Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 88
Filtrar
Más filtros

Medicinas Complementárias
Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Phytomedicine ; 128: 155349, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38522315

RESUMEN

BACKGROUND: Trimethylamine N-oxide (TMAO), a metabolite produced by intestinal microbiota through metabolizing phosphatidylcholine, choline, l-carnitine and betaine in the diet, has been implicated in the pathogenesis of atherosclerosis (AS). Concurrently, dietary polyphenols have garnered attention for their potential to ameliorate obesity, diabetes and atherosclerosis primarily by modulating the intestinal microbial structure. Hickory (Carya cathayensis) nut, a polyphenol-rich food product favored for its palatability, emerges as a candidate for exploration. HYPOTHESIS/PURPOSE: The relationship between polyphenol of hickory nut and atherosclerosis prevention will be firstly clarified, providing theoretical basis for the discovery of natural products counteracting TMAO-induced AS process in hickory nut. STUDY DESIGN AND METHODS: Employing Enzyme-linked Immunosorbent Assay (ELISA) and histological examination of aortic samples, the effects of total polyphenol extract on obesity index, inflammatory index and pathological changes of atherosclerosis in C57BL/6 J mice fed with high-fat and high choline diet were evaluated. Further, the composition, abundance, and function of mouse gut microbiota were analyzed through 16srDNA sequencing. Concurrently, the levels of TMAO and the expression of key enzymes (CutC and FMO3) involved in its synthesis are quantified using ELISA, Western Blot and Real-Time Quantitative PCR (RT-qPCR). Additionally, targeted metabolomic profiling of the hickory nut polyphenol extract was conducted, accompanied by molecular docking simulations to predict interactions between candidate polyphenols and the CutC/FMO3 using Autodock Vina. Finally, the docking prediction were verified by microscale thermophoresis (MST) . RESULTS: Polyphenol extracts of hickory nut improved the index of obesity and inflammation, and alleviated the pathological changes of atherosclerosis in C57BL/6 J mice fed with high-fat and high-choline diet. Meanwhile, these polyphenol extracts also changed the composition and function of intestinal microbiota, and increased the abundance of microorganisms in mice. Notably, the abundance of intestinal microbiota endowed with CutC gene was significantly reduced, coherent with expression of CutC catalyzing TMA production. Moreover, polyphenol extracts also decreased the expression of FMO3 in the liver, contributing to the reduction of TMAO levels in serum. Furthermore, metabonomic profile analysis of these polyphenol extracts identified 647 kinds of polyphenols. Molecular docking predication further demonstrated that Casuariin and Cinnamtannin B2 had the most potential inhibition on the enzymatic activities of CutC or FMO3, respectively. Notably, MST analysis corroborated the potential for direct interaction between CutC enzyme and available polyphenols such as Corilagin, (-)-Gallocatechin gallate and Epigallocatechin gallate. CONCLUSION: Hickory polyphenol extract can mitigate HFD-induced AS by regulating intestinal microflora in murine models. In addition, TMA-FMO3-TMAO pathway may play a key role in this process. This research unveils, for the inaugural time, the complex interaction between hickory nut-derived polyphenols and gut microbial, providing novel insights into the role of dietary polyphenols in AS prevention.


Asunto(s)
Aterosclerosis , Microbioma Gastrointestinal , Metilaminas , Ratones Endogámicos C57BL , Oxigenasas , Polifenoles , Animales , Polifenoles/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Metilaminas/metabolismo , Aterosclerosis/prevención & control , Aterosclerosis/tratamiento farmacológico , Masculino , Ratones , Nueces/química , Dieta Alta en Grasa/efectos adversos , Colina , Extractos Vegetales/farmacología , Extractos Vegetales/química , Obesidad/prevención & control , Simulación del Acoplamiento Molecular
2.
Atherosclerosis ; 391: 117431, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38408412

RESUMEN

BACKGROUND AND AIMS: The gut microbe-derived metabolite trimethylamine-N-oxide (TMAO) has been implicated in the development of cardiovascular fibrosis. Endoplasmic reticulum (ER) stress occurs after the dysfunction of ER and its structure. The three signals PERK/ATF-4, IRE-1α/XBP-1s and ATF6 are activated upon ER stress. Recent reports have suggested that the activation of PERK/ATF-4 and IRE-1α/XBP-1s signaling contributes to cardiovascular fibrosis. However, whether TMAO mediates aortic valve fibrosis by activating PERK/ATF-4 and IRE-1α/XBP-1s signaling remains unclear. METHODS: Human aortic valve interstitial cells (AVICs) were isolated from aortic valve leaflets. PERK IRE-1α, ATF-4, XBP-1s and CHOP expression, and production of collagen Ⅰ and TGF-ß1 were analyzed following treatment with TMAO. The role of PERK/ATF-4 and IRE-1α/XBP-1s signaling pathways in TMAO-induced fibrotic formation was determined using inhibitors and small interfering RNA. RESULTS: Diseased valves produced greater levels of ATF-4, XBP-1, collagen Ⅰ and TGF-ß1. Interestingly, diseased cells exhibited augmented PERK/ATF-4 and IRE-1α/XBP-1s activation after TMAO stimulation. Inhibition and silencing of PERK/ATF-4 and IRE-1α/XBP-1s each resulted in enhanced suppression of TMAO-induced fibrogenic activity in diseased cells. Mice treated with dietary choline supplementation had substantially increased TMAO levels and aortic valve fibrosis, which were reduced by 3,3-dimethyl-1-butanol (DMB, an inhibitor of trimethylamine formation) treatment. Moreover, a high-choline and high-fat diet remodeled the gut microbiota in mice. CONCLUSIONS: TMAO promoted aortic valve fibrosis through activation of PERK/ATF-4 and IRE-1α/XBP-1s signaling pathways in vitro and in vivo. Modulation of diet, gut microbiota, TMAO, PERK/ATF-4 and IRE1-α/XBP-1s may be a promising approach to prevent aortic valve fibrosis.


Asunto(s)
Microbioma Gastrointestinal , Factor de Crecimiento Transformador beta1 , Ratones , Humanos , Animales , Factor de Crecimiento Transformador beta1/metabolismo , Válvula Aórtica/metabolismo , Metilaminas/toxicidad , Metilaminas/metabolismo , Fibrosis , Colágeno , Colina , Óxidos
3.
Nutrients ; 15(3)2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36771270

RESUMEN

Trimethylamine N-oxide (TMAO) is a microbial metabolite derived from nutrients, such as choline, L-carnitine, ergothioneine and betaine. Recently, it has come under the spotlight for its close interactions with gut microbiota and implications for gastrointestinal cancers, cardiovascular disease, and systemic inflammation. The culprits in the origin of these pathologies may be food sources, in particular, high fat meat, offal, egg yolk, whole dairy products, and fatty fish, but intercalated between these food sources and the production of pro-inflammatory TMAO, the composition of gut microbiota plays an important role in modulating this process. The aim of this review is to explain how the gut microbiota interacts with the conversion of specific compounds into TMA and its oxidation to TMAO. We will first cover the correlation between TMAO and various pathologies such as dysbiosis, then focus on cardiovascular disease, with a particular emphasis on pro-atherogenic factors, and then on systemic inflammation and gastrointestinal cancers. Finally, we will discuss primary prevention and therapies that are or may become possible. Possible treatments include modulation of the gut microbiota species with diets, physical activity and supplements, and administration of drugs, such as metformin and aspirin.


Asunto(s)
Enfermedades Cardiovasculares , Microbiota , Neoplasias , Animales , Colina/metabolismo , Metilaminas/metabolismo , Inflamación , Neoplasias/tratamiento farmacológico , Neoplasias/prevención & control
4.
Food Funct ; 13(23): 12039-12050, 2022 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-36331311

RESUMEN

Metabolic syndrome (MS) is a collection of risk factors of serious metabolic diseases. L-Carnitine is an essential nutrient for human health, and the precursor of trimethylamine N-oxide (TMAO). Previous studies have shown that the effect of L-carnitine on MS is controversial, and no studies have considered the role of gut microbiota in the regulation of MS by L-carnitine. In the present study, we established a high-fat diet (HFD)-induced obese mice model and systematically explored the effect of a broad range of dietary L-carnitine concentrations (0.2% to 4%) on the major components of MS. The results show that L-carnitine (0.5%-4%) reduced HFD-caused body-weight gain, visceral adipose tissue, glucose intolerance, hyperglycemia, HOMA-IR index, hyperlipemia, hypertension, and hyperuricemia. The elevation in the concentrations of IL-6, IL-1ß, and TNF-α and decline in IL-10 in both serum and adipose tissue were also attenuated by L-carnitine. Furthermore, dietary L-carnitine increased the serum levels of TMAO produced by gut microbes. High-dose L-carnitine (2% and 4%), but not low-dose L-carnitine (0.2%-1%), notably modulated the composition of gut microbiota and partially attenuated HFD-induced gut microbiota dysbiosis. These results suggest that the ameliorative effect of L-carnitine on MS was independent of TMAO production and only partially related to the regulation of gut microbiota. This study provides crucial evidence for the utilization of L-carnitine as a safe and effective supplement for MS.


Asunto(s)
Microbioma Gastrointestinal , Síndrome Metabólico , Humanos , Ratones , Animales , Carnitina/metabolismo , Dieta Alta en Grasa/efectos adversos , Ratones Obesos , Síndrome Metabólico/tratamiento farmacológico , Metilaminas/metabolismo , Suplementos Dietéticos
5.
Atherosclerosis ; 362: 47-55, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36347649

RESUMEN

BACKGROUND AND AIMS: Choline has been shown to exert atherogenic effects in Apoe-/- and Ldlr-/- mice, related to its conversion by gut bacteria into trimethylamine (TMA) that is converted by the liver into the proinflammatory metabolite trimethylamine-N-oxide (TMAO). Since butyrate beneficially modulates the gut microbiota and has anti-inflammatory and antiatherogenic properties, the aim of the present study was to investigate whether butyrate can alleviate choline-induced atherosclerosis. To this end, we used APOE*3-Leiden.CETP mice, a well-established atherosclerosis-prone model with human-like lipoprotein metabolism. METHODS: Female APOE*3-Leiden.CETP mice were fed an atherogenic diet alone or supplemented with choline, butyrate or their combination for 16 weeks. RESULTS: Interestingly, choline protected against fat mass gain, increased the abundance of anti-inflammatory gut microbes, and increased the expression of gut microbial genes involved in TMA and TMAO degradation. Butyrate similarly attenuated fat mass gain and beneficially modulated the gut microbiome, as shown by increased abundance of anti-inflammatory and short chain fatty acid-producing microbes, and inhibited expression of gut microbial genes involved in lipopolysaccharide synthesis. Both choline and butyrate upregulated hepatic expression of flavin-containing monooxygenases, and their combination resulted in highest circulating TMAO levels. Nonetheless, choline, butyrate and their combination did not influence atherosclerosis development, and TMAO levels were not associated with atherosclerotic lesion size. CONCLUSIONS: While choline and butyrate have been reported to oppositely modulate atherosclerosis development in Apoe-/- and Ldlr-/- mice as related to changes in the gut microbiota, both dietary constituents did not affect atherosclerosis development while beneficially modulating the gut microbiome in APOE*3-Leiden.CETP mice.


Asunto(s)
Aterosclerosis , Butiratos , Colina , Microbioma Gastrointestinal , Animales , Femenino , Ratones , Apolipoproteínas E/genética , Aterosclerosis/genética , Aterosclerosis/prevención & control , Aterosclerosis/metabolismo , Butiratos/farmacología , Proteínas de Transferencia de Ésteres de Colesterol/genética , Colina/farmacología , Metilaminas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
6.
Phytomedicine ; 104: 154305, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35792446

RESUMEN

BACKGROUND: Recent studies have shown that plasma trimethylamine-N-oxide (TMAO) level is highly correlated with the risk of atherosclerosis (AS), and the elevated level is significantly positively correlated with the incidence of AS. PURPOSE: The purpose of this article is to offer a useful summary of the correlation between TMAO and AS, and the effect of herbal monomers, herbal extracts, and formulas on anti-atherosclerosis mediated by TMAO. METHOD: The data contained in this article comes from PubMed, Web of Science, and China National Knowledge Infrastructure. RESULTS: This review discusses the main mechanism of AS induced by TMAO, including endothelial dysfunction, macrophage foaming, platelet reactivity, and cholesterol metabolism, and summarizes 6 herb monomers, 5 herb extracts, and 2 formulas that have been tested for their anti-TMAO activity. CONCLUSION: The current understanding of possible ways to reduce TMAO generation is discussed, with the effect and potential of herb monomers, herb extracts, and formulas highlighted.


Asunto(s)
Aterosclerosis , Medicina Tradicional China , Aterosclerosis/tratamiento farmacológico , Aterosclerosis/metabolismo , Humanos , Metabolismo de los Lípidos , Metilaminas/metabolismo
7.
Food Funct ; 13(15): 8022-8037, 2022 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-35839103

RESUMEN

The production of pro-atherogenic trimethylamine N-oxide (TMAO) is dependent on the gut microbiota metabolism of quaternary amines (i.e., choline) into trimethylamine (TMA). Nutritional strategies that target microbial conversion of choline into TMA could reduce cardiovascular disease and atherosclerosis burden by reducing subsequent formation of TMAO. This study aimed to evaluate (1) whether beverages rich in known inhibitors of TMA production (chlorogenic acid, catechin and epicatechin) can reduce TMA formation and (2) the effect of upper gastrointestinal digestion on efficacy. To do this, either raw or digested coffee, tea and cocoa beverages were evaluated for their TMA-d9 production inhibition in our ex vivo-in vitro fermentation model with human fecal slurries and choline-d9 substrate. Results showed that digestion was required to unlock the TMA-d9 production inhibition potential of coffee and cocoa beverages, and that teas did not possess a strong inhibition potential either digested or undigested. By fractionating digested bioactive beverages, we determined that those fractions rich in chlorogenic acid were the most bioactive. Overall, this study suggests that regular cocoa and coffee consumption could be a nutritional strategy able to reduce TMAO levels. In vivo studies should be carried out to confirm the potential of these beverages as strategies to inhibit TMA production.


Asunto(s)
Aterosclerosis , Café , Aterosclerosis/metabolismo , Bacterias/metabolismo , Bebidas , Ácido Clorogénico , Colina/metabolismo , Fermentación , Humanos , Metilaminas/metabolismo
8.
Gut Microbes ; 14(1): 2077602, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35613310

RESUMEN

Maladaptive inflammatory and immune responses are responsible for intestinal barrier integrity and function dysregulation. Proline/serine-rich coiled-coil protein 1 (PSRC1) critically contributes to the immune system, but direct data on the gut microbiota and the microbial metabolite trimethylamine N-oxide (TMAO) are lacking. Here, we investigated the impact of PSRC1 deletion on TMAO generation and atherosclerosis. We first found that PSRC1 deletion in apoE-/- mice accelerated atherosclerotic plaque formation, and then the gut microbiota and metabolites were detected using metagenomics and untargeted metabolomics. Our results showed that PSRC1 deficiency enriched trimethylamine (TMA)-producing bacteria and functional potential for TMA synthesis and accordingly enhanced plasma betaine and TMAO production. Furthermore, PSRC1 deficiency resulted in a proinflammatory colonic phenotype that was significantly associated with the dysregulated bacteria. Unexpectedly, hepatic RNA-seq indicated upregulated flavin monooxygenase 3 (FMO3) expression following PSRC1 knockout. Mechanistically, PSRC1 overexpression inhibited FMO3 expression in vitro, while an ERα inhibitor rescued the downregulation. Consistently, PSRC1-knockout mice exhibited higher plasma TMAO levels with a choline-supplemented diet, which was gut microbiota dependent, as evidenced by antibiotic treatment. To investigate the role of dysbiosis induced by PSRC1 deletion in atherogenesis, apoE-/- mice were transplanted with the fecal microbiota from either apoE-/- or PSRC1-/-apoE-/- donor mice. Mice that received PSRC1-knockout mouse feces showed an elevation in TMAO levels, as well as plaque lipid deposition and macrophage accumulation, which were accompanied by increased plasma lipid levels and impaired hepatic cholesterol transport. Overall, we identified PSRC1 as an atherosclerosis-protective factor, at least in part, attributable to its regulation of TMAO generation via a multistep pathway. Thus, PSRC1 holds great potential for manipulating the gut microbiome and alleviating atherosclerosis.


Asunto(s)
Aterosclerosis , Microbioma Gastrointestinal , Metilaminas , Oxigenasas , Fosfoproteínas , Animales , Aterosclerosis/genética , Aterosclerosis/microbiología , Bacterias/genética , Bacterias/metabolismo , Microbioma Gastrointestinal/fisiología , Metilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxigenasas de Función Mixta/metabolismo , Oxigenasas/metabolismo , Fosfoproteínas/deficiencia , Placa Aterosclerótica/metabolismo , Placa Aterosclerótica/microbiología
9.
Pharm Biol ; 60(1): 131-143, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34978949

RESUMEN

CONTEXT: The bulb of Lilium brownii F. E. Brown (Liliaceae) (LB) is a common Chinese medicine to relieve insomnia. OBJECTIVE: To investigate the molecular mechanism of LB relieving insomnia. MATERIALS AND METHODS: Insomnia model was induced by intraperitoneally injection p-chlorophenylalanine (PCPA) in Wistar rats. Rats were divided into three groups: Control, PCPA (400 mg/kg, i.p. 2 days), LB (598.64 mg/kg, oral 7 days). The levels of 5-hydroxytryptamine (5-HT), norepinephrine (NE), melatonin (MT), and the expression of GABAA, 5-HT1A and MT receptors, as well as pathological changes in hypothalamus, were evaluated. 16S rDNA sequencing and UPLC-MS/MS were used to reveal the change of the intestinal flora and metabolic profile. RESULTS: The adverse changes in the abundance and diversity of intestinal flora and faecal metabolic phenotype altered by PCPA in rats were reversed after LB treatment, accompanied by the up-regulated levels of 5-HT as 8.14 ng/mL, MT as 16.16 pg/mL, 5-HT1A R and GABAA R, down-regulated level of NE as 0.47 ng/mL, and the improvement of pathological phenomena of cells in the hypothalamus. And the arachidonic acid metabolism and tryptophan metabolism pathway most significantly altered by PCPA were markedly regulated by LB. Besides, it was also found that LB reduced the levels of kynurenic acid related to psychiatric disorders and trimethylamine-N-oxide associated with cardiovascular disease. CONCLUSION: The mechanism of LB relieving insomnia involves regulating flora and metabolites to resemble the control group. As a medicinal and edible herb, LB could be considered for development as a health-care food to relieve increasing insomniacs in the future.


Asunto(s)
Medicamentos Herbarios Chinos/farmacología , Lilium/química , Enfermedades Metabólicas/tratamiento farmacológico , Trastornos del Inicio y del Mantenimiento del Sueño/tratamiento farmacológico , Animales , Cromatografía Líquida de Alta Presión , Fenclonina , Microbioma Gastrointestinal/efectos de los fármacos , Ácido Quinurénico/metabolismo , Masculino , Metilaminas/metabolismo , Ratas , Ratas Wistar , Espectrometría de Masas en Tándem
10.
Elife ; 112022 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-35084335

RESUMEN

There is mounting evidence that microbes residing in the human intestine contribute to diverse alcohol-associated liver diseases (ALD) including the most deadly form known as alcohol-associated hepatitis (AH). However, mechanisms by which gut microbes synergize with excessive alcohol intake to promote liver injury are poorly understood. Furthermore, whether drugs that selectively target gut microbial metabolism can improve ALD has never been tested. We used liquid chromatography tandem mass spectrometry to quantify the levels of microbe and host choline co-metabolites in healthy controls and AH patients, finding elevated levels of the microbial metabolite trimethylamine (TMA) in AH. In subsequent studies, we treated mice with non-lethal bacterial choline TMA lyase (CutC/D) inhibitors to blunt gut microbe-dependent production of TMA in the context of chronic ethanol administration. Indices of liver injury were quantified by complementary RNA sequencing, biochemical, and histological approaches. In addition, we examined the impact of ethanol consumption and TMA lyase inhibition on gut microbiome structure via 16S rRNA sequencing. We show the gut microbial choline metabolite TMA is elevated in AH patients and correlates with reduced hepatic expression of the TMA oxygenase flavin-containing monooxygenase 3 (FMO3). Provocatively, we find that small molecule inhibition of gut microbial CutC/D activity protects mice from ethanol-induced liver injury. CutC/D inhibitor-driven improvement in ethanol-induced liver injury is associated with distinct reorganization of the gut microbiome and host liver transcriptome. The microbial metabolite TMA is elevated in patients with AH, and inhibition of TMA production from gut microbes can protect mice from ethanol-induced liver injury.


Asunto(s)
Bacterias/metabolismo , Enfermedad Hepática Crónica Inducida por Sustancias y Drogas/metabolismo , Microbioma Gastrointestinal , Hepatitis/metabolismo , Metilaminas/metabolismo , Animales , Etanol/efectos adversos , Femenino , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria
11.
J Phys Chem Lett ; 12(51): 12411-12418, 2021 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-34939822

RESUMEN

Trimethylamine N-oxide (TMAO), a choline-containing dietary supplement obtained from red meat, egg, and other animal resources, on excess accumulation is known to cause cardiovascular diseases (CVDs) like atherosclerosis. To understand the molecular mechanism of the pathogenesis of TMAO-induced CVDs, we have set up 1,2-dimyristoyl-sn-glycero-3-phosphocholine (DMPC) membrane in water that mimicked the endothelial cell membrane-blood interface of the artery wall and investigated the effect of an elevated concentration of TMAO on the membrane. We found that TMAO exerts an "action at a distance" mechanism through electrostatic force of attraction that significantly alters various properties of the membrane, like hydrophobicity, lateral organization, and interfacial water dynamics, which elevates the rigidity of the membrane. Such an effect was found to be further amplified in the presence of known causes of CVDs, i.e., high content of cholesterol (Chol). Therefore, TMAO-induced membrane rigidity may restrict the intrinsic elasticity of an artery membrane, expected to be introducing "hardening of the arteries", which makes the membrane atherosclerotic.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Membrana Dobles de Lípidos/metabolismo , Metilaminas/efectos adversos , Metilaminas/metabolismo , Nutrientes/metabolismo , Fosfolípidos/metabolismo , Animales , Enfermedades Cardiovasculares/inducido químicamente , Enfermedades Cardiovasculares/patología , Suplementos Dietéticos , Humanos , Membrana Dobles de Lípidos/química , Metilaminas/administración & dosificación , Nutrientes/administración & dosificación , Nutrientes/efectos adversos , Fosfolípidos/química , Electricidad Estática
12.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-34948275

RESUMEN

L-alpha glycerylphosphorylcholine (GPC), a nutritional supplement, has been demonstrated to improve neurological function. However, a new study suggests that GPC supplementation increases incident stroke risk thus its potential adverse effects warrant further investigation. Here we show that GPC promotes atherosclerosis in hyperlipidemic Apoe-/- mice. GPC can be metabolized to trimethylamine N-oxide, a pro-atherogenic agent, suggesting a potential molecular mechanism underlying the observed atherosclerosis progression. GPC supplementation shifted the gut microbial community structure, characterized by increased abundance of Parabacteroides, Ruminococcus, and Bacteroides and decreased abundance of Akkermansia, Lactobacillus, and Roseburia, as determined by 16S rRNA gene sequencing. These data are consistent with a reduction in fecal and cecal short chain fatty acids in GPC-fed mice. Additionally, we found that GPC supplementation led to an increased relative abundance of choline trimethylamine lyase (cutC)-encoding bacteria via qPCR. Interrogation of host inflammatory signaling showed that GPC supplementation increased expression of the proinflammatory effectors CXCL13 and TIMP-1 and activated NF-κB and MAPK signaling pathways in human coronary artery endothelial cells. Finally, targeted and untargeted metabolomic analysis of murine plasma revealed additional metabolites associated with GPC supplementation and atherosclerosis. In summary, our results show GPC promotes atherosclerosis through multiple mechanisms and that caution should be applied when using GPC as a nutritional supplement.


Asunto(s)
Aterosclerosis/etiología , Glicerilfosforilcolina/efectos adversos , Glicerilfosforilcolina/metabolismo , Animales , Apolipoproteínas E/genética , Aterosclerosis/inducido químicamente , Aterosclerosis/metabolismo , Ciego/metabolismo , Ciego/microbiología , Línea Celular , Suplementos Dietéticos/efectos adversos , Células Endoteliales/metabolismo , Ácidos Grasos Volátiles/metabolismo , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Glicerilfosforilcolina/farmacología , Humanos , Masculino , Metilaminas/efectos adversos , Metilaminas/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética , ARN Ribosómico 16S/metabolismo
13.
Food Funct ; 12(20): 9880-9893, 2021 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-34664588

RESUMEN

The present study investigated the induction of the glycolysis product methylglyoxal by trimethylamine (TMA) lyase synthesis in the intestinal microbiota and investigated the intervention mechanism of the effects of dietary fiber on methylglyoxal formation. Intestinal digesta samples, collected from the ceca of mice fed with choline-rich and fiber-supplemented diets, were incubated in an anaerobic environment at 37 °C and pH 7.0 with choline, glycine, and methylglyoxal as inductive factors. The differences between the gut microbiota and its metagenomic and metabonomics profiles were determined using 16S rRNA gene sequencing analysis. The results elucidated that the different dietary interventions could induce differences in the composition of the microbiota, gene expression profiles associated with glycine metabolism, and glycolysis. As compared to the gut microbiota of choline-diet fed mice, fiber supplementation effectively altered the composition of the microbiota and inhibited the genes involved in choline metabolism, glycine and methylglyoxal accumulation, and TMA lyase expression, and improved the methylglyoxal utilization by regulating the pathway related to pyruvate production. However, the intervention of exogenous methylglyoxal significantly decreased these effects. These findings successfully revealed the correlations between the TMA lyase expression and glycine level, as well as the inhibitory effects of dietary fiber on the glycine level, thereby highlighting the role of common glycolytic metabolites as a potential target for TMA production.


Asunto(s)
Colina/farmacología , Fibras de la Dieta/farmacología , Microbioma Gastrointestinal/efectos de los fármacos , Liasas/metabolismo , Metilaminas/metabolismo , Piruvaldehído/metabolismo , Animales , Dieta/métodos , Heces/microbiología , Femenino , Glicina/metabolismo , Glucólisis , Liasas/genética , Metabolómica/métodos , Metagenómica/métodos , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/genética
14.
Pharmacol Res ; 173: 105884, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34530121

RESUMEN

Trimethylamine-N-oxide (TMAO) has emerged as a promising new therapeutic target for the treatment of central nervous system diseases, atherosclerosis and other diseases. However, its origin in the brain is unclear. Gynostemma pentaphyllum (Thunb.) Makino can reduce the increase of TMAO level caused by a high fat diet. But its effective chemical composition and specific mechanism have not been reported. The study confirmed that TMA was more easily to penetrate blood brain barrier than TMAO, the MAO enzyme was partly involved in the transformation of the TMA in brain, which further supplemented the choline-TMA-TMAO pathway. Based on the above metabolic pathway, using multi-omics approaches, such as microbiodiversity, metagenomics and lipidomics, it was demonstrated that the reduction of plasma TMAO levels by gypenosides did not act on FMO3 and MAO in the pathway, but remodeled the microbiota and affected the trimethylamine lyase needed in the conversion of choline to TMA in intestinal flora. At the same time, gypenosides interfered with enzymes associated with TCA and lipid metabolism, thus affecting TMAO and lipid metabolism. Considering the bidirectional transformation of phosphatidycholine and choline, lipid metabolism and TMAO metabolism could affected each other to some extent. In conclusion, our study revealed the intrinsic correlation between long-term application of gypenosides to lipid reduction and nervous system protection, and explained why gypenosides were used to treat brain diseases, even though they had a poor ability to enter the brain. Besides, it provided a theoretical basis for clinical application of gypenosides and the development of new drugs.


Asunto(s)
Metilaminas/metabolismo , Animales , Encéfalo/metabolismo , Colina/farmacología , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/genética , Gynostemma , Metabolismo de los Lípidos/efectos de los fármacos , Metilaminas/sangre , Metilaminas/líquido cefalorraquídeo , Ratones Endogámicos BALB C , Microsomas Hepáticos/metabolismo , Oxigenasas/metabolismo , Extractos Vegetales/farmacología , ARN Ribosómico 16S
15.
NPJ Biofilms Microbiomes ; 7(1): 36, 2021 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863898

RESUMEN

Trimethylamine-N-oxide (TMAO), a derivative from the gut microbiota metabolite trimethylamine (TMA), has been identified to be an independent risk factor for promoting atherosclerosis. Evidences suggest that berberine (BBR) could be used to treat obesity, diabetes and atherosclerosis, however, its mechanism is not clear mainly because of its poor oral bioavailability. Here, we show that BBR attenuated TMA/TMAO production in the C57BL/6J and ApoE KO mice fed with choline-supplemented chow diet, and mitigated atherosclerotic lesion areas in ApoE KO mice. Inhibition of TMA/TMAO production by BBR-modulated gut microbiota was proved by a single-dose administration of d9-choline in vivo. Metagenomic analysis of cecal contents demonstrated that BBR altered gut microbiota composition, microbiome functionality, and cutC/cntA gene abundance. Furthermore, BBR was shown to inhibit choline-to-TMA conversion in TMA-producing bacteria in vitro and in gut microbial consortium from fecal samples of choline-fed mice and human volunteers, and the result was confirmed by transplantation of TMA-producing bacteria in mice. These results offer new insights into the mechanisms responsible for the anti-atherosclerosis effects of BBR, which inhibits commensal microbial TMA production via gut microbiota remodeling.


Asunto(s)
Aterosclerosis/etiología , Aterosclerosis/metabolismo , Berberina/farmacología , Colina/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Metilaminas/metabolismo , Animales , Aterosclerosis/patología , Dieta , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Disbiosis , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE
16.
J Cardiovasc Pharmacol ; 77(4): 458-469, 2021 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-33657052

RESUMEN

ABSTRACT: Chronic stable angina (CSA) is caused by coronary atherosclerosis. The gut microbiota (GM) and their metabolite trimethylamine-N-oxide (TMAO) levels are associated with atherosclerosis. Danlou tablet (DLT) combined with Salvia miltiorrhiza ligustrazine (SML) injection has been used to treat CSA. This study aims to investigate how DLT combined with SML (DLT-SML) regulates serum lipids, inflammatory cytokines, GM community, and microbial metabolite in patients with CSA. In this study, 30 patients with CSA were enrolled in the DLT-SML group, and 10 healthy volunteers were included in the healthy control group. The patients in the DLT-SML group were subdivided as the normal total cholesterol (TC) group and high-TC group according to their serum TC level before treatment. Blood samples were collected to investigate the (1) lipid content, including triglyceride (TG), TC, high-density lipoprotein cholesterol, and low-density lipoprotein cholesterol, (2) fasting blood glucose (Glu), (3) inflammatory cytokines, including interleukin-1 beta (IL-1ß), interleukin-6 (IL-6), interleukin-10 (IL-10), and tumor necrosis factor-α (TNF-α), and (4) gut-derived metabolite, including lipopolysaccharides and TMAO level. GM composition was analyzed by sequencing 16S rRNA of fecal samples. Results showed that DLT-SML significantly decreased serum TG, TC, low-density lipoprotein cholesterol, IL-1ß, TNF-α, and TMAO levels of patients with CSA. DLT-SML increased the abundance of Firmicutes and decreased Proteobacteria, which were significantly lower or higher in patients with CSA, respectively, compared with the healthy control group. In particular, DLT-SML increased the microbial diversity and decreased Firmicutes/Bacteroidetes ratio of patients with high-TC. The abundance of Sarcina, Anaerostipes, Streptococcus, Weissella, and Erysipelatoclostridium was decreased, whereas Romboutsia, Faecalibacterium, and Subdoligranulum were increased by DLT-SML treatment in patients with CSA. These findings indicated that DLT-SML improved patients with CSA by ameliorating dyslipidemia profile, decreasing the circulating inflammatory cytokines, and regulating the GM composition and their metabolites.


Asunto(s)
Angina Estable/tratamiento farmacológico , Antiinflamatorios/uso terapéutico , Bacterias/efectos de los fármacos , Medicamentos Herbarios Chinos/uso terapéutico , Dislipidemias/tratamiento farmacológico , Microbioma Gastrointestinal/efectos de los fármacos , Hipolipemiantes/uso terapéutico , Inflamación/tratamiento farmacológico , Pirazinas/uso terapéutico , Adulto , Anciano , Angina Estable/sangre , Angina Estable/diagnóstico , Angina Estable/microbiología , Antiinflamatorios/efectos adversos , Bacterias/metabolismo , Biomarcadores/sangre , China , Citocinas/sangre , Quimioterapia Combinada , Medicamentos Herbarios Chinos/efectos adversos , Disbiosis , Dislipidemias/sangre , Dislipidemias/diagnóstico , Femenino , Humanos , Hipolipemiantes/efectos adversos , Inflamación/sangre , Inflamación/diagnóstico , Mediadores de Inflamación/sangre , Lípidos/sangre , Masculino , Metilaminas/metabolismo , Persona de Mediana Edad , Pirazinas/efectos adversos , Factores de Tiempo , Resultado del Tratamiento
17.
Sci Rep ; 11(1): 518, 2021 01 12.
Artículo en Inglés | MEDLINE | ID: mdl-33436815

RESUMEN

Patients with chronic kidney disease (CKD) have elevated circulating levels of trimethylamine N-oxide (TMAO), a metabolite derived from gut microbes and associated with cardiovascular diseases. High circulating levels of TMAO and its dietary precursor, choline, predict increased risk for development of CKD in apparently healthy subjects, and studies in mice fed TMAO or choline suggest that TMAO can contribute to kidney impairment and renal fibrosis. Here we examined the interactions between TMAO, kidney disease, and cardiovascular disease in mouse models. We observed that while female hyperlipidemic apoE KO mice fed a 0.2% adenine diet for 14 weeks developed CKD with elevated plasma levels of TMAO, provision of a non-lethal inhibitor of gut microbial trimethylamine (TMA) production, iodomethylcholine (IMC), significantly reduced multiple markers of renal injury (plasma creatinine, cystatin C, FGF23, and TMAO), reduced histopathologic evidence of fibrosis, and markedly attenuated development of microalbuminuria. In addition, while the adenine-induced CKD model significantly increased heart weight, a surrogate marker for myocardial hypertrophy, this was largely prevented by IMC supplementation. Surprisingly, adenine feeding did not increase atherosclerosis and significantly decreased the expression of inflammatory genes in the aorta compared to the control groups, effects unrelated to TMAO levels. Our data demonstrate that inhibition of TMAO production attenuated CKD development and cardiac hypertrophy in mice, suggesting that TMAO reduction may be a novel strategy in treating CKD and its cardiovascular disease complications.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Metilaminas/efectos adversos , Metilaminas/metabolismo , Insuficiencia Renal Crónica/etiología , Adenina/administración & dosificación , Adenina/efectos adversos , Albuminuria/etiología , Animales , Cardiomegalia/etiología , Cardiomegalia/prevención & control , Colina/administración & dosificación , Colina/efectos adversos , Colina/análogos & derivados , Colina/farmacología , Modelos Animales de Enfermedad , Femenino , Factor-23 de Crecimiento de Fibroblastos , Fibrosis , Riñón/patología , Metilaminas/administración & dosificación , Ratones , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/prevención & control
18.
Biomed Pharmacother ; 134: 111156, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33401080

RESUMEN

Cardiac disorders contribute to one of the major causes of fatality across the world. Hypertensive patients, even well maintained on drugs, possess a high risk to cardiovascular diseases. It is, therefore, highly important to identify different factors and pathways that lead to risk and progression of cardiovascular disorders. Several animals and human studies suggest that taxonomical alterations in the gut are involved in the cardiovascular physiology. In this article, with the help of various experimental evidences, we suggest that the host gut-microbiota plays an important in this pathway. Short chain fatty acids (SCFAs) and Trimethyl Amine -n-Oxide (TMAO) are the two major products of gut microbiome. SCFAs present a crucial role in regulating the blood pressure, while TMAO is involved in pathogenesis of atherosclerosis and other coronary artery diseases, including hypertension. We prove that there exists a triangular bridge connecting the gap between dietary salt, hypertension and gut microbiome. We also present some of the dietary interventions which can regulate and control microbiota that can prevent cardiovascular complications.We strongly believe that this article would improve the understanding the role of gut microbiota in hypertension, and will be helpful in the development of novel therapeutic strategies for prevention of hypertension through restoring gut microbiome homeostasis in the near future.


Asunto(s)
Bacterias/metabolismo , Presión Sanguínea , Microbioma Gastrointestinal , Hipertensión/etiología , Intestinos/microbiología , Cloruro de Sodio Dietético/efectos adversos , Animales , Dieta Saludable , Dieta Hiposódica , Fibras de la Dieta/uso terapéutico , Suplementos Dietéticos , Disbiosis , Ácidos Grasos Volátiles/metabolismo , Humanos , Hipertensión/dietoterapia , Hipertensión/microbiología , Hipertensión/fisiopatología , Metilaminas/metabolismo , Medición de Riesgo , Factores de Riesgo
19.
Microbiome ; 8(1): 162, 2020 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-33213511

RESUMEN

The capability of gut microbiota in degrading foods and drugs administered orally can result in diversified efficacies and toxicity interpersonally and cause significant impact on human health. Production of atherogenic trimethylamine N-oxide (TMAO) from carnitine is a gut microbiota-directed pathway and varies widely among individuals. Here, we demonstrated a personalized TMAO formation and carnitine bioavailability from carnitine supplements by differentiating individual TMAO productivities with a recently developed oral carnitine challenge test (OCCT). By exploring gut microbiome in subjects characterized by TMAO producer phenotypes, we identified 39 operational taxonomy units that were highly correlated to TMAO productivity, including Emergencia timonensis, which has been recently discovered to convert γ-butyrobetaine to TMA in vitro. A microbiome-based random forest classifier was therefore constructed to predict the TMAO producer phenotype (AUROC = 0.81) which was then validated with an external cohort (AUROC = 0.80). A novel bacterium called Ihubacter massiliensis was also discovered to be a key microbe for TMA/TMAO production by using an OCCT-based humanized gnotobiotic mice model. Simply combining the presence of E. timonensis and I. massiliensis could account for 43% of high TMAO producers with 97% specificity. Collectively, this human gut microbiota phenotype-directed approach offers potential for developing precision medicine and provides insights into translational research. Video Abstract.


Asunto(s)
Carnitina/farmacología , Metilaminas/metabolismo , Microbiota/efectos de los fármacos , Administración Oral , Adulto , Animales , Carnitina/administración & dosificación , Clostridiales/efectos de los fármacos , Clostridiales/metabolismo , Femenino , Humanos , Masculino , Ratones , Microbiota/genética
20.
Sci Rep ; 10(1): 14555, 2020 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-32884086

RESUMEN

The current study aimed to explore whether metformin, the most widely prescribed oral medication for the treatment of type 2 diabetes, alters plasma levels of cardiometabolic disease-related metabolite trimethylamine N-oxide (TMAO) in db/db mice with type 2 diabetes. TMAO plasma concentration was up to 13.2-fold higher in db/db mice when compared to control mice, while in db/db mice fed choline-enriched diet, that mimics meat and dairy product intake, TMAO plasma level was increased 16.8-times. Metformin (250 mg/kg/day) significantly decreased TMAO concentration by up to twofold in both standard and choline-supplemented diet-fed db/db mice plasma. In vitro, metformin significantly decreased the bacterial production rate of trimethylamine (TMA), the precursor of TMAO, from choline up to 3.25-fold in K. pneumoniae and up to 26-fold in P. Mirabilis, while significantly slowing the growth of P. Mirabilis only. Metformin did not affect the expression of genes encoding subunits of bacterial choline-TMA-lyase microcompartment, the activity of the enzyme itself and choline uptake, suggesting that more complex regulation beyond the choline-TMA-lyase is present. To conclude, the TMAO decreasing effect of metformin could be an additional mechanism behind the clinically observed cardiovascular benefits of the drug.


Asunto(s)
Metformina/uso terapéutico , Metilaminas/sangre , Animales , Microbioma Gastrointestinal/efectos de los fármacos , Klebsiella pneumoniae/metabolismo , Masculino , Metilaminas/metabolismo , Ratones , Ratones Endogámicos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA