the dysregulation of the gut microbiome. Objective: This review focus on the mechanisms by which the gut microbiota induces prostate inflammation and benign prostatic hyperplasia and its therapeutic implications. Materials and methods: Publications related to gut microbiota, prostate inflammation, and benign prostatic hyperplasia (BPH) until April 2023 were systematically reviewed. The research questions were formulated using the Problem, Intervention, Comparison/Control, and Outcome (PICO) frameworks. Results: Fifteen articles covering the relationship between the gut microbiota and prostate inflammation/BPH, the mechanisms by which the gut microbiota influences prostate inflammation and BPH, and potential therapeutic approaches targeting the gut microbiota for these conditions were included. Conclusion: Short-chain fatty acids (SCFAs), which are metabolites of the intestinal microbiota, protect the integrity of the intestinal barrier, regulate immunity, and inhibit inflammation. However, dysregulation of the gut microbiota significantly reduces the SCFA content in feces and impairs the integrity of the gut barrier, leading to the translocation of bacteria and bacterial components such as lipopolysaccharide, mediating the development of prostate inflammation through microbe-associated molecular patterns (MAMPs).

8.
Arch Toxicol ; 2024 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-39370473

RESUMEN

Despite offering significant conveniences, plastic materials contribute substantially in developing environmental hazards and pollutants. Plastic trash that has not been adequately managed may eventually break down into fragments caused by human or ecological factors. Arguably, the crucial element for determining the biological toxicities of plastics are micro/nano-forms of plastics (MPs/NPs), which infiltrate the mammalian tissue through different media and routes. Infiltration of MPs/NPs across the intestinal barrier leads to microbial architectural dysfunction, which further modulates the population of gastrointestinal microbes. Thereby, it triggers inflammatory mediators (e.g., IL-1α/ß, TNF-α, and IFN-γ) by activating specific receptors located in the gut barrier. Mounting evidence indicates that MPs/NPs disrupt host pathophysiological function through modification of junctional proteins and effector cells. Moreover, the alteration of microbial diversity by MPs/NPs causes the breakdown of the blood-brain barrier and translocation of metabolites (e.g., SCFAs, LPS) through the vagus nerve. Potent penetration affects the neuronal networks, neuronal protein accumulation, acceleration of oxidative stress, and alteration of neurofibrillary tangles, and hinders distinctive communicating pathways. Conclusively, alterations of these neurotoxic factors are possibly responsible for the associated neurodegenerative disorders due to the exposure of MPs/NPs. In this review, the hypothesis on MPs/NPs associated with gut microbial dysbiosis has been interlinked to the distinct neurological impairment through the gut-brain axis.

9.
Gut Microbes ; 16(1): 2409207, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39360560

RESUMEN

BACKGROUND: Despite achieving endoscopic remission, over 20% of inflammatory bowel disease (IBD) patients experience chronic abdominal pain. Visceral pain and the microbiome exhibit sex-dependent interactions, while visceral pain in IBD shows a sex bias. Our aim was to evaluate whether post-inflammatory microbial perturbations contribute to visceral hypersensitivity in a sex-dependent manner. METHODS: Males, cycling females, ovariectomized, and sham-operated females were given dextran sodium sulfate to induce colitis and allowed to recover. Germ-free recipients received sex-appropriate and cross-sex fecal microbial transplants (FMT) from post-inflammatory donor mice. Visceral sensitivity was assessed by recording visceromotor responses to colorectal distention. The composition of the microbiota was evaluated via 16S rRNA gene V4 amplicon sequencing, while the metabolome was assessed using targeted (short chain fatty acids - SCFA) and semi-targeted mass spectrometry. RESULTS: Post-inflammatory cycling females developed visceral hyperalgesia when compared to males. This effect was reversed by ovariectomy. Both post-inflammatory males and females exhibited increased SCFA-producing species, but only males had elevated fecal SCFA content. FMT from post-inflammatory females transferred visceral hyperalgesia to both males and females, while FMT from post-inflammatory males could only transfer visceral hyperalgesia to males. CONCLUSIONS: Female sex, hormonal status as well as the gut microbiota play a role in pain modulation. Our data highlight the importance of considering biological sex in the evaluation of visceral pain.


Asunto(s)
Colitis , Disbiosis , Microbioma Gastrointestinal , Dolor Visceral , Masculino , Femenino , Animales , Disbiosis/microbiología , Dolor Visceral/microbiología , Dolor Visceral/fisiopatología , Dolor Visceral/metabolismo , Colitis/microbiología , Ratones , Ratones Endogámicos C57BL , Trasplante de Microbiota Fecal , Factores Sexuales , Bacterias/clasificación , Bacterias/aislamiento & purificación , Bacterias/genética , Bacterias/metabolismo , ARN Ribosómico 16S/genética , Heces/microbiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/análisis , Dolor Crónico/microbiología , Dolor Crónico/fisiopatología , Inflamación/microbiología , Hiperalgesia/microbiología
10.
Int Immunopharmacol ; 143(Pt 1): 113271, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39368133

RESUMEN

BACKGROUND AND OBJECTIVE: Terminalia chebula is a classical medicine for the treatment of lingering dysentery, and both raw and processed T. chebula can alleviate ulcerative colitis (UC). The therapeutic efficacy of T. chebula is enhanced after processing, but the mechanism that processing improves this efficacy is still unknown. We investigated the medicinal effects of raw and processed T. chebula on dextran sulfate sodium (DSS)-induced UC model rats using intestinal flora and metabolomics analyses, in order to elucidate the mechanism by which processing enhances the therapeutic effect. METHODS: The major constituents of raw and processed T. chebula were detected by high-performance liquid chromatography (HPLC). UC model was replicated using the DSS method, and then UC rats were administered raw and processed T. chebula. The general physical signs, disease activity index (DAI) scores, colon histopathological morphology, and the expressions of inflammatory cytokines were used to evaluate the therapeutic effect of T. chebula. In addition, 16 s rRNA sequencing and gas chromatography-mass spectrometry (GC-MS) were used to characterize the intestinal flora and contents of short-chain fatty acids (SCFAs). Ultra-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF-MS) was utilized to identify the nontargeted fecal metabolites. RESULTS: Raw and processed T. chebula significantly improved the general physical signs and colon inflammatory symptoms and decreased DAI scores of UC rats. Both raw and processed T. chebula mitigated intestinal flora disorders in UC rats, increasing probiotic bacteria, including Lactobacillus and Romboutsia. However, the effect of processed T. chebula was more pronounced. Moreover, the levels of SCFAs of DSS-induced UC rats were restored after drug administration, and the processed T. chebula had a better regulatory effect than raw T. chebula. In the fecal nontargeted metabolomics analysis, differential metabolites such as lipids and amino acids were identified. The processed T. chebula can regulate purine metabolism and other pathways to improve UC, and the levels of the disordered metabolites gradually approached those of the control group. CONCLUSION: Raw and processed T. chebula had the capacity to mitigate DSS-induced UC by rebalancing the intestinal flora, restoring the contents of SCFAs, and regulating fecal metabolites, while processed T. chebula showed preferable effects.

11.
Front Endocrinol (Lausanne) ; 15: 1392418, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39363899

RESUMEN

Bone is a dynamic tissue that is constantly remodeled throughout adult life. Recently, it has been shown that bone turnover decreases shortly after food consumption. This process has been linked to the fermentation of non-digestible food ingredients such as inulin by gut microbes, which results in the production of the short-chain fatty acids (SCFAs) acetate, propionate and butyrate. SCFAs exert various metabolic functions, which in part can be explained by activation of G protein-coupled receptors (Gpr) 41 and 43. However, the potential relevance of a SCFA-Gpr41/43 signaling axis for bone metabolism has not been established. The aim of our study is to investigate the role of Gpr41/43 in bone metabolism and osteogenic differentiation of mesenchymal stem cells. For this purpose, we analyzed the skeletal phenotype of wild type controls (WT) and Gpr41/43 double knockout (Gpr41/43 dKO) mice fed either a chow or an inulin-enriched diet. In addition, we isolated bone marrow derived mesenchymal stem cells from WT and Gpr41/43 dKO mice and differentiated them into osteoblasts in the absence or presence of acetate. MicroCT scanning of femoral bones of Gpr41/43 dKO mice revealed a significant increase of trabecular bone volume and trabecular compared to WT controls. Treatment of WT bone marrow-derived osteoblasts with acetate resulted in decreased mineralization and substantial downregulation of bone formation markers such as Phex, Ptgs2 and Col1a1. Notably, this effect was strongly attenuated in differentiated osteoblasts lacking Gpr41/43. Inversely, acetate supplementation resulted in higher levels of adipocyte marker genes including Pparg, Lpl and Adipoq in bone marrow-derived cells from WT mice, an effect blunted in differentiated cells isolated from Gpr41/43 dKO mice. Overall, these data indicate that acetate regulates bone architecture via SCFA-Gpr41/43 signaling by modulating the osteogenic versus adipogenic differentiation of mesenchymal stem cells.


Asunto(s)
Adipogénesis , Diferenciación Celular , Células Madre Mesenquimatosas , Ratones Noqueados , Osteogénesis , Receptores Acoplados a Proteínas G , Animales , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Adipogénesis/fisiología , Osteogénesis/fisiología , Ácidos Grasos Volátiles/metabolismo , Ratones Endogámicos C57BL , Densidad Ósea , Masculino , Osteoblastos/metabolismo , Osteoblastos/citología , Células Cultivadas
12.
Front Nutr ; 11: 1465940, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39364150

RESUMEN

Background: The disorder of uric acid metabolism is closely associated with gut microbiota and short-chain fatty acids (SCFAs) dysregulation, but the biological mechanism is unclear, limiting the development of uric acid-lowering active polysaccharides. Konjac glucomannan (KGM) could attenuate metabolic disturbance of uric acid and modulate the gut microbiota. However, the relationship between uric acid metabolism and gut microbiota is still unknown. Methods: In this study, The fecal samples were provided by healthy volunteers and hyperuricemia (HUA) patients. Fecal samples from healthy volunteers was regarded as the NOR group. Similarly, 10% HUA fecal suspension was named as the HUA group. Then, fecal supernatant was inoculated into a growth basal medium containing glucose or KGM, and healthy fecal samples were designated as the NOR-GLU and NOR-KGM groups, while HUA fecal samples were designated as the HUA-GLU and HUA-KGM groups. All samples were cultured in an anaerobic bag system. After fermentation for 24 h, the samples were collected for further analysis of composition of intestinal microbiota, SCFAs concentration and XOD enzyme activity. Results: The results showed that KGM could be utilized and degraded by the gut microbiota from HUA subjects, and it could modulate the composition and structure of their HUA gut microbiota to more closely resemble that of a healthy group. In addition, KGM showed a superior modulated effect on HUA gut microbiota by increasing Megasphaera, Faecalibacterium, Lachnoclostridium, Lachnospiraceae, Anaerostipes, and Ruminococcus levels and decreasing Butyricicoccus, Eisenbergiella, and Enterococcus levels. Furthermore, the fermentation solution of KGM showed an inhibitory effect on xanthine oxidase (XOD) enzyme activity, which might be due to metabolites such as SCFAs. Conclusion: In conclusion, the effect of KGM on hyperuricemia subjects was investigated based on the gut microbiota in vitro. In the present study. It was found that KGM could be metabolized into SCFAs by HUA gut microbiota. Furthermore, KGM could modulate the structure of HUA gut microbiota. At the genus level, KGM could decrease the relative abundances of Butyricicoccus, Eisenbergiella, and Enterococcus, while Lachnoclostridium and Lachnospiraceae in HUA gut microbiota were significantly increased by the addition of KGM. The metabolites of gut microbiota, such as SCFAs, might be responsible for the inhibition of XOD activity. Thus, KGM exhibited a superior probiotic function on the HUA gut microbiota, which is expected as a promising candidate for remodeling the HUA gut microbiota.

13.
J Sci Food Agric ; 2024 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-39412364

RESUMEN

BACKGROUND: Postweaning intestinal damage in piglets is a challenging issue in the livestock industry. Short-chain fatty acids (SCFAs) are important metabolic products of the gut microbiota and are widely recognized for their role in maintaining normal colonic function and regulating the intestinal immune system. However, the effects of branched short-chain fatty acid (BSCFA) isobutyrate on intestinal health remain largely unknown. This study aims to explore the potential of isobutyrate for alleviating postweaning intestinal damage. RESULTS: This study indicates that isobutyrate can alleviate diarrhea in weaned piglets, enhance their growth performance, and optimize the gut microbiota. This is mainly achieved through increasing the relative abundance of probiotic bacteria such as Lactobacillus, Megasphaera, and Prevotellaceae_UCG-003, while concurrently reducing the relative abundance of potentially harmful bacteria such as Clostridium_sensu_stricto-1 and Escherichia-Shigella. It promotes the production of SCFAs, including acetate, isobutyrate, and butyrate. Furthermore, it activates G-protein-coupled receptors (GPR43/109A), inhibits the TLR4/MyD88 signaling pathway, strengthens the intestinal barrier function, and regulates the expression of related cytokines. CONCLUSION: In summary, exogenous isobutyrate can be considered a promising feed additive for improving the intestinal microbiota and regulating intestinal health in piglets. © 2024 Society of Chemical Industry.

14.
Environ Pollut ; 363(Pt 1): 125064, 2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39366448

RESUMEN

Diethyl ethylphosphonate (DEEP) as a novel organophosphorus flame retardant received increasing attention and its structure was discovered. But there are currently insufficient studies on how DEEP exposure affects the gut microbiome. In this study, the effects of DEEP on the structure and function of the human gut microbiota were examined using the SHIME system. Results from high-throughput sequencing of the 16S rRNA gene show that the high dose DEEP exposure reduced the Shannon and Simpson index in the transverse and descending colon. The Bacillota had the highest proportion while the proportion of Proteobacteria gradually decreased at the phylum level. The abundance of Escherichia, Prevotella, and Bilophila at the genus level increased with increasing doses of DEEP exposure. On the contrary, the abundance of Megasphaera, Klebsiella, and Phascolarctobacterium decreased. The short-chain fatty acids had a significant shift. With increasing doses of DEEP exposure, the concentration of acetic acid and propionic acid increased, while the concentration of butyric acid reached the highest at the medium dose of exposure. In addition, Bilophila, Psychrobacter, Escherichia, and Nostoe showed strong beneficial associations with acetic and propionic acids under DEEP exposure. Phocaeicola, Agathobacter, Klebsiella, Megasphaera, Phascolarctobacterium, and Bacteroides were negatively association with acetic and propionic acids. In a word, the study verified that exposure to different doses of DEEP can cause changes in the composition of the gut microbiome and metabolite SCFAs, which provides ideas for the investigation of other potential hazards of DEEP on human beings.

15.
Int J Biol Macromol ; 281(Pt 1): 136390, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39383910

RESUMEN

Polygalacturonic acid (PGA) restored the alpha-diversity of gut microbiota and promoted T cells infiltration in tumors. Here, we investigated whether oral administration of PGA could improve the anti-cancer effect of lipopolysaccharide-encapsulated PLGA-PEG-PLGA (LPS/PPP) in mice bearing CT26 tumors. Hydrogels with rapid thermogelling properties can achieve localized and controlled release of LPS, thus retaining the anti-cancer effect of LPS and avoiding a robust inflammatory storm. LPS/PPP promoted M1 macrophage polarization, TLR4 expression, and phagocytosis in tumors. The combination of PGA and LPS/PPP (PGA_LPS) notably repressed CT26 tumor growth and the inhibition rate reached 67.6 %. PGA_LPS triggered the recruitment of helper and cytotoxic T cells, IFN-γ level, decreased the proportion of immunosuppressive regulatory T cells. PGA_LPS also restored the beta-diversity of gut microbiota and increased short chain fatty acids abundance (butyric acid, 608.93 % vs. model group, P < 0.01). PGA_LPS followed by αPD-L1 resulted in obvious inhibition of both CT26 and 4T1 tumor growth, promoted cleaved-caspase 3 and Bax expression, T cell responses and the rescue of T cells exhaustion. These results confirmed that PGA_LPS reinforced the anticancer effect of αPD-L1, probably by reshaping the tumor microenvironment and intestinal flora, which sheds light on the combination approach to intensify the effect of immune checkpoint inhibitors.

16.
Int J Biol Macromol ; 281(Pt 3): 136226, 2024 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-39383919

RESUMEN

High-fat diets (HFDs) are widely used in aquaculture due to their lipid and protein-conserving effects, thereby reducing feed costs. However, prolonged feeding of HFD often leads to metabolic disorders in fish, such as disruption of hepatic lipid homeostasis, liver injury, and disruption of glucose homeostasis. Fibroblast growth factor 1 (FGF1) plays an essential role in controlling glucose levels in the body and dampening immune reactions. However, its impact on teleosts remains poorly researched. The therapeutic potential of recombinant FGF1 (rFGF1) was examined in a 6-week culture experiment involving rainbow trout (Oncorhynchus mykiss) that were fed an HFD. The results revealed that rFGF1 significantly reduced serum glucose levels and hepatic PEPCK and G6PC activities, but improved hepatic glycogen (P < 0.05), compared to the HFD + PBS group. Further experiments indicated that the inhibitory effect of rFGF1 on hepatic gluconeogenesis was mediated by the cAMP signaling pathway and was dependent on the high expression of PDE4D. In addition, rFGF1 increased hepatic glycogen content, which involves the AKT-GSK3ß axis. Despite this increase, rFGF1 did not lead to glycogen storage disease, as shown by reduced hepatic inflammation as a result of decreased GOT (glutamic oxaloacetic transaminase), GPT (glutamic pyruvic transaminase), and elevated SOD (superoxide dismutase) in the rFGF1-treated group, accompanied by decreased il-1ß, il-6, and xbp-1, and elevated nrf2 and number of hepatocyte autophagosomes. Alterations in gut microbes and short-chain fatty acids (SCFAs) were noted, indicating that rFGF1 caused a notable rise in intestinal Lactobacillus, acetic acid, and butyric acid levels. This study investigated the molecular mechanisms of rFGF1 on glucose metabolism and inflammatory responses in an HFD-fed rainbow trout model, providing new insights to improve the regulation of glucose metabolism in carnivorous fish.

17.
Int J Mol Sci ; 25(19)2024 Oct 02.
Artículo en Inglés | MEDLINE | ID: mdl-39408965

RESUMEN

Polycystic ovary syndrome (PCOS) is a complex disorder that impacts both the endocrine and metabolic systems, often resulting in infertility, obesity, insulin resistance, and cardiovascular complications. The aim of this study is to investigate the role of intestinal flora and its metabolites, particularly short-chain fatty acids (SCFAs), in the development of PCOS, and to assess the effects of metformin therapy on these components. SCFA levels in fecal and blood samples from women with PCOS (n=69) and healthy controls (n=18) were analyzed using Gas Chromatography-Mass Spectrometry (GC/MS) for precise measurement. Fecal microbiota were quantitatively detected by real-time polymerase chain reaction (PCR). To assess the efficacy of six months of metformin treatment, changes in the microbiota and SCFAs in the PCOS group (n=69) were also evaluated. The results revealed that women with PCOS exhibited a significant reduction in beneficial bacteria (namely, the C. leptum group and Prevotella spp.) alongside a notable overgrowth of opportunistic microorganisms (C. perfringens, C. difficile, Staphylococcus spp., and Streptococcus spp.). An overproduction of acetic acid (AA, FC=0.47, p<0.05) and valeric acid (VA, FC=0.54, p<0.05) suggests a link between elevated SCFAs and the development of obesity and PCOS. Interestingly, AA in the bloodstream might offer a protective effect against PCOS by ameliorating key symptoms such as high body mass index (r=-0.33, p=0.02), insulin resistance (r=-0.39, p=0.02), and chronic inflammation. Although serum SCFA levels showed non-significant changes following metformin treatment (p>0.05), the normalization of AA in the gut underscores that metformin exerts a more pronounced effect locally within the gastrointestinal tract. Furthermore, the study identified the most effective model for predicting the success of metformin therapy, based on serum concentrations of butyric acid (BA) and VA, achieving a 91% accuracy rate, 100% sensitivity, and 80% specificity. These promising findings highlight the potential for developing targeted interventions and personalized treatments, ultimately improving clinical outcomes for women with PCOS.


Asunto(s)
Ácidos Grasos Volátiles , Microbioma Gastrointestinal , Metformina , Síndrome del Ovario Poliquístico , Humanos , Síndrome del Ovario Poliquístico/tratamiento farmacológico , Síndrome del Ovario Poliquístico/microbiología , Síndrome del Ovario Poliquístico/metabolismo , Metformina/farmacología , Metformina/uso terapéutico , Femenino , Ácidos Grasos Volátiles/metabolismo , Ácidos Grasos Volátiles/sangre , Microbioma Gastrointestinal/efectos de los fármacos , Adulto , Heces/microbiología , Hipoglucemiantes/uso terapéutico , Hipoglucemiantes/farmacología , Adulto Joven , Estudios de Casos y Controles , Cromatografía de Gases y Espectrometría de Masas
18.
Sci Rep ; 14(1): 23789, 2024 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-39394233

RESUMEN

Lamb diarrhea is primarily induced by bacterial infections, causing great economic and health challenges. Traditional antibiotic treatments raise concerns over drug resistance and environmental contamination. We explored the therapeutic potential of a compound extract from Dracocephalum rupestre Hance and Berberidis Radix against Salmonella-induced diarrhea in lamb. Twenty-five five-week-old Kunming mice (20 ± 5 g) were used. A controlled laboratory experiment, combing histological examinations, serum cytokine level analysis, gut microbiota composition analysis, and short-chain fatty acid quantification were conducted. Results demonstrated significant reparative effects on intestinal mucosal damage of the compound. Compound treatment notably reduced serum levels of inflammatory cytokines (IL-6, IL-8, sigA, and TNF-α), indicating an anti-inflammatory effect. Gene expression analysis of mucosal repair markers (PCNA, TGF, and EGFR) confirmed the positive impacts on intestinal recovery processes after treatment. Microbiota analysis revealed concentration-dependent alterations in gut microbial composition, with a notable increase in beneficial bacterial genera such as Muribaculum and Prevotella, suggesting the role of the compound in promoting gut health. Additionally, short-chain fatty acid analysis indicated an increase in beneficial acids, which are critical for the gut and overall health. This investigation highlights the potential therapeutic benefits of Dracocephalum rupestre Hance combining Berberidis Radix in lamb with Salmonella-induced diarrhea.


Asunto(s)
Diarrea , Microbioma Gastrointestinal , Extractos Vegetales , Animales , Diarrea/tratamiento farmacológico , Diarrea/microbiología , Extractos Vegetales/farmacología , Extractos Vegetales/uso terapéutico , Ratones , Ovinos , Microbioma Gastrointestinal/efectos de los fármacos , Citocinas/metabolismo , Salmonella/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Mucosa Intestinal/metabolismo
19.
Adv Genet ; 112: 367-409, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39396841

RESUMEN

Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death worldwide. In recent years, the impact of the gut microbiota on the development of CRC has become clear. The gut microbiota is the community of microorganisms living in the gut symbiotic relationship with the host. These microorganisms contribute to the development of CRC through various mechanisms that are not yet fully understood. Increasing scientific evidence suggests that metabolites produced by the gut microbiota may influence CRC development by exerting protective and deleterious effects. This article reviews the metabolites produced by the gut microbiota, which are derived from the intake of complex carbohydrates, proteins, dairy products, and phytochemicals from plant foods and are associated with a reduced risk of CRC. These metabolites include short-chain fatty acids (SCFAs), indole and its derivatives, conjugated linoleic acid (CLA) and polyphenols. Each metabolite, its association with CRC risk, the possible mechanisms by which they exert anti-tumour functions and their relationship with the gut microbiota are described. In addition, other gut microbiota-derived metabolites that are gaining importance for their role as CRC suppressors are included.


Asunto(s)
Neoplasias Colorrectales , Microbioma Gastrointestinal , Humanos , Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/metabolismo , Ácidos Grasos Volátiles/metabolismo , Polifenoles/metabolismo , Animales
20.
EClinicalMedicine ; 76: 102844, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39391015

RESUMEN

Background: Obesity drives metabolic disease development. Preventing weight gain during early adulthood could mitigate later-life chronic disease risk. Increased dietary fibre intake, leading to enhanced colonic microbial fermentation and short-chain fatty acid (SCFA) production, is associated with lower body weight. Despite national food policy recommendations to consume 30 g of dietary fibre daily, only 9% of adults achieve this target. Inulin-propionate ester (IPE) selectively increases the production of the SCFA propionate in the colon. In previous studies, IPE has prevented weight gain in middle-aged adults over 6 months, compared with the inulin control. IPE is a novel food ingredient that can be added to various commonly consumed foods with a potential health benefit. This 12-month study aimed to determine whether using IPE to increase colonic propionate prevents further weight gain in overweight younger adults. Methods: This multi-centre randomised-controlled, double-blind trial was conducted in London and Glasgow, UK. Recruited participants were individuals at risk of weight gain, aged between 20 and 40 years and had an overweight body mass index. Sealed Envelope Software was used to randomise participants to consume 10 g of IPE or inulin (control), once per day for 12 months. The primary outcome was the weight gained from baseline to 12 months, analysed by an 'Intention to Treat' strategy. The safety profile and tolerability of IPE were monitored through adverse events and compliance. This study is registered with the International Standard Randomised Controlled Trials (ISRCT) Database (ISRCT number: 16299902). Findings: Participants (n = 135 per study arm) were recruited from July 2019 to October 2021. At 12 months, there was no significant difference in baseline-adjusted mean weight gain for IPE compared with control (1.02 kg, 95% CI: -0.37 to 2.41; p = 0.15; n = 226). Neither the IPE (+1.22 kg) nor the control arm (+0.07 kg) unadjusted mean gains in body weight reached the expected 2 kg threshold. In the IPE arm, fat-free mass was greater by 1.07 kg (95% CI: 0.21-1.93), and blood glucose elevated by 0.11 mmol/L (95% CI: 0.01-0.21). Compliance, determined by intake of ≥50% sachets, was reached by 63% of IPE participants. There were no unexpected adverse events or safety concerns. Interpretation: Our study indicates that at 12 months, IPE did not differentially affect weight gain, compared with the inulin control, in adults between 20 and 40 years of age, at risk of obesity. Funding: NIHR EME Programme (15/185/16).

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...