Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 878
Filtrar
Mais filtros

Medicinas Complementares
Intervalo de ano de publicação
1.
Appl Environ Microbiol ; 90(5): e0004624, 2024 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-38563787

RESUMO

Dietary fiber metabolism by gut microorganisms plays important roles in host physiology and health. Alginate, the major dietary fiber of daily diet seaweeds, is drawing more attention because of multiple biological activities. To advance the understanding of alginate assimilation mechanism in the gut, we show the presence of unsaturated alginate oligosaccharides (uAOS)-specific alginate utilization loci (AUL) in human gut microbiome. As a representative example, a working model of the AUL from the gut microorganism Bacteroides clarus was reconstructed from biochemistry and transcriptome data. The fermentation of resulting monosaccharides through Entner-Doudoroff pathway tunes the metabolism of short-chain fatty acids and amino acids. Furthermore, we show that uAOS feeding protects the mice against dextran sulfate sodium-induced acute colitis probably by remodeling gut microbiota and metabolome. IMPORTANCE: Alginate has been included in traditional Chinese medicine and daily diet for centuries. Recently discovered biological activities suggested that alginate-derived alginate oligosaccharides (AOS) might be an active ingredient in traditional Chinese medicine, but how these AOS are metabolized in the gut and how it affects health need more information. The study on the working mechanism of alginate utilization loci (AUL) by the gut microorganism uncovers the role of unsaturated alginate oligosaccharides (uAOS) assimilation in tuning short-chain fatty acids and amino acids metabolism and demonstrates that uAOS metabolism by gut microorganisms results in a variation of cell metabolites, which potentially contributes to the physiology and health of gut.


Assuntos
Alginatos , Microbioma Gastrointestinal , Oligossacarídeos , Alginatos/metabolismo , Oligossacarídeos/metabolismo , Camundongos , Animais , Humanos , Colite/microbiologia , Colite/induzido quimicamente , Camundongos Endogâmicos C57BL , Ácidos Graxos Voláteis/metabolismo , Inflamação/metabolismo , Sulfato de Dextrana , Fibras na Dieta/metabolismo
2.
Cytokine ; 179: 156608, 2024 07.
Artigo em Inglês | MEDLINE | ID: mdl-38631185

RESUMO

BACKGROUND: Mounting evidence revealed that an imbalance of Gut Microbiota (GM) leads to metabolic disorders. Synbiotics through regulation of GM composition can be an effective intervention in the management of metabolic diseases. This study aimed to investigate the effects of multi-species synbiotic supplementation on serum interleukin10 (IL-10) and fecal Short Chain Fatty Acids (SCFAs) in patients with dyslipidemia. METHODS: In this double-blind, randomized, placebo-controlled clinical trial, fifty-six adult men with dyslipidemia were randomly allocated to intervention and control groups and received either synbiotic or placebo powder twice a day for 12 weeks. Each synbiotic sachet contained 6 species of probiotic microorganisms with a total dose of 3 × 1010 Colony Forming Unit (CFU) and 5 gr inulin and Fructooligosaccharide (FOS) as prebiotics. Blood and stool samples were collected at the baseline and end of the study. Dietary intake, physical activity, anthropometric measurements, serum IL-10, and fecal SCFAs were assessed before and after the intervention. RESULT: There were no significant differences between the baseline characteristics of patients in the two groups. Serum IL-10 was increased in the synbiotic group (p < 0.0001). Moreover, synbiotic supplementation increased fecal concentration of acetate (p < 0.0001), butyrate (p = 0.043), propionate (p < 0.0001), and valerate (p < 0.026). A significant positive correlation was observed between the changes in fecal butyrate level and serum IL-10 concentration in the control group (r = 0.48, p = 0.01). CONCLUSIONS: A Twelve-week synbiotic supplementation increased fecal SCFAs and improved inflammation in adult men with dyslipidemia.


Assuntos
Suplementos Nutricionais , Dislipidemias , Ácidos Graxos Voláteis , Fezes , Interleucina-10 , Simbióticos , Humanos , Masculino , Fezes/microbiologia , Fezes/química , Simbióticos/administração & dosagem , Método Duplo-Cego , Interleucina-10/sangue , Dislipidemias/sangue , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/sangue , Pessoa de Meia-Idade , Adulto , Microbioma Gastrointestinal , Oligossacarídeos
3.
Nat Commun ; 15(1): 3502, 2024 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-38664378

RESUMO

Beneficial gut bacteria are indispensable for developing colonic mucus and fully establishing its protective function against intestinal microorganisms. Low-fiber diet consumption alters the gut bacterial configuration and disturbs this microbe-mucus interaction, but the specific bacteria and microbial metabolites responsible for maintaining mucus function remain poorly understood. By using human-to-mouse microbiota transplantation and ex vivo analysis of colonic mucus function, we here show as a proof-of-concept that individuals who increase their daily dietary fiber intake can improve the capacity of their gut microbiota to prevent diet-mediated mucus defects. Mucus growth, a critical feature of intact colonic mucus, correlated with the abundance of the gut commensal Blautia, and supplementation of Blautia coccoides to mice confirmed its mucus-stimulating capacity. Mechanistically, B. coccoides stimulated mucus growth through the production of the short-chain fatty acids propionate and acetate via activation of the short-chain fatty acid receptor Ffar2, which could serve as a new target to restore mucus growth during mucus-associated lifestyle diseases.


Assuntos
Colo , Fibras na Dieta , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Mucosa Intestinal , Receptores de Superfície Celular , Animais , Fibras na Dieta/metabolismo , Ácidos Graxos Voláteis/metabolismo , Camundongos , Colo/metabolismo , Colo/microbiologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/genética , Feminino , Camundongos Endogâmicos C57BL , Muco/metabolismo , Transplante de Microbiota Fecal , Simbiose , Propionatos/metabolismo , Clostridiales/metabolismo , Acetatos/metabolismo , Adulto
4.
Int Immunopharmacol ; 132: 112027, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38603860

RESUMO

BACKGROUND AND PURPOSE: Osteoporosis (OP) is a frequent clinical problem for the elderly. Traditional Chinese Medicine (TCM) has achieved beneficial results in the treatment of OP. Ziyuglycoside II (ZGS II) is a major active compound of Sanguisorba officinalis L. that has shown anti-inflammation and antioxidation properties, but little information concerning its anti-OP potential is available. Our research aims to investigate the mechanism of ZGS II in ameliorating bone loss by inflammatory responses and regulation of gut microbiota and short chain fatty acids (SCFAs) in ovariectomized (OVX) mice. METHODS: We predicted the mode of ZGS II action on OP through network pharmacology and molecular docking, and an OVX mouse model was employed to validate its anti-OP efficacy. Then we analyzed its impact on bone microstructure, the levels of inflammatory cytokines and pain mediators in serum, inflammation in colon, intestinal barrier, gut microbiota composition and SCFAs in feces. RESULTS: Network pharmacology identified 55 intersecting targets of ZGS II related to OP. Of these, we predicted IGF1 may be the core target, which was successfully docked with ZGS II and showed excellent binding ability. Our in vivo results showed that ZGS II alleviated bone loss in OVX mice, attenuated systemic inflammation, enhanced intestinal barrier, reduced the pain threshold, modulated the abundance of gut microbiota involving norank_f__Muribaculaceae and Dubosiella, and increased the content of acetic acid and propanoic acid in SCFAs. CONCLUSIONS: Our data indicated that ZGS II attenuated bone loss in OVX mice by relieving inflammation and regulating gut microbiota and SCFAs.


Assuntos
Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Simulação de Acoplamento Molecular , Osteoporose , Ovariectomia , Animais , Microbioma Gastrointestinal/efeitos dos fármacos , Ácidos Graxos Voláteis/metabolismo , Feminino , Camundongos , Osteoporose/tratamento farmacológico , Osteoporose/imunologia , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Saponinas/farmacologia , Saponinas/uso terapêutico , Humanos , Citocinas/metabolismo , Farmacologia em Rede , Inflamação/tratamento farmacológico
5.
J Transl Med ; 22(1): 222, 2024 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429821

RESUMO

BACKGROUND: Colonoscopy is a classic diagnostic method with possible complications including abdominal pain and diarrhoea. In this study, gut microbiota dynamics and related metabolic products during and after colonoscopy were explored to accelerate gut microbiome balance through probiotics. METHODS: The gut microbiota and fecal short-chain fatty acids (SCFAs) were analyzed in four healthy subjects before and after colonoscopy, along with seven individuals supplemented with Clostridium butyricum. We employed 16S rRNA sequencing and GC-MS to investigate these changes. We also conducted bioinformatic analysis to explore the buk gene, encoding butyrate kinase, across C. butyricum strains from the human gut. RESULTS: The gut microbiota and fecal short-chain fatty acids (SCFAs) of four healthy subjects were recovered on the 7th day after colonoscopy. We found that Clostridium and other bacteria might have efficient butyric acid production through bioinformatic analysis of the buk and assessment of the transcriptional level of the buk. Supplementation of seven healthy subjects with Clostridium butyricum after colonoscopy resulted in a quicker recovery and stabilization of gut microbiota and fecal SCFAs on the third day. CONCLUSION: We suggest that supplementation of Clostridium butyricum after colonoscopy should be considered in future routine clinical practice.


Assuntos
Clostridium butyricum , Microbioma Gastrointestinal , Microbiota , Humanos , Clostridium butyricum/genética , Clostridium butyricum/metabolismo , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Ácidos Graxos Voláteis/metabolismo , Colonoscopia , Ácido Butírico/farmacologia , Ácido Butírico/metabolismo
6.
Pain ; 165(9): e106-e114, 2024 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-38452211

RESUMO

ABSTRACT: Opioids are commonly prescribed to patients with chronic pain. Chronic opioid usage comes with a slew of serious side effects, including opioid-induced hyperalgesia (OIH). The patients with long-term opioid treatment experience paradoxical increases in nociceptive hypersensitivity, namely, OIH. Currently, treatment options for OIH are extremely lacking. In this study, we show that the ketogenic diet recovers the abnormal pain behavior caused by chronic morphine treatment in male mice, and we further show that the therapeutic effect of the ketogenic diet is mediated through gut microbiome. Our 16S rRNA sequencing demonstrates that chronic morphine treatment causes changes in mouse gut microbiota, specifically a decrease in short-chain fatty acids-producing bacteria, and the sequencing data also show that the ketogenic diet rescues those bacteria in the mouse gut. More importantly, we show that supplementation with short-chain fatty acids (butyrate, propionate, and acetate) can delay the onset of OIH, indicating that short-chain fatty acids play a direct role in the development of OIH. Our findings suggest that gut microbiome could be targeted to treat OIH, and the ketogenic diet can be used as a complementary approach for pain relief in patients with chronic opioid treatment. We only used male mice in this study, and thus, our findings cannot be generalized to both sexes.


Assuntos
Analgésicos Opioides , Dieta Cetogênica , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Hiperalgesia , Morfina , Animais , Dieta Cetogênica/métodos , Hiperalgesia/dietoterapia , Hiperalgesia/metabolismo , Hiperalgesia/induzido quimicamente , Masculino , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Camundongos , Ácidos Graxos Voláteis/metabolismo , Morfina/farmacologia , Camundongos Endogâmicos C57BL
7.
Clin Nutr ; 43(4): 969-980, 2024 04.
Artigo em Inglês | MEDLINE | ID: mdl-38452522

RESUMO

BACKGROUND & AIMS: Improving maternal gut health in pregnancy and lactation is a potential strategy to improve immune and metabolic health in offspring and curtail the rising rates of inflammatory diseases linked to alterations in gut microbiota. Here, we investigate the effects of a maternal prebiotic supplement (galacto-oligosaccharides and fructo-oligosaccharides), ingested daily from <21 weeks' gestation to six months' post-partum, in a double-blinded, randomised placebo-controlled trial. METHODS: Stool samples were collected at multiple timepoints from 74 mother-infant pairs as part of a larger, double-blinded, randomised controlled allergy intervention trial. The participants were randomised to one of two groups; with one group receiving 14.2 g per day of prebiotic powder (galacto-oligosaccharides GOS and fructo-oligosaccharides FOS in ratio 9:1), and the other receiving a placebo powder consisting of 8.7 g per day of maltodextrin. The faecal microbiota of both mother and infants were assessed based on the analysis of bacterial 16S rRNA gene (V4 region) sequences, and short chain fatty acid (SCFA) concentrations in stool. RESULTS: Significant differences in the maternal microbiota profiles between baseline and either 28-weeks' or 36-weeks' gestation were found in the prebiotic supplemented women. Infant microbial beta-diversity also significantly differed between prebiotic and placebo groups at 12-months of age. Supplementation was associated with increased abundance of commensal Bifidobacteria in the maternal microbiota, and a reduction in the abundance of Negativicutes in both maternal and infant microbiota. There were also changes in SCFA concentrations with maternal prebiotics supplementation, including significant differences in acetic acid concentration between intervention and control groups from 20 to 28-weeks' gestation. CONCLUSION: Maternal prebiotic supplementation of 14.2 g per day GOS/FOS was found to favourably modify both the maternal and the developing infant gut microbiome. These results build on our understanding of the importance of maternal diet during pregnancy, and indicate that it is possible to intervene and modify the development of the infant microbiome by dietary modulation of the maternal gut microbiome.


Assuntos
Microbiota , Prebióticos , Feminino , Humanos , Lactente , Gravidez , Suplementos Nutricionais , Ácidos Graxos Voláteis/metabolismo , Lactação , Mães , Oligossacarídeos , Pós , RNA Ribossômico 16S , Recém-Nascido
8.
Phytomedicine ; 128: 155413, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38513377

RESUMO

AIM OF THE STUDY: To evaluate the in vitro and in vivo anti-metastasis efficacy of Jianpi Yangzheng (JPYZ) decoction against gastric cancer (GC) and its potential mechanisms. MATERIALS AND METHODS: The distant metastasis of GC cells administered via tail vein injection was assessed using the pre-metastatic niche (PMN) model. 16S rRNA sequencing and GC-MS/MS were applied to determine the component of the gut microbiota and content of short-chain fatty acids (SCFAs) in feces of mice, respectively. The proportion of myeloid-derived suppressor cells (MDSCs) in the lung was evaluated by flow cytometry and immunofluorescence. Serum or tissue levels of inflammation factors including IL-6, IL-10 and TGF-ß were determined by ELISA or Western blot respectively. RESULTS: Injecting GC cells into the tail vein of mice led to the development of lung metastases and also resulted in alterations in the composition of gut microbiota and the levels of SCFAs produced. Nevertheless, JPYZ treatment robustly impeded the effect of GC cells administration. Mechanically, JPYZ treatment not only prevented the alteration in gut microbiota structure, but also restored the SCFAs content induced by GC cells administration. Specifically, JPYZ treatment recovered the relative abundance of genera Moryella, Helicobacter, Lachnoclostridium, Streptococcus, Tuzzerella, GCA-900066575, uncultured_Lachnospiraceae, Rikenellaceae_RC9_gut_group and uncultured_bacterium_Muribaculaceae to near the normal control levels. In addition, JPYZ abrogated MDSCs accumulation in the lung tissue and blocked inflammation factors overproduction in the serum and lung tissues, which subsequently impede the formation of the immunosuppressive microenvironment. Correlation analysis revealed that the prevalence of Rikenellaceae in the model group exhibited a positive correlation with MDSCs proportion and inflammation factor levels. Conversely, the scarcity of Muribaculaceae in the model group showed a negative correlation with these parameters. This suggests that JPYZ might exert an influence on the gut microbiota and their metabolites, such as SCFAs, potentially regulating the formation of the PMN and consequently impacting the outcome of GC metastasis. CONCLUSION: These findings suggest that GC cells facilitate metastasis by altering the gut microbiota composition, affecting the production of SCFAs, and recruiting MDSCs to create a pro-inflammatory pre-metastatic niche. JPYZ decoction counteracts this process by reshaping the gut microbiota structure, enhancing SCFA production, and inhibiting the formation of the pre-metastatic microenvironment, thereby exerting an anti-metastatic effect.


Assuntos
Medicamentos de Ervas Chinesas , Microbioma Gastrointestinal , Neoplasias Pulmonares , Células Supressoras Mieloides , Neoplasias Gástricas , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Medicamentos de Ervas Chinesas/farmacologia , Camundongos , Células Supressoras Mieloides/efeitos dos fármacos , Linhagem Celular Tumoral , Ácidos Graxos Voláteis/metabolismo , Camundongos Endogâmicos BALB C , Humanos , RNA Ribossômico 16S , Masculino , Fezes/microbiologia , Feminino
9.
Nutrients ; 16(5)2024 Feb 27.
Artigo em Inglês | MEDLINE | ID: mdl-38474789

RESUMO

BACKGROUND: Regular exercise has been described to modify both the diversity and the relative abundance of certain bacterial taxa. To our knowledge, the effect of a cycling stage race, which entails extreme physiological and metabolic demands, on the gut microbiota composition and its metabolic activity has not been analysed. OBJECTIVE: The aim of this cohort study was to analyse the dynamics of faecal microbiota composition and short-chain fatty acids (SCFAs) content of professional cyclists over a Grand Tour and their relationship with performance and dietary intake. METHODS: 16 professional cyclists competing in La Vuelta 2019 were recruited. Faecal samples were collected at four time points: the day before the first stage (A); after 9 stages (B); after 15 stages (C); and on the last stage (D). Faecal microbiota populations and SCFA content were analysed using 16S rRNA sequencing and gas chromatography, respectively. A principal component analysis (PCA) followed by Generalised Estimating Equation (GEE) models were carried out to explore the dynamics of microbiota and SCFAs and their relationship with performance. RESULTS: Bifidobacteriaceae, Coriobacteriaceae, Erysipelotrichaceae, and Sutterellaceae dynamics showed a strong final performance predictive value (r = 0.83, ranking, and r = 0.81, accumulated time). Positive correlations were observed between Coriobacteriaceae with acetate (r = 0.530) and isovalerate (r = 0.664) and between Bifidobacteriaceae with isobutyrate (r = 0.682). No relationship was observed between SCFAs and performance. The abundance of Erysipelotrichaceae at the beginning of La Vuelta was directly related to the previous intake of complex-carbohydrate-rich foods (r = 0.956), while during the competition, the abundance of Bifidobacteriaceae was negatively affected by the intake of simple carbohydrates from supplements (r = -0.650). CONCLUSIONS: An ecological perspective represents more realistically the relationship between gut microbiota composition and performance compared to single-taxon approaches. The composition and periodisation of diet and supplementation during a Grand Tour, particularly carbohydrates, could be designed to modulate gut microbiota composition to allow better performance.


Assuntos
Microbioma Gastrointestinal , Humanos , RNA Ribossômico 16S/genética , Estudos de Coortes , Ácidos Graxos Voláteis/metabolismo , Fezes/microbiologia , Ingestão de Alimentos , Exercício Físico , Carboidratos/análise
10.
Water Res ; 254: 121401, 2024 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-38447378

RESUMO

Although being viewed as a promising technology for reclamation of carbon and phosphorus from excess sludge, anaerobic fermentation (AF) grapples with issues such as a low yield of volatile fatty acids (VFAs) and high phosphorus recovery costs. In this study, we synthesized Fe3O4@MOF-808 (FeM) with abundant defects and employed it to simultaneously enhance VFAs and phosphorus recovery during sludge anaerobic fermentation. Through pre-oxidization of sludge catalyzed by FeM-induced peroxydisulfate, the soluble organic matter increased by 2.54 times, thus providing ample substrate for VFAs production. Subsequent AF revealed a remarkable 732.73 % increase in VFAs and a 1592.95 % increase in phosphate. Factors contributing to the high VFAs yield include the non-biological catalysis of unsaturated Zr active sites in defective FeM, enhancing protein hydrolysis, and the inhibition of methanogenesis due to electron competition arising from the transformation between Fe(III) and Fe(II) under Zr influence. Remarkably, FeM exhibited an adsorption capacity of up to 92.64 % for dissolved phosphate through ligand exchange and electrostatic attractions. Furthermore, FeM demonstrated magnetic separation capability from the fermentation broth, coupled with excellent stability and reusability in both catalysis and adsorption processes.


Assuntos
Fósforo , Esgotos , Fermentação , Esgotos/química , Anaerobiose , Carbono , Compostos Férricos , Ácidos Graxos Voláteis/metabolismo , Fosfatos , Concentração de Íons de Hidrogênio
11.
Gut Microbes ; 16(1): 2307542, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38319728

RESUMO

The gut microbiota and Short-chain fatty acids (SCFAs) can influence the progression of diseases, yet the role of these factors on gastric cancer (GC) remains uncertain. In this work, the analysis of the gut microbiota composition and SCFA content in the blood and feces of both healthy individuals and GC patients indicated that significant reductions in the abundance of intestinal bacteria involved in SCFA production were observed in GC patients compared with the controls. ABX mice transplanted with fecal microbiota from GC patients developed more tumors during the induction of GC and had lower levels of butyric acid. Supplementation of butyrate during the induction of gastric cancer along with H. pylori and N-methyl-N-nitrosourea (MNU) in WT in GPR109A-/-mice resulted in fewer tumors and more IFN-γ+ CD8+ T cells, but this effect was significantly weakened after knockout of GPR109A. Furthermore, In vitro GC cells and co-cultured CD8+ T cells or CAR-Claudin 18.2+ CD8+ T cells, as well as in vivo tumor-bearing studies, have indicated that butyrate enhanced the killing function of CD8+ T cells or CAR-Claudin 18.2+ CD8+ T cells against GC cells through G protein-coupled receptor 109A (GPR109A) and homologous domain protein homologous box (HOPX). Together, these data highlighted that the restoration of gut microbial butyrate enhanced CD8+ T cell cytotoxicity via GPR109A/HOPX, thus inhibiting GC carcinogenesis, which suggests a novel theoretical foundation for GC management against GC.


Assuntos
Microbioma Gastrointestinal , Neoplasias Gástricas , Humanos , Camundongos , Animais , Butiratos/metabolismo , Microbioma Gastrointestinal/fisiologia , Linfócitos T CD8-Positivos , Ácidos Graxos Voláteis/metabolismo , Ácido Butírico , Claudinas
12.
J Ethnopharmacol ; 325: 117776, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38307354

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Honeysuckle, first documented in the Miscellaneous Records of Famous Physicians, is known for its ability to expel toxin and cool blood to stop diarrhea. Modern pharmacological research has shown that honeysuckle has anti-inflammatory, antibacterial, antioxidant, and immune-regulating effects and is widely used in clinical practice. However, the effect of honeysuckle on ulcerative colitis (UC) is still not fully understood, which presents challenges for quality control, research and development. AIM OF THE STUDY: This study aimed to determine the anti-inflammatory properties and mechanism of action of aqueous extracts of honeysuckle in the treatment of ulcerative colitis. MATERIALS AND METHODS: The dextran sodium sulfate (DSS) induced-ulcerative colitis mouse model was established, and the mice were divided into five groups: the control group, the model group, and the low, medium, and high dose honeysuckle treatment groups. RESULTS: All dose groups of honeysuckle were found to significantly reduce IL-6 and TNF-α levels and regulate DSS-induced mRNA levels of CLDN4, COX-2, IL-6, INOS, MUC-2, occludin and NLRP3. The high-dose group displayed the most effective inhibition, and a differentially expressed mRNA detection indicated abnormal mRNA expression. The 16sRNA sequencing revealed that the honeysuckle was able to significantly upregulate the abundance of beneficial bacteria and downregulate the abundance of harmful bacteria. The study of short-chain fatty acids revealed that the levels of acetic, propionic, isobutyric, valeric and isovaleric acids were significantly increased after administering honeysuckle at medium and high doses. CONCLUSION: Honeysuckle reduces the production of pro-inflammatory cytokines, increases the content of short-chain fatty acids and restores the intestinal ecological balance, resulting in better therapeutic effects.


Assuntos
Colite Ulcerativa , Colite , Lonicera , Camundongos , Animais , Colite Ulcerativa/induzido quimicamente , Colite Ulcerativa/tratamento farmacológico , Colite Ulcerativa/metabolismo , Colo , Interleucina-6/genética , Interleucina-6/metabolismo , Anti-Inflamatórios/efeitos adversos , RNA Mensageiro/metabolismo , Ácidos Graxos Voláteis/metabolismo , Sulfato de Dextrana/toxicidade , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Colite/tratamento farmacológico
13.
Anim Sci J ; 95(1): e13929, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38400743

RESUMO

This study aimed to investigate the effect of supplementing Isochrysis galbana (I. galbana) at levels of 0 (control), 1, 2, 3, 4, and 5 (g/100 g DM) of the diet on the gas production kinetics, methane production, rumen fermentation parameters, and relative microbial population in vitro. Supplementation of I. galbana at high level (5 g/100 g DM) caused a significant decrease in total gas production (p < 0.05). High supplementation rates (4 and 5 g/100 g DM) decreased CH4 production relative to the control by 18.4% and 23.2%, respectively. Although rumen ammonia nitrogen (N-NH3) and total volatile fatty acids (VFA) concentrations were affected by dietary treatments, but the VFA profile did not changed. The relative proportion of protozoa and methanogenic archaea as well as Anaerovibrio lipolytica, Prevotella spp., Ruminococcus flavefaciens, and Fibrobacter succinogenes were decreased significantly as a result of microalgae supplementation. However, the relative abundance of Ruminococcus albus, Butyrivibrio fibrisolvens and Selenomonas ruminantium were significantly increased (p < 0.05), related to the control group. As well, the pH was not affected by dietary treatments. It was concluded that I. galbana reduced in vitro CH4 production and methanogenic archaea that its worth to be investigated further in in vivo studies.


Assuntos
Suplementos Nutricionais , Haptófitas , Animais , Suplementos Nutricionais/análise , Rúmen/metabolismo , Fermentação , Dieta , Ácidos Graxos Voláteis/metabolismo , Archaea , Metano/metabolismo , Ração Animal/análise , Digestão
14.
Zhongguo Zhong Yao Za Zhi ; 49(1): 208-215, 2024 Jan.
Artigo em Chinês | MEDLINE | ID: mdl-38403353

RESUMO

This study aimed to investigate the regulatory effects of Zuogui Jiangtang Jieyu Formula(ZJJ) on the intestinal flora, short chain fatty acids(SCFAs), and neuroinflammation in rats with diabetes mellitus complicated depression(DD). The DD model was established in rats and model rats were randomly divided into a model group, a positive drug(metformin + fluoxetine) group, a ZJJ low-dose group, and a ZJJ high-dose group, with eight rats in each group. Another eight rats were assigned to the blank group. Subsequently, depressive-like behavior test was conducted on the rats, and cerebrospinal fluid samples were collected to measure pro-inflammatory cytokines [interleukin-1ß(IL-1ß), interleukin-6(IL-6), and tumor necrosis factor-α(TNF-α)]. Blood serum samples were collected to measure proteins related to the hypothalamic-pituitary-adrenal axis(HPA axis), including corticotropin-releasing hormone(CRH), adrenocorticotropic hormone(ACTH), and cortisol(CORT), as well as glucose metabolism. Gut contents were collected from each group for 16S rRNA sequencing analysis of intestinal flora and SCFAs sequencing. The results indicated that ZJJ not only improved glucose metabolism in DD rats(P<0.01) but also alleviated depressive-like behavior(P<0.05) and HPA axis hyperactivity(P<0.05 or P<0.01). Besides, it also improved the neuroinflammatory response in the brain, as evidenced by a significant reduction in pro-inflammatory cytokines in cerebrospinal fluid(P<0.05 or P<0.01). Additionally, ZJJ improved the intestinal flora, causing the intestinal flora in DD rats to resemble that of the blank group, characterized by an increased Firmicutes abundance. ZJJ significantly increased the levels of SCFAs(acetic acid, butyric acid, valeric acid, and isovaleric acid)(P<0.01). Therefore, it is deduced that ZJJ can effectively ameliorate intestinal flora dysbiosis, regulate SCFAs, and thereby improve both glucose metabolism disturbances and depressive-like behavior in DD.


Assuntos
Diabetes Mellitus , Medicamentos de Ervas Chinesas , Microbioma Gastrointestinal , Ratos , Animais , Sistema Hipotálamo-Hipofisário/metabolismo , Depressão/tratamento farmacológico , RNA Ribossômico 16S , Sistema Hipófise-Suprarrenal/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Citocinas/genética , Citocinas/metabolismo , Glucose/metabolismo , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia
15.
Phytomedicine ; 126: 155470, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38417242

RESUMO

BACKGROUND: Asthma affects 3% of the global population, leading to over 0.25 million deaths. Due to its complexity, asthma is difficult to cure or prevent, and current therapies have limitations. This has led to a growing demand for alternative asthma treatments. We found rosmarinic acid (RosA) as a potential new drug candidate from natural medicine. However, RosA has poor bioavailability and remains mainly in the gastrointestinal tract after oral administration, suggesting the involvement of gut microbiota in its bioactivity. PURPOSE: To investigate the mechanism of RosA in alleviating allergic asthma by gut-lung axis. METHODS: We used 16S rRNA gene sequencing and metabolites analysis to investigate RosA's modulation of gut microbiota. Techniques of molecular biology and metabolomics were employed to study the pharmacological mechanism of RosA. Cohousing was used to confirm the involvement of gut microbiota in RosA-induced improvement of allergic asthma. RESULTS: RosA decreased cholate levels from spore-forming bacteria, leading to reduced 5-hydroxytryptamine (5-HT) synthesis, bronchoconstriction, vasodilation, and inflammatory cell infiltration. It also increased short-chain fatty acids (SCFAs) levels, facilitating the expression of intestinal tight junction proteins to promote intestinal integrity. SCFAs upregulated intestinal monocarboxylate transporters (MCTs), thereby improving their systemic delivery to reduce Th2/ILC2 mediated inflammatory response and suppress eosinophil influx and mucus production in lung. Additionally, RosA inhibited lipopolysaccharide (LPS) production and translocation, leading to reduced TLR4-NFκB mediated pulmonary inflammation and oxidative stress. CONCLUSIONS: The anti-asthmatic mechanism of oral RosA is primarily driven by modulation of gut microbiota-derived 5-HT, SCFAs, and LPS, achieving a combined synergistic effect. RosA is a safe, effective, and reliable drug candidate that could potentially replace glucocorticoids for asthma treatment.


Assuntos
Asma , Ácido Rosmarínico , Humanos , Imunidade Inata , RNA Ribossômico 16S/genética , Lipopolissacarídeos , Serotonina , Linfócitos , Asma/tratamento farmacológico , Asma/metabolismo , Pulmão/metabolismo , Ácidos Graxos Voláteis/metabolismo
16.
Int J Biol Macromol ; 260(Pt 1): 129430, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38228199

RESUMO

In this study, a new polysaccharide (PSPJ) with specific molecular weight and monosaccharide compositions was isolated and purified from the water extract of Panacis Japonici Rhizoma (PJR). 16S rRNA analysis and untargeted metabolomic analysis were used to assess PSPJ's efficacy in averting non-alcoholic fatty liver disease (NAFLD). This study indicated that PSPJ significantly reduced liver fat accumulation, the increase in blood lipids and ALT caused by HFD, indicating that PSPJ can prevent NAFLD. We demonstrated through cell experiments that PSPJ does not directly affect liver cells. The gut microbiota disorder and alterations in short-chain fatty acids (SCFAs) induced by the high-fat diet (HFD) were ameliorated by PSPJ, as evidenced by the analysis of 16S rRNA. In particular, supplementing PSPJ reduced the abundance of Turicibacter, Dubosiella, and Staphylococcus, and increased the abundance of Bacteroides, Blautia, and Lactobacillus. Untargeted metabolomic analysis shows that PSPJ improves liver metabolic disorders by regulating arachidonic acid metabolism, carbohydrate digestion and absorption, fatty acid biosynthesis, fatty acid metabolism and retinol metabolism. The findings of our investigation indicate that PSPJ has the potential to modulate liver metabolism through alterations in the composition of intestinal bacteria, hence preventing NAFLD.


Assuntos
Microbioma Gastrointestinal , Hepatopatia Gordurosa não Alcoólica , Panax , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/metabolismo , RNA Ribossômico 16S/genética , RNA Ribossômico 16S/metabolismo , Panax/metabolismo , Fígado/metabolismo , Ácidos Graxos Voláteis/metabolismo , Polissacarídeos/farmacologia , Polissacarídeos/metabolismo , Dieta Hiperlipídica/efeitos adversos , Camundongos Endogâmicos C57BL
17.
J Anim Sci ; 1022024 Jan 03.
Artigo em Inglês | MEDLINE | ID: mdl-38206107

RESUMO

Research into the potential use of various dietary feed supplements to reduce methane (CH4) production from ruminants has proliferated in recent years. In this study, two 8-wk long experiments were conducted with mature ewes and incorporated the use of a variety of natural dietary feed supplements offered either independently or in combination. Both experiments followed a randomized complete block design. Ewes were offered a basal diet in the form of ad libitum access to grass silage supplemented with 0.5 kg concentrates/ewe/d. The entire daily dietary concentrate allocation, incorporating the respective feed supplement, was offered each morning, and this was followed by the daily silage allocation. In experiment 1, the experimental diets contained 1) no supplementation (CON), 2) Ascophyllum nodosum (SW), 3) A. nodosum extract (EX1), 4) a blend of garlic and citrus extracts (GAR), and 5) a blend of essential oils (EO). In experiment 2, the experimental diets contained 1) no supplementation (CON), 2) A. nodosum extract (EX2), 3) soya oil (SO), and 4) a combination of EX2 and SO (EXSO). Twenty ewes per treatment were individually housed during both experiments. Methane was measured using portable accumulation chambers. Rumen fluid was collected at the end of both experiments for subsequent volatile fatty acid (VFA) and ammonia analyses. Data were analyzed using mixed models ANOVA (PROC MIXED, SAS v9.4). Statistically significant differences between treatment means were considered when P < 0.05. Dry matter intake was not affected by diet in either experiment (P > 0.05). Ewes offered EO tended to have an increased feed:gain ratio relative to CON (P < 0.10) and SO tended to increase the average daily gain (P < 0.10) which resulted in animals having a higher final body weight (P < 0.05) than CON. Ewes offered EX1 and SO emitted 9% less CH4 g/d than CON. The only dietary treatment to have an effect on rumen fermentation variables relative to CON was SW, which enhanced total VFA production (P < 0.05). In conclusion, the A. nodosum extract had inconsistent results on CH4 emissions whereby EX1 reduced CH4 g/d while EX2 had no mitigating effect on CH4 production, likely due to the differences in PT content reported for EX1 and EX2. SO was the only dietary feed supplement assessed in the current study that enhanced animal performance whilst mitigating daily CH4 production.


Reducing methane emissions from agriculture is vital to minimize the effects of global warming and to meet greenhouse gas reduction targets set by EU policy. In this experiment, a range of natural feed supplements were offered to mature ewes through the concentrated portion of their diet. Soya oil and brown seaweed extract reduced daily methane emissions by 9% when offered independently of each other; however, no reduction in methane was observed when combined. Additionally, inclusion of soya oil improved animal weight gain. Results from the current experiment may contribute to the development of a targeted dietary strategy to reduce methane emissions from livestock.


Assuntos
Dieta , Metano , Ovinos , Animais , Feminino , Metano/metabolismo , Dieta/veterinária , Suplementos Nutricionais/análise , Ruminantes , Silagem/análise , Ácidos Graxos Voláteis/metabolismo , Rúmen/metabolismo , Óleo de Soja/metabolismo , Extratos Vegetais , Fermentação , Ração Animal/análise , Lactação , Digestão
18.
Shock ; 61(1): 120-131, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-37962207

RESUMO

ABSTRACT: M1 macrophage-mediated inflammation is critical in sepsis. We previously found the protective role of astragaloside intravenous (AS-IV) in sepsis-associated gut impairment, whose specific mechanism remains unknown. Gut microbiota modulates gut homeostatic balance to avoid excessive inflammation. Here, we aimed to investigate effects of AS-IV on gut macrophages polarization and potential roles of gut microbiota and short chain fatty acids (SCFAs) in septic gut damage. Mice were pretreated by AS-IV gavage for 7 days before cecal ligation and puncture. M1 polarization of gut lamina propria macrophages (LpMs) was promoted by cecal ligation and puncture, accompanied by abnormal cytokines release and intestinal barrier dysfunction. NLRP3 inflammasome was activated in M1 LpMs. 16S rRNA sequencing demonstrated gut microbiota imbalance. The levels of acetate, propionate, and butyrate in fecal samples decreased. Notably, AS-IV reversed LpMs M1/M2 polarization, lightened gut inflammation and barrier injury, reduced NLRP3 inflammasome expression in LpMs, restored the diversity of gut microbiome, and increased butyrate levels. Similarly, these benefits were mimicked by fecal microbiota transplantation or exogenous butyrate supplementation. In Caco-2 and THP-1 cocultured model, LPS and interferon γ caused THP-1 M1 polarization, Caco-2 barrier impairment, abnormal cytokines release, and high NLRP3 inflammasome expression in THP-1 cells, all of which were mitigated by butyrate administration. However, these protective effects of butyrate were abrogated by NLRP3 gene overexpression in THP-1. In conclusion, AS-IV can ameliorate sepsis-induced gut inflammation and barrier dysfunction by modulating M1/M2 polarization of gut macrophages, whose underlying mechanism may be restoring gut microbiome and SCFA to restrain NLRP3 inflammasome activation.


Assuntos
Microbioma Gastrointestinal , Saponinas , Sepse , Triterpenos , Humanos , Animais , Camundongos , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Inflamassomos/metabolismo , Células CACO-2 , RNA Ribossômico 16S/metabolismo , Ácidos Graxos Voláteis/metabolismo , Butiratos/metabolismo , Inflamação/metabolismo , Macrófagos/metabolismo , Sepse/metabolismo , Citocinas/metabolismo
19.
Food Chem ; 438: 137982, 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-37979272

RESUMO

Fecal samples from 20 healthy adults were collected for in vitro fermentation experiments to investigate the effects of combined probiotics on the utilization of grape seed extract in humans. After fermenting for 24 h, short-chain fatty acids, metabolites, and gut microbiota composition were analyzed. Short-chain fatty acids in the grape seed extract probiotics group were significantly higher than those in the grape seed extract group. Probiotics significantly enhanced the conversion and utilization of catechins and epicatechins in grape seed extract group and increased the production of 3-hydroxyphenylacetic acid. The 16S rRNA sequencing results revealed that compound probiotics significantly increased the relative abundance of Lacticaseibacillus, HT002, Bifidobacterium, and Lactobacillus and reduced that of Escherichia-Shigella. Our findings showed considerable individual variability in the metabolic utilization of grape seed extract in humans. The consumption of probiotics appears to significantly enhance the utilization.


Assuntos
Extrato de Sementes de Uva , Probióticos , Adulto , Humanos , Polifenóis , RNA Ribossômico 16S , Ácidos Graxos Voláteis/metabolismo
20.
Arthritis Rheumatol ; 76(4): 647-659, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37994265

RESUMO

OBJECTIVE: The objective for this study was to evaluate the effects of short chain fatty acids (SCFAs) on arthritic bone remodeling. METHODS: We treated a recently described preclinical murine model of psoriatic arthritis (PsA), R26STAT3Cstopfl/fl CD4Cre mice, with SCFA-supplemented water. We also performed in vitro osteoclast differentiation assays in the presence of serum-level SCFAs to evaluate the direct impact of these microbial metabolites on maturation and function of osteoclasts. We further characterized the molecular mechanism of SCFAs by transcriptional analysis. RESULTS: The osteoporosis condition in R26STAT3Cstopfl/fl CD4Cre animals is attributed primarily to robust osteoclast differentiation driven by an expansion of osteoclast progenitor cells (OCPs), accompanied by impaired osteoblast development. We show that SCFA supplementation can rescue the osteoporosis phenotype in this model of PsA. Our in vitro experiments revealed an inhibitory effect of the SCFAs on osteoclast differentiation, even at very low serum concentrations. This suppression of osteoclast differentiation enabled SCFAs to impede osteoporosis development in R26STAT3Cstopfl/fl CD4Cre mice. Further interrogation revealed that bone marrow-derived OCPs from diseased mice expressed a higher level of SCFA receptors than those of control mice and that the progenitor cells in the bone marrow of SCFA-treated mice presented a modified transcriptomic landscape, suggesting a direct impact of SCFAs on bone marrow progenitors in the context of osteoporosis. CONCLUSION: We demonstrated how gut microbiota-derived SCFAs can regulate distal pathology (ie, osteoporosis) and identified a potential therapeutic option for restoring bone density in rheumatic disease, further highlighting the critical role of the gut-bone axis in these disorders.


Assuntos
Artrite Psoriásica , Osteoporose , Camundongos , Animais , Osteoclastos/metabolismo , Artrite Psoriásica/metabolismo , Remodelação Óssea , Diferenciação Celular , Osteoporose/metabolismo , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA