Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
Microb Biotechnol ; 17(6): e14485, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38850270

RESUMO

Proanthocyanidin-rich grape seed extract (GSE) has been shown to have the potential to protect bones, although the underlying mechanism remains unknown. The current study aims to explore GSE's preventive and therapeutic impact on bone loss induced by oestrogen deficiency and the underlying mechanism through the gut microbiota (GM) and metabolomic responses. In oestrogen-deficient ovariectomized (OVX) mice, GSE ameliorated bone loss by inhibiting the expansion of bone marrow adipose tissue (BMAT), restoring BMAT lipolysis and promoting bone formation. GSE regulated OVX-induced GM dysbiosis by reducing the abundance of opportunistic pathogenic bacteria, such as Alistipes, Turicibacter and Romboutsia, while elevating the abundance of beneficial bacteria, such as Bifidobacterium. The modified GM primarily impacted lipid and amino acid metabolism. Furthermore, the serum metabolites of GSE exhibited a significant enrichment in lipid metabolism. In summary, GSE shows potential as a functional food for preventing oestrogen deficiency-induced bone loss by modulating GM and metabolite-mediated lipid metabolism.


Assuntos
Estrogênios , Microbioma Gastrointestinal , Extrato de Sementes de Uva , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Extrato de Sementes de Uva/farmacologia , Camundongos , Feminino , Estrogênios/deficiência , Estrogênios/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Disbiose/prevenção & controle , Camundongos Endogâmicos C57BL , Bactérias/metabolismo , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Osteoporose/prevenção & controle , Modelos Animais de Doenças , Tecido Adiposo/metabolismo , Ovariectomia
2.
Nutrients ; 15(11)2023 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-37299553

RESUMO

Pecans (Carya illinoinensis) are considered a functional food due to the high content of polyunsaturated fatty acids, dietary fiber and polyphenols. To determine the effect of whole pecans (WP) or a pecan polyphenol (PP) extract on the development of metabolic abnormalities in mice fed a high-fat (HF) diet, we fed C57BL/6 mice with a Control diet (7% fat), HF diet (23% fat), HF containing 30% WP or an HF diet supplemented with 3.6 or 6 mg/g of PP for 18 weeks. Supplementation of an HF diet with WP or PP reduced fat mass, serum cholesterol, insulin and HOMA-IR by 44, 40, 74 and 91%, respectively, compared to the HF diet. They also enhanced glucose tolerance by 37%, prevented pancreatic islet hypertrophy, and increased oxygen consumption by 27% compared to the HF diet. These beneficial effects were associated with increased thermogenic activity in brown adipose tissue, mitochondrial activity and AMPK activation in skeletal muscle, reduced hypertrophy and macrophage infiltration of subcutaneous and visceral adipocytes, reduced hepatic lipid content and enhanced metabolic signaling. Moreover, the microbial diversity of mice fed WP or PP was higher than those fed HF, and associated with lower circulating lipopolysaccharides (~83-95%). Additionally, a 4-week intervention study with the HF 6PP diet reduced the metabolic abnormalities of obese mice. The present study demonstrates that WP or a PP extract prevented obesity, liver steatosis and diabetes by reducing dysbiosis, inflammation, and increasing mitochondrial content and energy expenditure. Pecan polyphenols were mainly condensed tannin and ellagic acid derivatives including ellagitannins as determined by LC-MS. Herein we also propose a model for the progression of the HF diet-mediated metabolic disorder based on early and late events, and the possible molecular targets of WP and PP extract in preventive and intervention strategies. The body surface area normalization equation gave a conversion equivalent to a daily human intake dose of 2101-3502 mg phenolics that can be obtained from 110-183 g pecan kernels/day (22-38 whole pecans) or 21.6-36 g defatted pecan flour/day for an average person of 60 kg. This work lays the groundwork for future clinical studies.


Assuntos
Carya , Diabetes Mellitus , Fígado Gorduroso , Camundongos , Humanos , Animais , Dieta Hiperlipídica/efeitos adversos , Polifenóis/farmacologia , Polifenóis/metabolismo , Disbiose/prevenção & controle , Disbiose/metabolismo , Camundongos Endogâmicos C57BL , Obesidade/etiologia , Obesidade/prevenção & controle , Fígado Gorduroso/prevenção & controle , Fígado/metabolismo , Inflamação/prevenção & controle , Inflamação/metabolismo , Diabetes Mellitus/metabolismo , Hipertrofia , Metabolismo Energético
3.
ACS Appl Mater Interfaces ; 15(16): 19833-19846, 2023 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-37052616

RESUMO

Radiation gastroenteritis represents one of the most prevalent and hazardous complications of abdominopelvic radiotherapy, which not only severely reduces patients' life quality but also restricts radiotherapy efficacy. However, there is currently no clinically available oral radioprotector for this threatening disease due to its complex pathogenesis and the harsh gastrointestinal environment. To this end, this study developed a facile but effective oral radioprotector, ergothioneine hyaluronate (EGT@HA) gel, protecting against radiation gastroenteritis by synergistically regulating oxidative stress, inflammation, and gut microbiota. In vitro and cellular experiments verified the chemical stability and free radical scavenging ability of EGT and its favorable cellular radioprotective efficacy by inhibiting intracellular reactive oxidative species (ROS) generation, DNA damage, mitochondrial damage, and apoptosis. At the in vivo level, EGT@HA with prolonged gastrointestinal residence mitigated radiation-induced gastrointestinal tissue injury, apoptosis, neutrophil infiltration, and gut flora dysbiosis. For the first time, this work investigated the protective effects of EGT@HA gel on radiation gastroenteritis, which not only hastens the advancement of the novel gastrointestinal radioprotector but also provides a valuable gastrointestinal radioprotection paradigm by synergistically modulating oxidative stress, inflammation, and gut microbiota disturbance.


Assuntos
Ergotioneína , Gastroenterite , Microbioma Gastrointestinal , Lesões por Radiação , Humanos , Ergotioneína/genética , Ergotioneína/farmacologia , Antioxidantes/farmacologia , Disbiose/tratamento farmacológico , Disbiose/prevenção & controle , Apoptose , Inflamação/tratamento farmacológico , Inflamação/prevenção & controle
4.
Crit Rev Food Sci Nutr ; 63(21): 5206-5230, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34991393

RESUMO

The prevalence of inflammatory bowel disease (IBD) is increasing, which is concerning because IBD is a known risk factor for the development of colorectal cancer. Emerging evidence highlights environmental factors, particularly dietary factors and gut microbiota dysbiosis, as pivotal inducers of IBD onset. Goji berry, an ancient tonic food and a nutraceutical supplement, contains a range of phytochemicals such as polysaccharides, carotenoids, and polyphenols. Among these phytochemicals, L. barbarum polysaccharides (LBPs) are the most important functional constituents, which have protective effects against oxidative stress, inflammation, and neurodegeneration. Recently, the beneficial effects of goji berry and associated LBPs consumption were linked to prebiotic effects, which can prevent dysbiosis associated with IBD. This review assessed pertinent literature on the protective effects of goji berry against IBD focusing on the gut microbiota and their metabolites in mediating the observed beneficial effects.


Assuntos
Doenças Inflamatórias Intestinais , Lycium , Humanos , Prebióticos , Disbiose/prevenção & controle , Doenças Inflamatórias Intestinais/tratamento farmacológico , Doenças Inflamatórias Intestinais/prevenção & controle , Polissacarídeos/farmacologia , Compostos Fitoquímicos/farmacologia
5.
Front Cell Infect Microbiol ; 12: 969526, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36051242

RESUMO

The gut dysbiosis has emerged as a prominent player in the pathogenesis and development of colorectal cancer (CRC), which in turn intensifies dysregulated gut microbiota composition and inflammation. Since most drugs are given orally, this dysbiosis directly and indirectly impinges the absorption and metabolism of drugs in the gastrointestinal tract, and subsequently affects the clinical outcome of patients with CRC. Herbal medicine, including the natural bioactive products, have been used traditionally for centuries and can be considered as novel medicinal sources for anticancer drug discovery. Due to their various structures and pharmacological effects, natural products have been found to improve microbiota composition, repair intestinal barrier and reduce inflammation in human and animal models of CRC. This review summarizes the chemo-preventive effects of extracts and/or compounds derived from natural herbs as the promising antineoplastic agents against CRC, and will provide innovative strategies to counteract dysregulated microbiota and improve the lives of CRC patients.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Animais , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Disbiose/prevenção & controle , Medicina Herbária , Humanos , Inflamação
6.
Pharmacol Res ; 177: 106115, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35124207

RESUMO

The bidirectional interaction between carcinogens and gut microbiota that contributes to colorectal cancer is complicated. Reactivation of carcinogen metabolites by microbial ß-glucuronidase (ßG) in the gut potentially plays an important role in colorectal carcinogenesis. We assessed the chemoprotective effects and associated changes in gut microbiota induced by pre-administration of bacterial-specific ßG inhibitor TCH-3511 in carcinogen azoxymethane (AOM)-treated APCMin/+ mice. AOM induced intestinal ßG activity, which was reflected in increases in the incidence, formation, and number of tumors in the intestine. Notably, inhibition of gut microbial ßG by TCH-3511 significantly reduced AOM-induced intestinal ßG activity, decreased the number of polyps in both the small and large intestine to a frequency that was similar in mice without AOM exposure. AOM also led to lower diversity and altered composition in the gut microbiota with a significant increase in mucin-degrading Akkermansia genus. Conversely, mice treated with TCH-3511 and AOM exhibited a more similar gut microbiota structure as mice without AOM administration. Importantly, TCH-3511 treatment significant decreased Akkermansia genus and produced a concomitant increase in short-chain fatty acid butyrate-producing gut commensal microbes Lachnoospiraceae NK4A136 group genus in AOM-treated mice. Taken together, our results reveal a key role of gut microbial ßG in promoting AOM-induced gut microbial dysbiosis and intestinal tumorigenesis, indicating the chemoprotective benefit of gut microbial ßG inhibition against carcinogens via maintaining the gut microbiota balance and preventing cancer-associated gut microbial dysbiosis. Thus, the bacterial-specific ßG inhibitor TCH-3511 is a potential chemoprevention agent for colorectal cancer.


Assuntos
Neoplasias Colorretais , Microbioma Gastrointestinal , Animais , Azoximetano/toxicidade , Bactérias , Carcinogênese , Carcinógenos/toxicidade , Transformação Celular Neoplásica , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/prevenção & controle , Disbiose/prevenção & controle , Glucuronidase , Camundongos
7.
Oxid Med Cell Longev ; 2021: 6221012, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34950418

RESUMO

Antioxidant polyphenols from plants are potential dietary supplementation to alleviate early weaning-induced intestinal disorders in piglets. Recent evidences showed polyphenol quercetin could reshape gut microbiota when it functioned as anti-inflammation or antioxidation agents in rodent models. However, the effect of dietary quercetin supplementation on intestinal disorders and gut microbiota of weanling piglets, along with the role of gut microbiota in this effect, both remain unclear. Here, we determined the quercetin's effect on attenuating diarrhea, intestinal damage, and redox imbalance, as well as the role of gut microbiota by transferring the quercetin-treated fecal microbiota to the recipient piglets. The results showed that dietary quercetin supplementation decreased piglets' fecal scores improved intestinal damage by increasing tight junction protein occludin, villus height, and villus height/crypt depth ratio but decreased crypt depth and intestinal epithelial apoptosis (TUNEL staining). Quercetin also increased antioxidant capacity indices, including total antioxidant capacity, catalase, and glutathione/oxidized glutathione disulfide but decreased oxidative metabolite malondialdehyde in the jejunum tissue. Fecal microbiota transplantation (FMT) from quercetin-treated piglets had comparable effects on improving intestinal damage and antioxidative capacity than dietary quercetin supplementation. Further analysis of gut microbiota using 16S rDNA sequencing showed that dietary quercetin supplementation or FMT shifted the structure and increased the diversity of gut microbiota. Especially, anaerobic trait and carbohydrate metabolism functions of gut microbiota were enriched after dietary quercetin supplementation and FMT, which may owe to the increased antioxidative capacity of intestine. Quercetin increased the relative abundances of Fibrobacteres, Akkermansia muciniphila, Clostridium butyricum, Clostridium celatum, and Prevotella copri but decreased the relative abundances of Proteobacteria, Lactobacillus coleohominis, and Ruminococcus bromii. Besides, quercetin-shifted bacteria and carbohydrate metabolites short chain fatty acids were significantly related to the indices of antioxidant capacity and intestinal integrity. Overall, dietary quercetin supplementation attenuated diarrhea and intestinal damage by enhancing the antioxidant capacity and regulating gut microbial structure and metabolism in piglets.


Assuntos
Diarreia/prevenção & controle , Suplementos Nutricionais , Disbiose/prevenção & controle , Transplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Enteropatias/prevenção & controle , Quercetina/administração & dosagem , Ração Animal/análise , Animais , Antioxidantes/administração & dosagem , Bactérias/classificação , Bactérias/crescimento & desenvolvimento , Diarreia/microbiologia , Diarreia/patologia , Disbiose/microbiologia , Disbiose/patologia , Feminino , Enteropatias/microbiologia , Enteropatias/patologia , Suínos , Desmame
8.
Front Immunol ; 12: 756920, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34646279

RESUMO

High glucose and fructose intake have been proven to display pro-inflammatory roles during the progression of inflammatory diseases. However, mannose has been shown to be a special type of hexose that has immune regulatory functions. In this review, we trace the discovery process of the regulatory functions of mannose and summarize some past and recent studies showing the therapeutic functions of mannose in inflammatory diseases. We conclude that treatment with mannose can suppress inflammation by inducing regulatory T cells, suppressing effector T cells and inflammatory macrophages, and increasing anti-inflammatory gut microbiome. By summarizing all the important findings, we highlight that mannose treatment is a safe and promising novel strategy to suppress inflammatory diseases, including autoimmune disease and allergic disease.


Assuntos
Inflamação/tratamento farmacológico , Manose/uso terapêutico , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/imunologia , Disbiose/tratamento farmacológico , Disbiose/prevenção & controle , Frutose/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Glucose/efeitos adversos , Humanos , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/imunologia , Inflamação/induzido quimicamente , Inflamação/imunologia , Inflamação/prevenção & controle , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Manose/farmacologia , Camundongos , Obesidade/tratamento farmacológico , Sacarose/efeitos adversos , Subpopulações de Linfócitos T/efeitos dos fármacos , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
9.
ACS Appl Mater Interfaces ; 13(28): 32729-32742, 2021 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-34247476

RESUMO

Aristolochic acid (AA) has been reported to cause a series of health problems, including aristolochic acid nephropathy and liver cancer. However, AA-containing herbs are highly safe in combination with berberine (Ber)-containing herbs in traditional medicine, suggesting the possible neutralizing effect of Ber on the toxicity of AA. In the present study, in vivo systematic toxicological experiments performed in zebrafish and mice showed that the supramolecule self-assembly formed by Ber and AA significantly reduced the toxicity of AA and attenuated AA-induced acute kidney injury. Ber and AA can self-assemble into linear heterogenous supramolecules (A-B) via electrostatic attraction and π-π stacking, with the hydrophobic groups outside and the hydrophilic groups inside during the drug combination practice. This self-assembly strategy may block the toxic site of AA and hinder its metabolism. Meanwhile, A-B linear supramolecules did not disrupt the homeostasis of gut microflora as AA did. RNA-sequence analysis, immunostaining, and western blot of the mice kidney also showed that A-B supramolecules almost abolished the acute nephrotoxicity of AA in the activation of the immune system and tumorigenesis-related pathways.


Assuntos
Ácidos Aristolóquicos/toxicidade , Berberina/uso terapêutico , Medicamentos de Ervas Chinesas/toxicidade , Nefropatias/prevenção & controle , Substâncias Macromoleculares/uso terapêutico , Animais , Ácidos Aristolóquicos/química , Berberina/química , Interações Medicamentosas , Medicamentos de Ervas Chinesas/química , Disbiose/prevenção & controle , Microbioma Gastrointestinal/efeitos dos fármacos , Expressão Gênica/efeitos dos fármacos , Rim/efeitos dos fármacos , Rim/metabolismo , Rim/patologia , Nefropatias/induzido quimicamente , Nefropatias/patologia , Células Matadoras Naturais/efeitos dos fármacos , Substâncias Macromoleculares/química , Substâncias Macromoleculares/toxicidade , Masculino , Camundongos Endogâmicos C57BL , Neutrófilos/efeitos dos fármacos , Fator de Transcrição RelA/metabolismo , Peixe-Zebra , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Nutrients ; 13(7)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209683

RESUMO

Gut microbiota dysbiosis is a common feature in colorectal cancer (CRC) and inflammatory bowel diseases (IBD). Adoption of the Mediterranean diet (MD) has been proposed as a therapeutic approach for the prevention of multiple diseases, and one of its mechanisms of action is the modulation of the microbiota. We aimed to determine whether MD can be used as a preventive measure against cancer and inflammation-related diseases of the gut, based on its capacity to modulate the local microbiota. A joint meta-analysis of publicly available 16S data derived from subjects following MD or other diets and from patients with CRC, IBD, or other gut-related diseases was conducted. We observed that the microbiota associated with MD was enriched in bacteria that promote an anti-inflammatory environment but low in taxa with pro-inflammatory properties capable of altering intestinal barrier functions. We found an opposite trend in patients with intestinal diseases, including cancer. Some of these differences were maintained even when MD was compared to healthy controls without a defined diet. Our findings highlight the unique effects of MD on the gut microbiota and suggest that integrating MD principles into a person's lifestyle may serve as a preventive method against cancer and other gut-related diseases.


Assuntos
Neoplasias Colorretais/prevenção & controle , Dieta Mediterrânea , Disbiose/prevenção & controle , Gastroenteropatias/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Adulto , Colo/microbiologia , Neoplasias Colorretais/microbiologia , Disbiose/microbiologia , Feminino , Gastroenteropatias/microbiologia , Humanos , Doenças Inflamatórias Intestinais/microbiologia , Doenças Inflamatórias Intestinais/prevenção & controle , Masculino , Pessoa de Meia-Idade
11.
mBio ; 12(4): e0115521, 2021 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-34225483

RESUMO

Nonalcoholic fatty liver disease (NAFLD), characterized by excessive fat deposition in the liver unrelated to alcohol consumption, is highly prevalent worldwide. However, effective therapeutic agents approved for NAFLD treatment are lacking. An ileal bile acid transporter inhibitor (IBATi), which represents a new mode of treatment of chronic idiopathic constipation, leads to increased delivery of bile acids to the colon. We investigated the effect of IBATi against NAFLD through modification of the gut microbiota in mice. IBATi treatment significantly suppressed body weight gain, liver dysfunction, and serum low-density lipoprotein levels and significantly decreased NAFLD activity scores in high-fat diet (HFD) mice. Treatment with IBATi ameliorated the decreased hepatic cholesterol 7-a-monooxygenase (Cyp7a1) and increased ileal fibroblast growth factor 15 (Fgf15) mRNA expression in HFD mice. Further, IBATi treatment changed the α-diversity in the gut microbiota reduced by HFD, which was analyzed in feces using 16S rRNA sequencing. To establish the mechanism underlying improvement in NAFLD induced by IBATi, we recolonized antibiotic solution-treated mice by fecal microbiome transplantation (FMT) using stool from HFD or HFD plus IBATi mice. This is the first report that fecally transplanted gut microbiota from HFD plus IBATi mice prevented hepatic steatosis caused by HFD. In conclusion, IBATi improved hepatic steatosis by ameliorating gut microbiota dysbiosis in NAFLD model mice, suggesting a potential therapeutic agent for NAFLD treatment. IMPORTANCE NAFLD is an increasingly recognized condition that may progress to liver cirrhosis and hepatocellular carcinoma, and community surveys have assessed that the prevalence is 14 to 32% worldwide. The first line of treatment for NAFLD is lifestyle modification to achieve weight reduction, particularly through diet and exercise. However, weight reduction is difficult to achieve and maintain, and pharmacological agents approved for the treatment of NAFLD are lacking. This study investigated the influence of the gut microbiota and the effect of an IBATi on NAFLD using a murine model. Treatment with IBATi significantly improved NAFLD in HFD mice. Further, fecal microbiome transplantation using stool from HFD plus IBATi mice prevented hepatic steatosis caused by HFD. Our study makes a significant contribution to the literature because the study findings suggest a potential treatment strategy for NAFLD patients by ameliorating gut microbiota dysbiosis.


Assuntos
Disbiose/prevenção & controle , Fígado Gorduroso/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Hepatopatia Gordurosa não Alcoólica/microbiologia , Animais , Ácidos e Sais Biliares/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Disbiose/microbiologia , Fígado Gorduroso/microbiologia , Microbioma Gastrointestinal/genética , Fígado/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/genética
12.
Front Cell Infect Microbiol ; 11: 622327, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34178715

RESUMO

Neonatal colonization of the gastrointestinal tract depends on mother microbiome, thus mother microbiota dysbiosis is transmitted to the offspring during the delivery and shaped by breastmilk characteristics. Here we used a murine model of UC predisposition (Winnie-/-) to evaluate the effects of maternal diet during pregnancy and lactation. Using heterozygous breeders, we obtained both Winnie-/- and C57BL/6 littermates from the same mother and compared their microbiota at weaning and adult age, using a diet enriched with 1% tomato fruit of a line - named Bronze - highly enriched in bioactive polyphenols, or Control tomato. Females received enriched diets two weeks before the beginning of the breeding and never stopped for the following six months. No significant effect was observed in regard to the percentage of Winnie-/- offspring, as with both diets the percentage was about 25% as expected. Winnie littermates from breeders fed with the Bronze-enriched diet showed reduced dysbiosis at 4 weeks of age if compared with Winnie under the Control tomato diet. This effect was then reduced when mice reached adult age. Conversely, the microbiota of C57BL/6 does not change significantly, indicating that fortified mothers-diet significantly contribute to preventing dysbiosis in genetically predisposed offspring, but has mild effects on healthy littermates and adult mice. An overall tendency towards reduced inflammation was underlined by the colon weight and the percentage of Foxp3+ cells reduction in Winnie mice fed with Bronze diet. Control diet did not show similar tendency.


Assuntos
Colite Ulcerativa , Microbioma Gastrointestinal , Animais , Dieta , Modelos Animais de Doenças , Disbiose/prevenção & controle , Feminino , Lactação , Camundongos , Camundongos Endogâmicos C57BL , Polifenóis , Gravidez
13.
Nat Med ; 27(7): 1212-1222, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34183837

RESUMO

Inflammatory bowel disease (IBD) is a complex chronic inflammatory disorder of the gastrointestinal tract. Extracellular adenosine triphosphate (eATP) produced by the commensal microbiota and host cells activates purinergic signaling, promoting intestinal inflammation and pathology. Based on the role of eATP in intestinal inflammation, we developed yeast-based engineered probiotics that express a human P2Y2 purinergic receptor with up to a 1,000-fold increase in eATP sensitivity. We linked the activation of this engineered P2Y2 receptor to the secretion of the ATP-degrading enzyme apyrase, thus creating engineered yeast probiotics capable of sensing a pro-inflammatory molecule and generating a proportional self-regulated response aimed at its neutralization. These self-tunable yeast probiotics suppressed intestinal inflammation in mouse models of IBD, reducing intestinal fibrosis and dysbiosis with an efficacy similar to or higher than that of standard-of-care therapies usually associated with notable adverse events. By combining directed evolution and synthetic gene circuits, we developed a unique self-modulatory platform for the treatment of IBD and potentially other inflammation-driven pathologies.


Assuntos
Trifosfato de Adenosina/metabolismo , Apirase/metabolismo , Doenças Inflamatórias Intestinais/terapia , Probióticos/uso terapêutico , Receptores Purinérgicos P2Y2/metabolismo , Saccharomyces cerevisiae/metabolismo , Animais , Apirase/genética , Sistemas CRISPR-Cas/genética , Modelos Animais de Doenças , Disbiose/prevenção & controle , Feminino , Fibrose/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Humanos , Doenças Inflamatórias Intestinais/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Receptores Purinérgicos P2Y2/genética , Saccharomyces cerevisiae/genética
14.
Sci Rep ; 11(1): 7008, 2021 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-33772084

RESUMO

Dietary fiber functions as a prebiotic to determine the gut microbe composition. The gut microbiota influences the metabolic functions and immune responses in human health. The gut microbiota and metabolites produced by various dietary components not only modulate immunity but also impact various organs. Although recent findings have suggested that microbial dysbiosis is associated with several respiratory diseases, including asthma, cystic fibrosis, and allergy, the role of microbiota and metabolites produced by dietary nutrients with respect to pulmonary disease remains unclear. Therefore, we explored whether the gut microbiota and metabolites produced by dietary fiber components could influence a cigarette smoking (CS)-exposed emphysema model. In this study, it was demonstrated that a high-fiber diet including non-fermentable cellulose and fermentable pectin attenuated the pathological changes associated with emphysema progression and the inflammatory response in CS-exposed emphysema mice. Moreover, we observed that different types of dietary fiber could modulate the diversity of gut microbiota and differentially impacted anabolism including the generation of short-chain fatty acids, bile acids, and sphingolipids. Overall, the results of this study indicate that high-fiber diets play a beneficial role in the gut microbiota-metabolite modulation and substantially affect CS-exposed emphysema mice. Furthermore, this study suggests the therapeutic potential of gut microbiota and metabolites from a high-fiber diet in emphysema via local and systemic inflammation inhibition, which may be useful in the development of a new COPD treatment plan.


Assuntos
Fibras na Dieta/farmacologia , Enfisema/dietoterapia , Enfisema/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Prebióticos/administração & dosagem , Animais , Ácidos e Sais Biliares/biossíntese , Celulose/farmacologia , Fumar Cigarros/efeitos adversos , Dieta , Disbiose/prevenção & controle , Ácidos Graxos Voláteis/biossíntese , Feminino , Inflamação/dietoterapia , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Pectinas/farmacologia , Esfingolipídeos/biossíntese
15.
Food Res Int ; 141: 110153, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33642019

RESUMO

Tea polyphenols (TPs) are now widely used in foods for various biological activities. However, they are rarely used in foods to regulate gut microbiota dysbiosis induced by antibiotics. We assessed the regulation of TPs on gut microbiota with an antibiotic-induced intestinal flora disorder mouse model. The mice were orally administered with cefixime for 8 days, then received TPs for 28 days. We found that the antibiotic had a profound impact on the gut microbiota. Compared with the normal group, significant decreases in the species richness and diversity and the production of short-chain fatty acids (SCFAs) were still observed 28 days after the antibiotic treatment, although there was no significant difference in the colonic mucosa. TPs significantly alleviated the decrease of the richness and diversity of gut microbiota caused by the antibiotic treatment, and significantly increased the relative abundance of beneficial microbes such as Lactobacillus, Akkermansia, Blautia, Roseburia, and Eubacterium. The function prediction showed that TPs significantly decreased the relative abundance of genes related to human diseases, yet significantly increased the relative abundance of genes related to cell growth and death, cell motility, and energy metabolism. These showed that TPs could regulate the gut microbiota dysbiosis induced by antibiotics, thus decreasing the risk of diseases such as obesity, cancer, and diabetes. These suggest that TPs have a great potential to be used as a functional food ingredient to prevent or reduce adverse effects of antibiotics.


Assuntos
Disbiose , Microbioma Gastrointestinal , Animais , Antibacterianos/toxicidade , Disbiose/induzido quimicamente , Disbiose/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , Polifenóis/farmacologia , Chá
16.
Drug Discov Today ; 26(9): 2159-2163, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33639249

RESUMO

Homeostasis of the intestinal microbiota is currently recognized as a major contributor to human health. Furthermore, intestinal dysbiosis is associated with a multitude of consequences, including intestinal colonization by antibiotic-resistant or pathogenic bacteria, such as Clostridioides difficile, and reduced efficacy of promising anticancer immunotherapies. By far, the most immediate and drastic exposure leading to dysbiosis is antibiotic treatment. Many attempts have been made to prevent or repair antibiotic-associated dysbiosis. Here, we review these innovations and the difficulties associated with their development.


Assuntos
Antibacterianos/efeitos adversos , Disbiose/induzido quimicamente , Microbioma Gastrointestinal/efeitos dos fármacos , Animais , Disbiose/prevenção & controle , Humanos
17.
J Agric Food Chem ; 69(5): 1675-1683, 2021 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-33494608

RESUMO

Cadmium (Cd) is a heavy metal, which is widely used in the industry and daily life. It has a long half-life, so large amounts of Cd can accumulate in humans and become toxic. Chlorogenic acid (CGA) can eliminate free radicals and inhibit lipid peroxidation and is mainly used to prevent metal toxicity. In the present study, mice are given CGA by intraperitoneal injection or gavage, respectively, to explore the mechanism of preventing Cd toxicity. In acute Cd-exposed mice, CGA treatment (ip) alleviated Cd-induced oxidative damage and reduced the production of NO and MPO in the liver and kidney tissues, while TLR4 expression levels did not change significantly. After 8 weeks of Cd exposure, CGA administration (gavage) significantly alleviated gut dysbiosis by decreasing the Firmicutes to Bacteroidetes ratio, enhancing the relative abundances of bacteria, including Ruminiclostridium_9, Alloprevotella, and Rikenella, and inhibiting the activation of the TLR4/MyD88/NF-κB signaling pathway. These findings suggested that protection mechanisms underlying the oral administration of CGA against the Cd-induced hepatorenal injury was related to the regulation of the intestinal flora balance. CGA can be used as an effective component in daily diet to prevent Cd toxicity.


Assuntos
Cádmio/toxicidade , Ácido Clorogênico/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Hepatopatias/prevenção & controle , Administração Oral , Animais , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Disbiose/etiologia , Disbiose/metabolismo , Disbiose/microbiologia , Disbiose/prevenção & controle , Fígado/efeitos dos fármacos , Fígado/metabolismo , Hepatopatias/etiologia , Hepatopatias/metabolismo , Hepatopatias/microbiologia , Masculino , Camundongos , NF-kappa B/genética , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
18.
Gut Microbes ; 12(1): 1-18, 2020 11 09.
Artigo em Inglês | MEDLINE | ID: mdl-33300439

RESUMO

New evidence shows that host-microbiota crosstalk can be modulated via endogenous miRNAs. We have previously reported that miR-21 ablation protects against liver injury in cholestasis. In this study, we investigated the role of miR-21 in modulating the gut microbiota during cholestasis and its effects in liver dysfunction. Mice lacking miR-21 had reduced liver damage and were protected against small intestinal injury as well as from gut microbiota dysbiosis when subjected to bile duct ligation surgery. The unique microbiota profile of miR-21KO mice was characterized by an increase in Lactobacillus, a key microbiome genus for gut homeostasis. Interestingly, in vitro incubation of synthetic miR-21 directly reduced Lactobacillus load. Moreover, supplementation with Lactobacillus reuteri revealed reduced liver fibrosis in acute bile duct-ligated mice, mimicking the protective effects in miR-21 knockout mice. D-lactate, a main product of Lactobacillus, regulates gut homeostasis that may link with reduced liver fibrosis. Altogether, our results demonstrate that miR-21 promotes liver dysfunction through direct modulation of the gut microbiota and highlight the potential therapeutic effects of Lactobacillus supplementation in gut and liver homeostasis.


Assuntos
Microbioma Gastrointestinal/genética , Lactobacillus/genética , Cirrose Hepática/genética , Fígado/lesões , MicroRNAs/genética , Animais , Colestase/patologia , Disbiose/genética , Disbiose/prevenção & controle , Feminino , Microbioma Gastrointestinal/fisiologia , Ácido Láctico/metabolismo , Fígado/patologia , Cirrose Hepática/microbiologia , Cirrose Hepática/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
19.
Can J Surg ; 63(6): E581-E593, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33278908

RESUMO

Humans are destined to explore space, yet critical illness and injury may be catastrophically limiting for extraterrestrial travel. Humans are superorganisms living in symbiosis with their microbiomes, whose genetic diversity dwarfs that of humans. Symbiosis is critical and imbalances are associated with disease, occurring within hours of serious illness and injury. There are many characteristics of space flight that negatively influence the microbiome, especially deep space itself, with its increased radiation and absence of gravity. Prolonged weightlessness causes many physiologic changes that are detrimental; some resemble aging and will adversely affect the ability to tolerate critical illness or injury and subsequent treatment. Critical illness-induced intra-abdominal hypertension (IAH) may induce malperfusion of both the viscera and microbiome, with potentially catastrophic effects. Evidence from animal models confirms profound IAH effects on the gut, namely ischemia and disruption of barrier function, mechanistically linking IAH to resultant organ dysfunction. Therefore, a pathologic dysbiome, space-induced immune dysfunction and a diminished cardiorespiratory reserve with exacerbated susceptibility to IAH, imply that a space-deconditioned astronaut will be vulnerable to IAH-induced gut malperfusion. This sets the stage for severe gut ischemia and massive biomediator generation in an astronaut with reduced cardiorespiratory/immunological capacity. Fortunately, experiments in weightless analogue environments suggest that IAH may be ameliorated by conformational abdominal wall changes and a resetting of thoracoabdominal mechanics. Thus, review of the interactions of physiologic changes with prolonged weightlessness and IAH is required to identify appropriate questions for planning exploration class space surgical care.


L'humanité est à l'aube d'une nouvelle ère d'exploration spatiale, mais le risque de maladies et blessures graves pourrait restreindre de manière catastrophique le potentiel des voyages dans l'espace. L'être humain est un superorganisme vivant en symbiose avec son microbiote, dont la diversité génétique éclipse celle de l'hôte. Cette symbiose est essentielle : tout déséquilibre est associé à une dégradation de l'état de santé dans les heures suivant l'occurrence d'une blessure ou d'une maladie grave. Bon nombre de caractéristiques propres au vol spatial ont des répercussions négatives sur le microbiote; l'espace lointain présente des dangers particuliers en raison de l'exposition accrue au rayonnement et de l'absence de gravité. L'exposition prolongée à l'apesanteur cause une myriade de changements physiologiques nuisant à la santé. Certains ressemblent à des processus de vieillissement et réduiront la capacité à tolérer une blessure ou une maladie grave et son traitement. L'hypertension intra-abdominale (HIA) causée par une maladie grave peut réduire la perfusion des viscères et du microbiote, ce qui peut avoir des conséquences catastrophiques. Des études sur modèle animal ont confirmé les effets profondément délétères de l'HIA sur les intestins par l'apparition d'une ischémie et une altération de la barrière intestinale; cette découverte permettrait d'établir un lien mécanistique entre l'HIA et la défaillance d'organes résultante. Par conséquent, une dysbiose pathologique, associée à un dysfonctionnement immunitaire en apesanteur et à une réduction de la réserve cardiorespiratoire accompagnée d'une exacerbation de la susceptibilité à l'HIA, pourrait signifier qu'un astronaute exposé à l'effet déconditionnant de l'apesanteur serait vulnérable aux problèmes de perfusion de l'intestin découlant de l'HIA. Ce problème pourrait à son tour mener à une ischémie intestinale grave et à une production massive de biomédiateurs chez un astronaute présentant déjà une capacité cardiorespiratoire et immunitaire réduite. Heureusement, des expériences dans des environnements simulant l'apesanteur semblent indiquer que les effets de l'HIA pourraient être contrés par des changements conformationnels de la paroi abdominale et un rétablissement de la mécanique thoracoabdominale. Par conséquent, un examen des interactions des changements physiologiques associés à un état d'apesanteur prolongé et à l'HIA est requis pour déterminer les questions à poser afin de planifier adéquatement les soins chirurgicaux en contexte d'exploration spatiale.


Assuntos
Disbiose/fisiopatologia , Hipertensão Intra-Abdominal/fisiopatologia , Insuficiência de Múltiplos Órgãos/fisiopatologia , Voo Espacial , Ausência de Peso/efeitos adversos , Abdome/fisiopatologia , Animais , Estado Terminal , Disbiose/etiologia , Disbiose/prevenção & controle , Microbioma Gastrointestinal/fisiologia , Humanos , Hipertensão Intra-Abdominal/etiologia , Hipertensão Intra-Abdominal/prevenção & controle , Modelos Animais , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/prevenção & controle
20.
World J Gastroenterol ; 26(36): 5420-5436, 2020 Sep 28.
Artigo em Inglês | MEDLINE | ID: mdl-33024394

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the third leading cause of cancer mortality worldwide. The gut microbiota can help maintain healthy metabolism and immunity. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is a critical factor in promoting health and homeostasis; it promotes intestinal immunity, stimulates bone marrow precursors to generate macrophage colonies, and enhances the antibacterial and antitumor activity of circulating monocytes. As such, GM-CSF may protect against HCC development by regulating immunity as well as intestinal microecology. AIM: To investigate the impact of GM-CSF on the gut microbiome and metabolic characteristics of HCC. METHODS: Thirty-six male BALB/c nude mice were divided into three groups: Control (n = 10), HCC (n = 13), and HCC + GM-CSF (GM-CSF overexpression, n = 13). We utilized HCC cells to establish orthotopic transplantation tumor models of HCC with normal and over-expressing GM-CSF. Liver injury, immune inflammatory function and intestinal barrier function were evaluated. The fecal microbiome and metabolome were studied using 16S rRNA absolute quantification sequencing and gas chromatography-mass spectrometry. RESULTS: GM-CSF overexpression significantly affected the gut microbiome of mice with HCC and resulted in a high abundance of organisms of the genera Roseburia, Blautia and Butyricimonass, along with a significant reduction in Prevotella, Parabacteroides, Anaerotruncus, Streptococcus, Clostridium, and Mucispirillum. Likewise, GM-CSF overexpression resulted in a substantial increase in fecal biotin and oleic acid levels, along with a prominent decrease in the fecal succinic acid, adenosine, fumaric acid, lipoic acid, and maleic acid levels. Correlation analysis revealed that the intestinal microbiota and fecal metabolites induced by GM-CSF were primarily involved in pathways related to reducing the inflammatory response, biotin metabolism, and intestinal barrier dysfunction. CONCLUSION: GM-CSF can protect against HCC development by regulating immunity and modulating the abundance of specific intestinal microorganisms and their metabolites. This study provides new insights into the therapeutic approaches for HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Carcinoma Hepatocelular/prevenção & controle , Disbiose/prevenção & controle , Fator Estimulador de Colônias de Granulócitos e Macrófagos , Inflamação , Neoplasias Hepáticas/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , RNA Ribossômico 16S/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA