Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 658
Filtrar
1.
Front Immunol ; 14: 1186892, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37215145

RESUMO

A growing body of research suggests that short-chain fatty acids (SCFAs), metabolites produced by intestinal symbiotic bacteria that ferment dietary fibers (DFs), play a crucial role in the health status of symbiotes. SCFAs act on a variety of cell types to regulate important biological processes, including host metabolism, intestinal function, and immune function. SCFAs also affect the function and fate of immune cells. This finding provides a new concept in immune metabolism and a better understanding of the regulatory role of SCFAs in the immune system, which impacts the prevention and treatment of disease. The mechanism by which SCFAs induce or regulate the immune response is becoming increasingly clear. This review summarizes the different mechanisms through which SCFAs act in cells. According to the latest research, the regulatory role of SCFAs in the innate immune system, including in NLRP3 inflammasomes, receptors of TLR family members, neutrophils, macrophages, natural killer cells, eosinophils, basophils and innate lymphocyte subsets, is emphasized. The regulatory role of SCFAs in the adaptive immune system, including in T-cell subsets, B cells, and plasma cells, is also highlighted. In addition, we discuss the role that SCFAs play in regulating allergic airway inflammation, colitis, and osteoporosis by influencing the immune system. These findings provide evidence for determining treatment options based on metabolic regulation.


Assuntos
Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Imunidade , Ácidos Graxos Voláteis/biossíntese , Ácidos Graxos Voláteis/química , Ácidos Graxos Voláteis/metabolismo , Imunidade Inata , Humanos , Animais , Transdução de Sinais , Inflamação/imunologia , Inflamação/metabolismo
2.
Int J Food Sci Nutr ; 74(2): 219-233, 2023 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-36915255

RESUMO

We investigated the effects of (poly)phenol-rich sugarcane extract (PRSE), sugarcane fibre (SCFiber), and the combination of them (PRSE + SCFiber) on the gut microbiota and short-chain fatty acids (SCFA) production using in vitro digestion and pig faecal fermentation. Measuring total phenolic content and antioxidant activity through the in vitro digestion stages showed that PRSE + SCFiber increased the delivery of (poly)phenols to the in vitro colonic fermentation stage compared to PRSE alone. The PRSE + SCFiber modulated the faecal microbiota profile by enhancing the relative abundances of Prevotella, Lactobacillus, and Blautia, and reducing the relative abundance of Streptococcus. PRSE + SCFiber also mitigated the inhibitory effects of PRSE on SCFA production. These results suggest that the inclusion of sugarcane fibre with PRSE could increase the availability of phenolic compounds in the colon and modulate the gut microbiota towards a more favourable profile.


Assuntos
Fibras na Dieta , Fezes , Microbioma Gastrointestinal , Saccharum , Animais , Fibras na Dieta/administração & dosagem , Fibras na Dieta/análise , Fibras na Dieta/metabolismo , Digestão , Grão Comestível/química , Ácidos Graxos Voláteis/biossíntese , Fezes/química , Fezes/microbiologia , Fermentação , Suínos , Polifenóis/farmacologia , Extratos Vegetais/farmacologia , Microbioma Gastrointestinal/fisiologia
3.
Microbiol Spectr ; 10(1): e0231221, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35196806

RESUMO

Members of the gut-dwelling Bacteroides genus have remarkable abilities in degrading a diverse set of fiber polysaccharide structures, most of which are found in the mammalian diet. As part of their metabolism, they convert these fibers to organic acids that can in turn provide energy to their host. While many studies have identified and characterized the genes and corresponding proteins involved in polysaccharide degradation, relatively little is known about Bacteroides genes involved in downstream metabolic pathways. Bacteroides thetaiotaomicron is one of the most studied species from the genus and is representative of this group in producing multiple organic acids as part of its metabolism. We focused here on several organic acid synthesis pathways in B. thetaiotaomicron, including those involved in formate, lactate, propionate, and acetate production. We identified potential genes involved in each pathway and characterized these through gene deletions coupled to growth assays and organic acid quantification. In addition, we developed and employed a Golden Gate-compatible plasmid system to simplify alteration of native gene expression levels. Our work both validates and contradicts previous bioinformatic gene annotations, and we develop a model on which to base future efforts. A clearer understanding of Bacteroides metabolic pathways can inform and facilitate efforts to employ these bacteria for improved human health or other utilization strategies. IMPORTANCE Both humans and animals host a large community of bacteria and other microorganisms in their gastrointestinal tracts. This community breaks down dietary fiber and produces organic acids that are used as an energy source by the body and can also help the host resist infection by various pathogens. While the Bacteroides genus is one of the most common in the gut microbiota, it is only distantly related to bacteria with well-characterized metabolic pathways and it is therefore unclear whether research insights on organic acid production in those species can also be directly applied to the Bacteroides. By investigating multiple genetic pathways for organic acid production in Bacteroides thetaiotaomicron, we provide a basis for deeper understanding of these pathways. The work further enables greater understanding of Bacteroides-host relationships, as well as inter-species relationships in the microbiota, which are of importance for both human and animal gut health.


Assuntos
Bacteroides thetaiotaomicron/metabolismo , Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Bacteroides thetaiotaomicron/genética , Bacteroides thetaiotaomicron/isolamento & purificação , Vias Biossintéticas , Trato Gastrointestinal/microbiologia , Regulação Bacteriana da Expressão Gênica , Humanos
4.
Gastroenterology ; 162(2): 548-561.e4, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34687739

RESUMO

BACKGROUND AND AIMS: Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with altered gut microbiota composition. Phylogenetic groups of gut bacteria involved in the metabolism of short chain fatty acids (SCFAs) were depleted in SARS-CoV-2-infected patients. We aimed to characterize a functional profile of the gut microbiome in patients with COVID-19 before and after disease resolution. METHODS: We performed shotgun metagenomic sequencing on fecal samples from 66 antibiotics-naïve patients with COVID-19 and 70 non-COVID-19 controls. Serial fecal samples were collected (at up to 6 times points) during hospitalization and beyond 1 month after discharge. We assessed gut microbial pathways in association with disease severity and blood inflammatory markers. We also determined changes of microbial functions in fecal samples before and after disease resolution and validated these functions using targeted analysis of fecal metabolites. RESULTS: Compared with non-COVID-19 controls, patients with COVID-19 with severe/critical illness showed significant alterations in gut microbiome functionality (P < .001), characterized by impaired capacity of gut microbiome for SCFA and L-isoleucine biosynthesis and enhanced capacity for urea production. Impaired SCFA and L-isoleucine biosynthesis in gut microbiome persisted beyond 30 days after recovery in patients with COVID-19. Targeted analysis of fecal metabolites showed significantly lower fecal concentrations of SCFAs and L-isoleucine in patients with COVID-19 before and after disease resolution. Lack of SCFA and L-isoleucine biosynthesis significantly correlated with disease severity and increased plasma concentrations of CXCL-10, NT- proB-type natriuretic peptide, and C-reactive protein (all P < .05). CONCLUSIONS: Gut microbiome of patients with COVID-19 displayed impaired capacity for SCFA and L-isoleucine biosynthesis that persisted even after disease resolution. These 2 microbial functions correlated with host immune response underscoring the importance of gut microbial functions in SARS-CoV-2 infection pathogenesis and outcome.


Assuntos
COVID-19/microbiologia , Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal/genética , Imunidade/fisiologia , Isoleucina/biossíntese , Adulto , Biomarcadores/sangue , Estudos de Casos e Controles , Fezes/microbiologia , Feminino , Humanos , Masculino , Metagenômica , Pessoa de Meia-Idade , Filogenia , SARS-CoV-2 , Índice de Gravidade de Doença
5.
J Appl Microbiol ; 132(2): 1370-1383, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34470077

RESUMO

AIMS: Pre-eclampsia (PE) affects pregnant patients worldwide, but there is no effective treatment for this condition. We aimed to explore the effect of sodium butyrate (NaB) on PE. METHODS AND RESULTS: In this study, Nω-nitro-L-arginine methyl ester hydrochloride was used to induce PE in pregnant rats. We found that NaB significantly decreased the levels of blood pressure, 24-h protein urine and inflammatory factors (IL-1ß, IL-6 and TGF-ß), increased the foetal and placental weights and intestinal barrier markers (ZO-1, claudin-5 and occludin) expression. In addition, NaB intervention reduced the levels of soluble fms-like tyrosine kinase 1 and soluble endoglin and increased placental growth factor level. Meanwhile, after NaB treatment, the Treg/Th17 ratio of immune cells in the spleen and small intestine of pregnant rats decreased, while the level of pregnancy-related diamine oxidase increased. Notably, the PE rat treatment with NaB improved gut microbiota compositions, especially for the abundances of Firmicutes and Bacteroides, and significantly increased butyric acid and pentanoic acid levels, which might help to alleviate PE in pregnant rats. CONCLUSION: In the PE rat model, exogenous NaB improved intestinal barrier function and reduced adverse outcomes, which might be associated with the gut microbiota and its production of SCFA metabolites. SIGNIFICANCE AND IMPACT OF THE STUDY: NaB might alleviate the adverse outcomes of PE by regulating gut microbiota and its metabolite SCFA, which revealed that NaB might be a potential regulator of gut microbiota and a therapeutic substance for PE.


Assuntos
Ácido Butírico/farmacologia , Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal , Pré-Eclâmpsia , Animais , Feminino , Placenta , Fator de Crescimento Placentário , Pré-Eclâmpsia/tratamento farmacológico , Gravidez , Ratos
6.
Drug Des Devel Ther ; 15: 4849-4863, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34876807

RESUMO

Type 2 diabetes mellitus (T2DM) is a widespread metabolic disease characterized by chronic hyperglycemia. Human microbiota, which is regarded as a "hidden organ", plays an important role in the initiation and development of T2DM. In addition, anti-hyperglycemic agents and traditional Chinese medicine may affect the composition of gut microbiota and consequently improve glucose metabolism. However, the relationship between gut microbiota, T2DM and anti-hyperglycemic agents or traditional Chinese medicine is poorly understood. In this review, we summarized pre-clinical and clinical studies to elucidate the possible underlying mechanism. Some anti-hyperglycemic agents and traditional Chinese medicine may partly exert hypoglycemic effects by altering the gut microbiota composition in ways that reduce metabolic endotoxemia, maintain the integrity of intestinal mucosal barrier, promote the production of short-chain fatty acids (SCFAs), decrease trimethylamine-N-oxide (TMAO) and regulate bile acid metabolism. In conclusion, gut microbiota may provide some new therapeutic targets for treatment of patients with diabetes mellitus.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Medicamentos de Ervas Chinesas/química , Ácidos Graxos Voláteis/biossíntese , Humanos , Hipoglicemiantes/química , Medicina Tradicional Chinesa
7.
Nutrients ; 13(11)2021 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-34836151

RESUMO

Fermentation is an ancient food preservation process, and fermented products have been traditionally consumed in different cultures worldwide over the years. The interplay between human gut microbiota, diet and host health is widely recognized. Diet is one of the main factors modulating gut microbiota potentially with beneficial effects on human health. Fermented dairy products have received much attention, but other sources of probiotic delivery through food received far less attention. In this research, a combination of in vitro tools mimicking colonic fermentation and the intestinal epithelium have been applied to study the effect of different pasteurized and non-pasteurized water kefir products on gut microbiota, epithelial barrier function and immunomodulation. Water kefir increased beneficial short-chain fatty acid production at the microbial level, reduced detrimental proteolytic fermentation compounds and increased Bifidobacterium genus abundance. The observed benefits are enhanced by pasteurization. Pasteurized products also had a significant effect at the host level, improving inflammation-induced intestinal epithelial barrier disruption and increasing IL-10 and IL-1ß compared to the control condition. Our data support the potential health benefits of water kefir and demonstrate that pasteurization, performed to prolong shelf life and stability of the product, also enhanced these benefits.


Assuntos
Bebidas/análise , Citocinas/biossíntese , Microbioma Gastrointestinal , Kefir , Água/farmacologia , Colo/metabolismo , Colo/microbiologia , Ácidos Graxos Voláteis/biossíntese , Fermentação , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Pasteurização , Permeabilidade
8.
Int J Mol Sci ; 22(22)2021 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-34830266

RESUMO

Lactoferrin is a glycoprotein found at high concentrations within exocrine secretions, including tears. Low levels of lactoferrin have been implicated in the loss of tear secretion and ageing. Furthermore, lactoferrin possesses a range of functionalities, including anti-inflammatory properties and the ability to modulate the gut microbiota. Expanding evidence demonstrates a crucial role of the gut microbiota in immune regulation and development. The specific composition of bacterial species of the gut has a profound influence on local and systemic inflammation, leading to a protective capacity against a number of inflammatory diseases, potentially by the induction of regulatory immune cells. In this study, we demonstrated that oral administration of lactoferrin maintains tear secretion in a restraint and desiccating stress induced mouse model of dry eye disease. Furthermore, we revealed that lactoferrin induces the reduction of inflammatory cytokines, modulates gut microbiota, and induces short-chain fatty acid production. Whereas, the antibiotic vancomycin abrogates the effects of lactoferrin on dry eye disease and significantly reduces short-chain fatty acid concentrations. Therefore, this protective effect of LF against a mice model of DED may be explained by our observations of an altered gut microbiota and an enhanced production of immunomodulatory short-chain fatty acids.


Assuntos
Síndromes do Olho Seco/tratamento farmacológico , Síndromes do Olho Seco/metabolismo , Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal/efeitos dos fármacos , Lactoferrina/administração & dosagem , Substâncias Protetoras/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Administração Oral , Animais , Antibacterianos/administração & dosagem , Citocinas/metabolismo , DNA Bacteriano/genética , DNA Bacteriano/isolamento & purificação , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal/genética , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Lágrimas/metabolismo , Resultado do Tratamento , Vancomicina/administração & dosagem
9.
FASEB J ; 35(11): e21937, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34606628

RESUMO

Defective permeability barrier is considered to be an incentive of hyperuricemia, however, the link between them has not been proven. Here, we evaluated the potential preventive effects of Lactiplantibacillus plantarum N-1 (LPN1) on gut microbiota and intestinal barrier function in rats with hyperoxaluria-induced kidney stones. Male rats were supplied with 1% ethylene glycol (EG) dissolved in drinking water for 4 weeks to develop hyperoxaluria, and some of them were administered with LPN1 for 4 weeks before EG treatment as a preventive intervention. We found that EG not only resulted hyperoxaluria and kidney stone formation, but also promoted the intestinal inflammation, elevated intestinal permeability, and gut microbiota disorders. Supplementation of LPN1 inhibited the renal crystalline deposits through reducing urinary oxalic acid and renal osteopontin and CD44 expression and improved EG-induced intestinal inflammation and barrier function by decreasing the serum LPS and TLR4/NF-κB signaling and up-regulating tight junction Claudin-2 in the colon, as well as increasing the production of short-chain fatty acid (SCFAs) and the abundance of beneficial SCFAs-producing bacteria, mainly from the families of Lachnospiraceae and Ruminococcaceae. Probiotic LPN1 could prevent EG-induced hyperoxaluria by regulating gut microbiota and enhancing intestinal barrier function.


Assuntos
Etilenoglicol/efeitos adversos , Microbioma Gastrointestinal/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Cálculos Renais/induzido quimicamente , Cálculos Renais/prevenção & controle , Lactobacillaceae , Permeabilidade , Probióticos/administração & dosagem , Animais , Colo/metabolismo , Colo/microbiologia , Ácidos Graxos Voláteis/análise , Ácidos Graxos Voláteis/biossíntese , Fezes/química , Fezes/microbiologia , Hiperoxalúria/induzido quimicamente , Hiperoxalúria/prevenção & controle , Hiperuricemia/induzido quimicamente , Hiperuricemia/prevenção & controle , Inflamação/metabolismo , Masculino , RNA Ribossômico 16S/genética , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Junções Íntimas/metabolismo
10.
Microbiol Spectr ; 9(2): e0018221, 2021 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-34612695

RESUMO

Seasonal influenza outbreaks represent a large burden for the health care system as well as the economy. While the role of the microbiome has been elucidated in the context of various diseases, the impact of respiratory viral infections on the human microbiome is largely unknown. In this study, swine was used as an animal model to characterize the temporal dynamics of the respiratory and gastrointestinal microbiome in response to an influenza A virus (IAV) infection. A multi-omics approach was applied on fecal samples to identify alterations in microbiome composition and function during IAV infection. We observed significantly altered microbial richness and diversity in the gastrointestinal microbiome after IAV infection. In particular, increased abundances of Prevotellaceae were detected, while Clostridiaceae and Lachnospiraceae decreased. Moreover, our metaproteomics data indicated that the functional composition of the microbiome was heavily affected by the influenza infection. For instance, we identified decreased amounts of flagellin, correlating with reduced abundances of Lachnospiraceae and Clostridiaceae, possibly indicating involvement of a direct immune response toward flagellated Clostridia during IAV infection. Furthermore, enzymes involved in short-chain fatty acid (SCFA) synthesis were identified in higher abundances, while metabolome analyses revealed rather stable concentrations of SCFAs. In addition, 16S rRNA gene sequencing was used to characterize effects on the composition and natural development of the upper respiratory tract microbiome. Our results showed that IAV infection resulted in significant changes in the abundance of Moraxellaceae and Pasteurellaceae in the upper respiratory tract. Surprisingly, temporal development of the respiratory microbiome structure was not affected. IMPORTANCE Here, we used swine as a biomedical model to elucidate the impact of influenza A H1N1 infection on structure and function of the respiratory and gastrointestinal tract microbiome by employing a multi-omics analytical approach. To our knowledge, this is the first study to investigate the temporal development of the porcine microbiome and to provide insights into the functional capacity of the gastrointestinal microbiome during influenza A virus infection.


Assuntos
Bactérias/classificação , Bactérias/isolamento & purificação , Microbioma Gastrointestinal/fisiologia , Infecções por Orthomyxoviridae/patologia , Sistema Respiratório/microbiologia , Animais , Bactérias/genética , Modelos Animais de Doenças , Ácidos Graxos Voláteis/biossíntese , Fezes/microbiologia , Feminino , Perfilação da Expressão Gênica , Vírus da Influenza A Subtipo H1N1/patogenicidade , Masculino , Proteômica , RNA Ribossômico 16S/genética , Suínos
11.
Benef Microbes ; 12(6): 601-612, 2021 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-34590532

RESUMO

The gut microbiota has been indicated to play a crucial role in health and disease. Apart from changes in composition between healthy individuals and those with a disease or disorder, it has become clear that also microbial activity is important for health. For instance, butyrate has been proven to be beneficial for health, because, amongst others, it is a substrate for the colonocytes, and modulates the host's immune system and metabolism. Here, we studied the effect of a blend of three mushrooms (Ganoderma lucidum GL AM P-38, Grifola frondosa GF AM P36 and Pleurotus ostreatus PO AM-GP37)) on gut microbiota composition and activity in a validated, dynamic, computer-controlled in vitro model of the colon (TIM-2). Predigested mushroom blend at three doses (0.5, 1.0 and 1.5 g/day of ingested mushroom blend) was fed to a pooled microbiota of healthy adults for 72 h, and samples were taken every day for microbiota composition (sequencing of amplicons of the V3-V4 region of the 16S rRNA gene) and activity (short-chain fatty acid (SCFA) production). The butyrate producing genera Lachnospiraceae UCG-004, Lachnoclostridium, Ruminococcaceae UCG-002 and Ruminococcaceae NK4A214-group are all dose-dependently increased when the mushroom blend was fed. Entirely in line with the increase of these butyrate-producers, the cumulative amount of butyrate also dose-dependently increased, to roughly twice the amount compared to the control (medium without mushroom blend) on the high-dose mushroom blend. Butyrate proportionally made up 53.1% of the total SCFA upon feeding the high-dose mushroom blend, compared to 27% on the control medium. In conclusion, the (polysaccharides in the) mushroom blend led to substantial increase in butyrate by the gut microbiota. These results warrant future mechanistic research on the mushroom blend, as butyrate is considered to be one of the microbial metabolites that contributes to health, by increasing barrier function and modulating inflammation.


Assuntos
Agaricales , Produtos Biológicos/farmacologia , Butiratos/metabolismo , Microbioma Gastrointestinal , Adulto , Agaricales/química , Ácidos Graxos Voláteis/biossíntese , Grifola , Humanos , Pleurotus , RNA Ribossômico 16S/genética , Reishi
12.
Nutrients ; 13(9)2021 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-34579126

RESUMO

The human gut microbiota has been linked to the health status of the host. Modulation of human gut microbiota through pro- and prebiotic interventions has yielded promising results; however, the effect of novel prebiotics, such as chitin-glucan, on gut microbiota-host interplay is still not fully characterized. We assessed the effect of chitin-glucan (CG) and chitin-glucan plus Bifidobacterium breve (CGB) on human gut microbiota from the luminal and mucosal environments in vitro. Further, we tested the effect of filter-sterilized fecal supernatants from CG and CGB fermentation for protective effects on inflammation-induced barrier disruption and cytokine production using a co-culture of enterocytes and macrophage-like cells. Overall, CG and CGB promote health-beneficial short-chain fatty acid production and shift human gut microbiota composition, with a consistent effect increasing Roseburia spp. and butyrate producing-bacteria. In two of three donors, CG and CGB also stimulated Faecalibacterium prausniitzi. Specific colonization of B. breve was observed in the lumen and mucosal compartment; however, no synergy was detected for different endpoints when comparing CGB and CG. Both treatments included a significant improvement of inflammation-disrupted epithelial barrier and shifts on cytokine production, especially by consistent increase in the immunomodulatory cytokines IL10 and IL6.


Assuntos
Quitina/farmacologia , Citocinas/biossíntese , Microbioma Gastrointestinal/efeitos dos fármacos , Glucanos/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Prebióticos/administração & dosagem , Bifidobacterium breve/fisiologia , Células CACO-2 , Técnicas de Cocultura , Enterócitos , Ácidos Graxos Voláteis/biossíntese , Fezes/microbiologia , Fermentação , Microbioma Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/fisiologia , Probióticos/administração & dosagem , Células THP-1
13.
Int J Mol Sci ; 22(12)2021 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-34204294

RESUMO

Persistent infection with High Risk-Human Papilloma Viruses (HR-HPVs) is a primary cause of cervical cancer worldwide. Vaginal-dysbiosis-associated bacteria were correlated with the persistence of HR-HPVs infection and with increased cancer risk. We obtained strains of the most represented bacterial species in vaginal microbiota and evaluated their effects on the survival of cervical epithelial cells and immune homeostasis. The contribution of each species to supporting the antiviral response was also studied. Epithelial cell viability was affected by culture supernatants of most vaginal-dysbiosis bacteria, whereas Lactobacillus gasseri or Lactobacillus jensenii resulted in the best stimulus to induce interferon-γ (IFN-γ) production by human mononuclear cells from peripheral blood (PBMCs). Although vaginal-dysbiosis-associated bacteria induced the IFN-γ production, they were also optimal stimuli to interleukin-17 (IL-17) production. A positive correlation between IL-17 and IFN-γ secretion was observed in cultures of PBMCs with all vaginal-dysbiosis-associated bacteria suggesting that the adaptive immune response induced by these strains is not dominated by TH1 differentiation with reduced availability of IFN-γ, cytokine most effective in supporting virus clearance. Based on these results, we suggest that a vaginal microbiota dominated by lactobacilli, especially by L. gasseri or L. jensenii, may be able to assist immune cells with clearing HPV infection, bypasses the viral escape and restores immune homeostasis.


Assuntos
Antibiose , Disbiose , Homeostase , Lactobacillus/fisiologia , Mucosa/imunologia , Mucosa/microbiologia , Vagina/imunologia , Vagina/microbiologia , Sobrevivência Celular , Citocinas/biossíntese , Células Epiteliais/metabolismo , Ácidos Graxos Voláteis/biossíntese , Feminino , Humanos , Vagina/metabolismo
14.
PLoS One ; 16(7): e0254004, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34288919

RESUMO

The human gut microbiota is known for its highly heterogeneous composition across different individuals. However, relatively little is known about functional differences in its ability to ferment complex polysaccharides. Through ex vivo measurements from healthy human donors, we show that individuals vary markedly in their microbial metabolic phenotypes (MMPs), mirroring differences in their microbiota composition, and resulting in the production of different quantities and proportions of Short Chain Fatty Acids (SCFAs) from the same inputs. We also show that aspects of these MMPs can be predicted from composition using 16S rRNA sequencing. From experiments performed using the same dietary fibers in vivo, we demonstrate that an ingested bolus of fiber is almost entirely consumed by the microbiota upon passage. We leverage our ex vivo data to construct a model of SCFA production and absorption in vivo, and argue that inter-individual differences in quantities of absorbed SCFA are directly related to differences in production. Though in vivo studies are required to confirm these data in the context of the gut, in addition to in vivo read outs of SCFAs produced in response to specific fiber spike-ins, these data suggest that optimizing SCFA production in a given individual through targeted fiber supplementation requires quantitative understanding of their MMP.


Assuntos
Fibras na Dieta/metabolismo , Ácidos Graxos Voláteis/biossíntese , Fermentação , Microbioma Gastrointestinal/fisiologia , Adulto , Bactérias/genética , Bactérias/isolamento & purificação , Bactérias/metabolismo , Variação Biológica Individual , Carboidratos da Dieta/metabolismo , Fezes/microbiologia , Feminino , Seguimentos , Humanos , Absorção Intestinal , Inulina/análise , Aprendizado de Máquina , Masculino , Fenótipo , Polissacarídeos/metabolismo , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Ribotipagem , Adulto Jovem
15.
Food Chem ; 361: 130095, 2021 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-34091400

RESUMO

Highly resistant starch rice (HRSR) is of particular interest in terms of its capacity to deliver short-chain fatty acids (SCFAs) to the colon in the prevention of diabetes mellitus and obesity. In this study, HRSR was processed into cooked rice, rice milk, rice cake, and rice popcorn, and the in vitro digestion and fermentation processes were monitored. The results showed that the starch digestibility of the four samples conformed to a first-order two-phase equation, and the resistant starch content of rice cake was the highest (11.98%). Compared with inulin, rice cake had a slower fermentation rate, and the butyrate concentration increased by 67.85%. The abundances of Prevotellaceae, which promotes the synthesis of SCFAs, and anti-inflammatory Faecalibacterium increased. The abundances of Proteobacteria and Megamonas, markers of gut microbiota imbalance, decreased. The results might facilitate the design and production of functional food products for type 2 diabetic and obese patients and improving colonic health.


Assuntos
Digestão , Fezes , Fermentação , Microbioma Gastrointestinal/efeitos dos fármacos , Oryza/química , Oryza/metabolismo , Amido Resistente/análise , Ácidos Graxos Voláteis/biossíntese , Humanos
16.
Int J Biol Macromol ; 182: 1351-1361, 2021 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-34000312

RESUMO

The aim of this study was to investigate and compare the structural and physicochemical properties of native Banxia starch (BXS) and autoclaving-cooling treated Banxia starch (CTBXS) and its related impacts on production of short chain fatty acids (SCFAs) and human gut microbiota by in vitro fecal fermentation. BXS had semicircle to spherical granules, whereas CTBXS exhibited block-shape. According to XRD and TGA, BXS had a C-type crystalline pattern, while CTBXS had a B-type crystalline pattern. CTBXS had better thermal stability than BXS. In addition, BXS exhibited significantly higher solubility and swelling power than CTBXS, and CTBXS had higher content of SDS than BXS. Moreover, BXS and CTBXS could change the composition and abundance of gut microbiota, could also promote the production of SCFAs. This study is beneficial to well understand the in vitro digestion and fecal fermentation behaviors of BXS and CTBXS, and can be developed as a potential functional food with the aim of improving colonic health.


Assuntos
Temperatura Baixa , Microbioma Gastrointestinal , Pinellia/química , Amido/química , Bactérias/classificação , Cristalização , Ácidos Graxos Voláteis/biossíntese , Fezes , Fermentação , Filogenia , RNA Ribossômico 16S/genética , Solubilidade , Espectroscopia de Infravermelho com Transformada de Fourier , Termogravimetria
17.
Nutrients ; 13(5)2021 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-33922631

RESUMO

Glycerol monocaprylate (GMC) is a glycerol derivative of medium-chain fatty acids (MCFAs) and is widely used as a preservative in food processing. However, GMC and its hydrolytic acid (octylic acid) have antibacterial properties that may affect the physiology and intestinal microecology of the human body. Therefore, in this study, the effects of two different dosages of GMC (150 and 1600 mg kg-1) on glucose, lipid metabolism, inflammation, and intestinal microecology of normal diet-fed C57BL/6 mice were comprehensively investigated. The obtained results showed that the level of triglycerides (TGs) in the low-dose group down-regulated significantly, and the anti-inflammatory cytokine interleukin 10 (IL-10) significantly increased, while the pro-inflammatory cytokines monocyte chemotactic protein 1 (MCP-1) and interleukin 1beta (IL-1ß) in the high-dose group were significantly decreased. Importantly, GMC promoted the α-diversity of gut microbiota in normal-diet-fed mice, regardless of dosages. Additionally, it was found that the low-dose treatment of GMC significantly increased the abundance of Lactobacillus, while the high-dose treatment of GMC significantly increased the abundance of SCFA-producers such as Clostridiales, Lachnospiraceae, and Ruminococcus. Moreover, the content of short-chain fatty acids (SCFAs) was significantly increased by GMC supplementation. Thus, our research provides a novel insight into the effects of GMC on gut microbiota and physiological characteristics.


Assuntos
Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal/efeitos dos fármacos , Glicerol/farmacologia , Inflamação/microbiologia , Metabolismo/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Tamanho Celular/efeitos dos fármacos , Citocinas/sangue , Comportamento Alimentar/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Hormônios/sangue , Inflamação/sangue , Inflamação/genética , Inflamação/patologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL
18.
Int J Biol Sci ; 17(5): 1302-1314, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33867847

RESUMO

Aurora-A kinase, a serine/threonine mitotic kinase involved in mitosis, is overexpressed in several human cancers. A recent study showed that Aurora-A mediates glucose metabolism via SOX8/FOXK1 in ovarian cancer. However, the roles of Aurora-A in metabolic diseases remain unclear. This study found that Aurka loss in the intestinal epithelium promoted age-induced obesity and enlargement of lipid droplets in parallel with an increase in infiltrated macrophages in the white adipocyte tissue (WAT) of male mice. Moreover, loss of Aurka induced the expression of lipid metabolism regulatory genes, including acetyl-coenzyme A carboxylase 1 (Acc1), in association with an increase in the levels of p-AKT in the intestinal epithelium as well as WAT. Blockade of AKT activation reduced the expression of lipid metabolism regulatory genes. In subsequent experiments, we found that the Firmicutes abundance and the levels of short-chain fatty acids (SCFAs) in the gut were dramatically increased in Aurkaf/+;VillinCre/+ mice compared with Aurkaf/+ mice. Additionally, propionate increased the phosphorylation of AKT in vitro. These observations indicated that Aurka loss in the intestinal epithelium contributed to gut microbiota dysbiosis and higher levels of SCFAs, especially propionate, leading to AKT activation and lipid metabolism regulatory gene expression, which in turn promoted age-induced obesity.


Assuntos
Acetil-CoA Carboxilase/metabolismo , Envelhecimento/metabolismo , Aurora Quinase A/metabolismo , Disbiose , Mucosa Intestinal , Obesidade/metabolismo , Propionatos/metabolismo , Tecido Adiposo/metabolismo , Tecido Adiposo/patologia , Animais , Disbiose/metabolismo , Disbiose/microbiologia , Ativação Enzimática , Ácidos Graxos Voláteis/biossíntese , Microbioma Gastrointestinal/fisiologia , Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Metabolismo dos Lipídeos/genética , Macrófagos/metabolismo , Macrófagos/patologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo
19.
Nutrients ; 13(4)2021 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-33920564

RESUMO

Intestinal health relies on the association between the mucosal immune system, intestinal barrier and gut microbiota. Bioactive components that affect the gut microbiota composition, epithelial physical barrier and intestinal morphology were previously studied. The current systematic review evaluated evidence of anthocyanin effects and the ability to improve gut microbiota composition, their metabolites and parameters of the physical barrier; this was conducted in order to answer the question: "Does food source or extract of anthocyanin promote changes on intestinal parameters?". The data analysis was conducted following the PRISMA guidelines with the search performed at PubMed, Cochrane and Scopus databases for experimental studies, and the risk of bias was assessed by the SYRCLE tool. Twenty-seven studies performed in animal models were included, and evaluated for limitations in heterogeneity, methodologies, absence of information regarding allocation process and investigators' blinding. The data were analyzed, and the anthocyanin supplementation demonstrated positive effects on intestinal health. The main results identified were an increase of Bacteroidetes and a decrease of Firmicutes, an increase of short chain fatty acids production, a decrease of intestinal pH and intestinal permeability, an increase of the number of goblet cells and tight junction proteins and villi improvement in length or height. Thus, the anthocyanin supplementation has a potential effect to improve the intestinal health. PROSPERO (CRD42020204835).


Assuntos
Antocianinas/farmacocinética , Suplementos Nutricionais , Microbioma Gastrointestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Intestinos/efeitos dos fármacos , Bacteroidetes/metabolismo , Disponibilidade Biológica , Ácidos Graxos Voláteis/biossíntese , Firmicutes/metabolismo , Células Caliciformes/metabolismo , Humanos , Microvilosidades/efeitos dos fármacos , Permeabilidade/efeitos dos fármacos
20.
Food Funct ; 12(7): 2886-2900, 2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33877244

RESUMO

The effects of arabinoxylan (AX)-rich rye bran based diet (RB) and antibiotics on digestion, fermentation and short-chain fatty acids (SCFA) absorption were studied compared with an iso-dietary fibre (DF) cellulose based diet (CEL). Thirty female pigs (body weight 72.5 ± 3.9 kg) were fed a standard swine diet in week 1, CEL as wash-out for bran-associated bioactive components in week 2 and then divided into 3 groups fed either the CEL (n = 10) or RB (n = 20) for 2 weeks, where 10 pigs from RB had daily intramuscular antibiotic injections (RB+) and the other 10 pigs were untreated (RB-) in week 4. In RB, the degradation of AX mainly occurred in caecum and proximal colon (P < 0.01) and to a higher extent than cellulose, which on the other hand, irrespective of antibiotic treatment, was less degraded in the RB groups than in the CEL (P < 0.01). The apparent digestibility of fat and protein in the distal small intestine was lower for RB than CEL (P < 0.05), the protein digestibility remained lower in most of the colon, and the digestibility was not affected by treatment with antibiotics. The colonic concentrations of SCFA, acetate and propionate as well as the butyrate concentration in the distal colon were lower with the RB treatments compared with CEL (P < 0.01). Caecal butyrate concentrations were on the other hand higher, and a significant reduction was seen with antibiotic treatment (P < 0.001). The daily net absorption of SCFA and acetate was lower with RB than with CEL (P < 0.01). In conclusion, RB resulted in different DF degradation processes and SCFA production compared with CEL, whereas antibiotic treatment had marginal effects on the intestinal DF degradation but hampered butyrate production.


Assuntos
Antibacterianos/farmacologia , Fibras na Dieta/administração & dosagem , Digestão/efeitos dos fármacos , Ácidos Graxos Voláteis/farmacocinética , Fermentação/efeitos dos fármacos , Secale , Ração Animal , Animais , Butiratos/metabolismo , Celulose/administração & dosagem , Dieta , Ácidos Graxos Voláteis/biossíntese , Feminino , Absorção Intestinal/efeitos dos fármacos , Sus scrofa , Xilanos/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...