Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Front Cell Infect Microbiol ; 14: 1389925, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39027133

RESUMEN

Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.


Asunto(s)
Medicamentos Herbarios Chinos , Microbioma Gastrointestinal , Medicina Tradicional China , Microbioma Gastrointestinal/fisiología , Humanos , Medicamentos Herbarios Chinos/farmacología , Animales , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Diabetes Mellitus Tipo 2/microbiología , Diabetes Mellitus Tipo 2/metabolismo , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/tratamiento farmacológico
2.
Front Neurosci ; 18: 1388748, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38650617

RESUMEN

Background: There is evidence of an association between the gut microbiota and progression of stroke. However, the relationship between gut microbial metabolites, specifically bile acids (BAs), and post-ischemic stroke disability and poor functional outcomes remains unexplored. Methods: Patients with acute ischemic stroke (AIS) or transient ischemic attack (TIA) in the Third China National Stroke Registry were grouped according to total bile acid (TBA) quartile on admission. Association of TBA with disability and poor functional outcomes were evaluated using logistic regression models and restricted cubic splines. Results: Data for 9,536 patients were included. After adjusting for confounders, the risks of disability and poor functional outcomes were significantly lower in the highest TBA quartile than in the lowest TBA quartile at the 3-month follow-up, with respective odds ratios (ORs) of 0.65 (95% confidence interval [CI] 0.55-0.78; p < 0.001) and 0.66 (95% CI 0.55-0.78, p < 0.001). Each standard deviation increase in the TBA level reduced the risks of disability and poor functioning outcomes by 10% (adjusted ORs 0.9 [95% CI 0.83-0.98; p = 0.01] and 0.9 [95% CI 0.83-0.97; p < 0.001], respectively). This association remained similar at the 1-year follow-up. After stratification by TOAST subtype, the risk of disability or a poor functional outcome in patients with the large-artery atherosclerosis or "other" subtype was significantly lower in the highest quartile than in the lowest quartile (p < 0.05). Conclusion: Serum TBA is an independent risk factor for disability and poor functional outcomes after AIS or TIA, and exerts a protective effects on brain.

3.
J Cardiothorac Surg ; 19(1): 208, 2024 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-38616256

RESUMEN

BACKGROUND: Cardiac fibroblasts (CFs) are activated after initial injury, and then differentiate into myofibroblasts (MFs), which play a pivotal role as the primary mediator cells in pathological remodeling. Sodium butyrate (NaB), being a metabolite of gut microbiota, exhibits anti-inflammatory property in local therapies on sites other than the intestine. Thus, this study aimed to probe the mechanism by which NaB regulates CFs transdifferentiation through the NLRP3/Caspase-1 pyroptosis pathway. METHODS: CFs were cultured in vitro and induced into MFs by TGFß1. CFs were identified by immunofluorescence labelling technique of vimentin and α-SMA, followed by treatment with NaB or NLRP3 inflammasome inhibitor (CY-09) and its activator [nigericin sodium salt (NSS)]. The expression levels of α-SMA, GSDMD-N/NLRP3/cleaved Caspase-1 proteins, and inflammatory factors IL-1ß/IL-18/IL-6/IL-10 were determined using immunofluorescence, Western blot and ELISA. Cell proliferation and migration were evaluated using the CCK-8 assay and the cell scratch test, respectively. RESULTS: Following the induction of TGFß1, CFs exhibited increased expression levels of α-SMA proteins and IL-6/IL-10, as well as cell proliferative and migratory abilities. TGFß1 induced CFs to differentiate into MFs, while NaB inhibited this differentiation. NaB inactivated the NLRP3/Caspase-1 pyroptosis pathway. CY-09 demonstrated inhibitory effects on the NLRP3/Caspase-1 pyroptosis pathway, leading to a reduction in TGFß1-induced CFs transdifferentiation. NSS activated the NLRP3/Caspase-1 pyroptosis pathway, and thus partially counteracting the inhibitory effect of intestinal microbiota metabolite NaB on CFs transdifferentiation. CONCLUSION: NaB, a metabolite of the gut microbiota, inhibited the activation of the NLRP3/Caspase-1 pyroptosis pathway in TGFß1-induced CFs, repressed the transdifferentiation of CFs into MFs.


Asunto(s)
Microbioma Gastrointestinal , Humanos , Caspasa 1 , Proteína con Dominio Pirina 3 de la Familia NLR , Ácido Butírico , Interleucina-10 , Transdiferenciación Celular , Interleucina-6 , Piroptosis , Fibroblastos
4.
Microbiome ; 12(1): 66, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38549163

RESUMEN

BACKGROUND: Microdeletion of the human chromosomal region 16p11.2 (16p11.2 + / - ) is a prevalent genetic factor associated with autism spectrum disorder (ASD) and other neurodevelopmental disorders. However its pathogenic mechanism remains unclear, and effective treatments for 16p11.2 + / -  syndrome are lacking. Emerging evidence suggests that the gut microbiota and its metabolites are inextricably linked to host behavior through the gut-brain axis and are therefore implicated in ASD development. Despite this, the functional roles of microbial metabolites in the context of 16p11.2 + / -  are yet to be elucidated. This study aims to investigate the therapeutic potential of indole-3-propionic acid (IPA), a gut microbiota metabolite, in addressing behavioral and neural deficits associated with 16p11.2 + / - , as well as the underlying molecular mechanisms. RESULTS: Mice with the 16p11.2 + / -  showed dysbiosis of the gut microbiota and a significant decrease in IPA levels in feces and blood circulation. Further, these mice exhibited significant social and cognitive memory impairments, along with hyperactivation of hippocampal dentate gyrus neurons and reduced inhibitory synaptic transmission in this region. However, oral administration of IPA effectively mitigated the histological and electrophysiological alterations, thereby ameliorating the social and cognitive deficits of the mice. Remarkably, IPA treatment significantly increased the phosphorylation level of ERK1, a protein encoded by the Mapk3 gene in the 16p11.2 region, without affecting the transcription and translation of the Mapk3 gene. CONCLUSIONS: Our study reveals that 16p11.2 + / -  leads to a decline in gut metabolite IPA levels; however, IPA supplementation notably reverses the behavioral and neural phenotypes of 16p11.2 + / -  mice. These findings provide new insights into the critical role of gut microbial metabolites in ASD pathogenesis and present a promising treatment strategy for social and cognitive memory deficit disorders, such as 16p11.2 microdeletion syndrome. Video Abstract.


Asunto(s)
Trastorno del Espectro Autista , Propionatos , Humanos , Ratones , Animales , Transmisión Sináptica , Hipocampo , Indoles
5.
Heliyon ; 9(11): e21431, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38027795

RESUMEN

Oxidative stress is a state of imbalance between oxidation and antioxidation. It is caused by excess levels of free radicals and leads to the damage of DNA, proteins, and lipids. The crucial role of gut microbiota in regulating oxidative stress has been widely demonstrated. Studies have suggested that the redox regulatory effects of gut microbiota are related to gut microbiota metabolites, including fatty acids, lipopolysaccharides, tryptophan metabolites, trimethylamine-N-oxide and polyphenolic metabolites. In recent years, the potential benefits of probiotics have been gaining increasing scientific interest owing to their ability to modulate gut microbiota and oxidative stress. In this review, we summarise the adverse health effects of oxidative stress and discuss the role of the gut microbiota and its metabolites in redox regulation. Based on the influence of gut microbiota metabolites, the roles of probiotics in preventing oxidative stress are highlighted.

6.
Kaohsiung J Med Sci ; 39(10): 1002-1010, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37807941

RESUMEN

Butyrate (BU), a gut microbiota-derived metabolite, has been reported to play a neuroprotective role in Parkinson's disease (PD). However, the specific molecular mechanism of BU has not been fully interpreted. This work aimed to verify the protective effects of BU against MPTP/MPP+ -induced neurotoxicity and explore the mechanisms involved. The results showed that BU protected against MPTP-induced motor dysfunction and decreased tyrosine hydroxylase (TH) and dopamine transporter (DAT) levels. Additionally, BU pretreatment improved PC12 cell viability and reduced MPP+ -induced PC12 cell apoptosis. BU treatment also attenuated MPP+ -stimulated oxidative stress and inflammatory response in PC12 cells. Furthermore, BU inhibited MPTP/MPP+ -induced hyperactivation of the JAK2/STAT3 signaling in mice and PC12 cells. Besides, a JAK2 agonist, Coumermycin A1 (C-A1), substantially reversed BU-mediated inhibition on JAK2/STAT3 phosphorylation in MPP+ -challenged PC12 cells and abated BU-induced repression on MPP+ -triggered apoptosis, oxidative stress, and inflammatory response in PC12 cells. To sum up, BU might exert neuroprotective effects against MPP+ /MPTP-induced neurotoxicity by inactivating the JAK2/STAT3 signaling.


Asunto(s)
Microbioma Gastrointestinal , Intoxicación por MPTP , Fármacos Neuroprotectores , Enfermedad de Parkinson , Ratas , Ratones , Animales , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/metabolismo , Butiratos , Intoxicación por MPTP/tratamiento farmacológico , Intoxicación por MPTP/metabolismo , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Transducción de Señal , Células PC12 , Ratones Endogámicos C57BL
7.
Biomed Pharmacother ; 165: 115243, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37517290

RESUMEN

Brain injury, a common complication in preterm infants, includes the destruction of the key structural and functional connections of the brain and causes neurodevelopmental disorders; it has high morbidity and mortality rates. The exact mechanism underlying brain injury in preterm infants is unclear. Intestinal flora plays a vital role in brain development and the maturation of the immune system in infants; however, detailed understanding of the gut microbiota-metabolite-brain axis in preterm infants is lacking. In this review, we summarise the key mechanisms by which the intestinal microbiota contribute to neurodevelopment and brain injury in preterm infants, with special emphasis on the influence of microorganisms and their metabolites on the regulation of neurocognitive development and neurodevelopmental risks related to preterm birth, infection and neonatal necrotising enterocolitis (NEC). This review provides support for the development and application of novel therapeutic strategies, including probiotics, prebiotics, synbiotics, and faecal bacteria transplantation targeting at brain injury in preterm infants.


Asunto(s)
Lesiones Encefálicas , Microbiota , Nacimiento Prematuro , Probióticos , Femenino , Recién Nacido , Humanos , Recien Nacido Prematuro , Probióticos/uso terapéutico , Lesiones Encefálicas/etiología , Encéfalo
8.
Biochem Biophys Res Commun ; 669: 134-142, 2023 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-37271025

RESUMEN

Non-alcoholic fatty liver disease (NAFLD) has become the most common chronic liver disease worldwide but still lacks specific treatment modalities. The gut microbiota and its metabolites have been shown to be intimately involved in NAFLD development, participating in and regulating disease progression. Trimethylamine N-oxide (TMAO), a metabolite highly dependent on the gut microbiota, has been shown to play deleterious regulatory roles in cardiovascular disease, but the relationship between it and NAFLD lacks validation from basic experiments. This research applied TMAO intervention by constructing fatty liver cell models in vitro to observe its effect on fatty liver cells and potential key genes and performed siRNA interference on the gene to verify the action. The results showed that TMAO intervention promoted the appearance of more red-stained lipid droplets in Oil-red O staining results, increased triglyceride (TG) levels and increased mRNA levels of liver fibrosis-related genes, and also identified one of the key genes, keratin17 (KRT17) via transcriptomics. Following the reduction in its expression level, under the same treatment, there were decreased red-stained lipid droplets, decreased TG levels, decreased indicators of impaired liver function as well as decreased mRNA levels of liver fibrosis-related genes. In conclusion, the gut microbiota metabolite TMAO could promote lipid deposition and fibrosis process via the KRT17 gene in fatty liver cells in vitro.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad del Hígado Graso no Alcohólico , Humanos , Fibrosis , Metilaminas/farmacología , Metilaminas/metabolismo , Cirrosis Hepática , Lípidos
9.
Free Radic Biol Med ; 193(Pt 1): 213-226, 2022 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-36265794

RESUMEN

Metabolic dysfunction-associated fatty liver disease (MAFLD) has become the most common chronic liver disorders in the world, and yet has no approved pharmacotherapy due to the etiology is complex. In the last ten years, increasing evidence have identified the environmental pollutants as risk factors for MAFLD. However, the underlying mechanism remains unclear. Our study found that bromoacetic acid (BAA, a typical kind of environmental toxin) increased triglycerides and total cholesterol levels as well as induced obvious hepatic steatosis and inflammation. The lipidomics showed that ferroptosis was implicated in the environmental toxin-linked MAFLD. Besides, the analysis of microbial metabolomics showed significant change of gut microbiome in BAA groups and the content of gut microbiota metabolite (glycochenodeoxycholate, GCDCA) increased sharply. In vitro study, we observed features of ferroptotic cells by transmission electron microscopy after BAA/GCDCA treatment. Besides, we demonstrated that BAA/GCDCA significantly increased iron contents, with upregulating transferrin receptor (TFR) and acyl-CoA synthetase long-chain family 4 (ACSL4) expression levels. By contrast, iron chelator or silencing TFR relieved BAA/GCDCA-induced lipid metabolism disorder and inflammation. What's more, the interaction between TFR and ACSL4 was also identified. Taken together, we found that, in response to environmental toxin, gut microbiota metabolite GCDCA activates TFR-ACSL4-mediated ferroptosis, which triggered subsequent lipid metabolism disorder and inflammation. Moreover, these findings firstly highlighted the functional relevance among ferroptosis, lipid metabolism and gut microbiota metabolite during environmental pollutant exposure, which shed light on the deep mechanism of environmental toxin-related MAFLD, providing potential targets for the prevention of MAFLD.


Asunto(s)
Ferroptosis , Microbioma Gastrointestinal , Humanos , Ácido Glicoquenodesoxicólico , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo , Receptores de Transferrina , Inflamación
10.
Metab Brain Dis ; 37(7): 2511-2520, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35895243

RESUMEN

Gastrointestinal symptoms are common in the early-stage Parkinson's disease (PD), but its potential pathogenesis remains unclear. Therefore, in the present study, we used the 16S ribosomal RNA gene sequencing and gas chromatography coupled with mass spectrometry-based metabolomics to investigate the alterations of gut microbiome and serum amino acid levels in the early-stage PD mice model induced with rotenone. The results demonstrated that the microbial taxa at phylum, family and genus levels remarkably altered in rotenone-induced mice relative to vehicle-induced mice. The rotenone-induced mice had higher relative abundance of Flavobacteriaceae, Staphylococcaceae, and Prevotellaceae as well as lower relative abundance of Lachnospiraceae_UCG-001, Ruminiclostridium, and Prevotellaceae_NK3B31_group than vehicle-induced mice. The evaluation of serum amino acids revealed the alterations in several classes of amino acids, including L-proline, L-alanine, L-serine, L-asparagine, L-threonine, L-glutamine, L-methionine, and L-4-hydroxyproline. Notably, the altered serum amino acid levels were significantly associated with the abundance of gut microbiota, especially Ruminococcaceae and Ruminiclostridium. Our study explored the possible role of the gut-microbiota-metabolite axis in the early-stage PD and provided the possibility of prevention and treatment of PD by gut-microbiota-metabolite axis in the future.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad de Parkinson , Animales , Ratones , Enfermedad de Parkinson/metabolismo , Rotenona/toxicidad , Glutamina , Hidroxiprolina , Asparagina , Alanina , Metionina , Serina , Treonina
11.
Am Heart J Plus ; 23: 100219, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-38560653

RESUMEN

Study objective: The present systematic review investigates the hypothesis that specific components of the intestinal microbiome and/or their metabolites are associated with early stages of subclinical arterial damage (SAD). Design: Based on the MOOSE criteria, we conducted a systematic review of the literature (Scopus, Medline) investigating the potential association between gut microbiota and the most widely applied arterial biomarkers of SAD. Participants: All studies included individuals without established cardiovascular disease, either with or without SAD. Intervention: No interventions were made. Main outcome measures: Association between exposure (components/metabolites of microbiota) and outcome (presence of SAD). Results: Fourteen articles met the predefined criteria. Due to the large heterogeneity, their meta-analysis was not possible. Our review revealed (a) two studies on endothelial dysfunction, out of which one found an inverse relation between plasma trimethylamine N-oxide levels and FMD and the other did not substantiate a statistically significant correlation with RHI. (b) Twelve studies on atheromatosis, assessed as intimal-medial thickness (IMT), coronary artery calcium (CAC) and arterial plaque, of which, seven studies showed statistically significant associations (negative or positive depending on the microorganism or microbiota metabolite) with IMT, one study revealed significant associations with coronary artery calcium, while one showed absence of correlation and four studies reported statistically significant correlations with arterial plaque. (c) Three studies on arterial stiffness (pulse wave velocity - PWV) with two of them concluding in statistically significant association while the third study did not. Some articles investigated multiple of the correlations described and therefore, belonged to more than one section. Conclusion: Evidence of both positive and inverse associations of gut microbiota composition and their metabolites with different types of SVD has been found. However the small number and heterogeneity of available studies cannot allow to confirm or disprove the hypothesis.

12.
Ann Palliat Med ; 10(11): 11512-11523, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34872276

RESUMEN

BACKGROUND: The gut microbiota-dependent metabolite trimethylamine N-oxide (TMAO) has recently been recognized as one of the novel marker for adverse cardiovascular events and risk of death. However, data on the relationship between TMAO and atrial fibrillation (AF) is limited. The current study was performed to quantify and evaluate the relationship between circulating TMAO levels and AF occurrence. METHODS: The electronic databases PubMed, Cochrane Library, and Embase were systematically searched to March 20, 2021. Research studies were considered that recorded or analyzed the prevalence of AF in individuals in specific populations as well as their circulating TMAO levels. A meta-analysis of two-class variables was used to obtain pooled effects. A dose-response meta-analysis was used to investigate the dose-response relationship between TMAO levels and the risk of AF. RESULTS: Six studies with a total of 8,837 individuals and 1,668 AF cases were included in the present meta-analysis. Compared with a lower circulating TMAO level, a higher TMAO level was associated with a higher prevalence of AF [odds ratio (OR): 1.40; 95% confidence interval (CI): 1.23, 1.59; I2=19.8%]. The dose-response analysis revealed the risk of AF increased by 6% per 1-µmol/L increment (OR: 1.06; 95% CI: 1.00, 1.11), 32% per 5-µmol/L increment (OR: 1.32; 95% CI: 1.03, 1.70), and 73% per 10-µmol/L increment (OR: 1.73; 95% CI: 1.05, 2.86) of the circulating TMAO level. DISCUSSION: This is the first systematic literature review and meta-analysis to demonstrate a significant dose-dependent relationship between increased AF risk and circulating TMAO levels.


Asunto(s)
Fibrilación Atrial , Microbioma Gastrointestinal , Fibrilación Atrial/epidemiología , Humanos , Incidencia , Metilaminas , Factores de Riesgo
13.
Nutr J ; 19(1): 76, 2020 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-32731904

RESUMEN

AIMS: Several epidemiological studies have examined the association between trimethylamine N-Oxide (TMAO) and stroke risk; however, the results are still inconclusive. The purpose of this meta-analysis was to evaluate the relationship between TMAO concentrations and stroke risk. METHODS: PubMed, Scopus, Cochrane and ProQuest search engines were systematically searched up to 18 June 2019. All of the studies that evaluated the relationship between TMAO and stroke were included in the systematic review and eligible studies were included into the meta-analysis. Meta-regression and subgroup analysis were also employed to find the source of heterogeneity. RESULTS: Eight studies (two cross-sectional studies, two cohort studies, three case-control studies and one nested case-control study) with a total of 6150 participants were included in the meta-analysis. The overall result showed that being in the highest category of TMAO increased the odds of stroke by 68% (OR: 1.675; CI: 0.866-3.243; P = 0.047) and mean TMAO concentrations was 2.201 µmol/L higher in patients with stroke rather than non-stroke controls (weighted mean difference (WMD): 2.20; CI: 1.213-3.188; P < 0.001). Furthermore, we observed revealed a non-linear association between increased TMAO levels and increased odds of stroke (P- for nonlinearity < 0.001). In addition, visual inspection of the funnel plot revealed a significant asymmetry among studies examining the differences in TMAO in patients with stroke versus control group. CONCLUSION: This is the first meta-analysis to show positive dose-dependent relations between circulating TMAO concentration and stroke risk. However, further interventional studies and long-term studies are needed to better explain causality.


Asunto(s)
Microbioma Gastrointestinal , Accidente Cerebrovascular , Estudios de Casos y Controles , Estudios Transversales , Humanos , Metilaminas , Factores de Riesgo , Accidente Cerebrovascular/epidemiología
14.
Front Microbiol ; 11: 1450, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32670255

RESUMEN

Radiation therapy is a cornerstone of modern management methods for malignancies but is accompanied by diverse side effects. In the present study, we showed that food additives such as polysorbate 80 (P80) exacerbate irradiation-induced gastrointestinal (GI) tract toxicity. A 16S ribosomal RNA high-throughput sequencing analysis indicated that P80 consumption altered the abundance and composition of the gut microbiota, leading to severe radiation-induced GI tract injury. Mice harboring fecal microbes from P80-treated mice were highly susceptible to irradiation, and antibiotics-challenged mice also represented more sensitive to radiation following P80 treatment. Importantly, butyrate, a major metabolite of enteric microbial fermentation of dietary fibers, exhibited beneficial effects against P80 consumption-aggravated intestinal toxicity via the activation of G-protein-coupled receptors (GPCRs) and maintenance of the intestinal bacterial composition in irradiated animals. Moreover, butyrate had broad therapeutic effects on common radiation-induced injury. Collectively, our findings demonstrate that P80 are potential risk factors for cancer patients during radiotherapy and indicate that butyrate might be employed as a therapeutic option to mitigate the complications associated with radiotherapy.

15.
Nutrition ; 78: 110856, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32592979

RESUMEN

The gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) has been regarded as one of the potent risk factors for cardiovascular events and diabetes. In the current meta-analysis we quantitatively summarized and updated the results of studies regarding the association between TMAO and mortality. A systematic search was performed from PubMed, ProQuest, Scopus, and Embase. All of the studies that evaluated the association between TMAO and mortality were included in the systematic review and meta-analysis. Subgroup analysis and meta-regression were performed to identify the source of heterogeneity. There were 31 230 participants included and the results showed that being in the highest category of TMAO increased the hazard ratio (HR) of mortality by 47%. Moreover, there was a non-linear association between increased TMAO concentrations and HR of mortality. In the current dose-response meta-analysis, we revealed a positive association between TMAO and mortality risk among an adult population.


Asunto(s)
Microbioma Gastrointestinal , Adulto , Humanos , Metilaminas , Modelos de Riesgos Proporcionales , Factores de Riesgo
16.
Microbiome ; 8(1): 69, 2020 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-32434586

RESUMEN

BACKGROUND: We have proved fecal microbiota transplantation (FMT) is an efficacious remedy to mitigate acute radiation syndrome (ARS); however, the mechanisms remain incompletely characterized. Here, we aimed to tease apart the gut microbiota-produced metabolites, underpin the therapeutic effects of FMT to radiation injuries, and elucidate the underlying molecular mechanisms. RESULTS: FMT elevated the level of microbial-derived indole 3-propionic acid (IPA) in fecal pellets from irradiated mice. IPA replenishment via oral route attenuated hematopoietic system and gastrointestinal (GI) tract injuries intertwined with radiation exposure without precipitating tumor growth in male and female mice. Specifically, IPA-treated mice represented a lower system inflammatory level, recuperative hematogenic organs, catabatic myelosuppression, improved GI function, and epithelial integrity following irradiation. 16S rRNA gene sequencing and subsequent analyses showed that irradiated mice harbored a disordered enteric bacterial pattern, which was preserved after IPA administration. Notably, iTRAQ analysis presented that IPA replenishment retained radiation-reprogrammed protein expression profile in the small intestine. Importantly, shRNA interference and hydrodynamic-based gene delivery assays further validated that pregnane X receptor (PXR)/acyl-CoA-binding protein (ACBP) signaling played pivotal roles in IPA-favored radioprotection in vitro and in vivo. CONCLUSIONS: These evidences highlight that IPA is a key intestinal microbiota metabolite corroborating the therapeutic effects of FMT to radiation toxicity. Owing to the potential pitfalls of FMT, IPA might be employed as a safe and effective succedaneum to fight against accidental or iatrogenic ionizing ARS in clinical settings. Our findings also provide a novel insight into microbiome-based remedies toward radioactive diseases. Video abstract.


Asunto(s)
Inhibidor de la Unión a Diazepam , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal , Indoles , Traumatismos por Radiación , Animales , Línea Celular , Inhibidor de la Unión a Diazepam/metabolismo , Heces/química , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de la radiación , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Hematopoyesis/efectos de los fármacos , Indoles/administración & dosificación , Indoles/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Receptor X de Pregnano/metabolismo , ARN Ribosómico 16S/genética , Traumatismos por Radiación/terapia , Transducción de Señal/efectos de los fármacos
17.
Crit Rev Food Sci Nutr ; 60(16): 2801-2823, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32462890

RESUMEN

The gut microbiota-derived metabolite trimethylamine N-oxide (TMAO) has been regarded as one of the potent risk factors of cardiovascular events and diabetes. However, its association with possible inflammatory mediators has not been revealed yet. In the current meta-analysis, we quantitatively summarized the results of studies regarding the association between TMAO and inflammation. Electronic databases including PubMed, ProQuest, Scopus, and Embase were systematically searched and a total of 586 manuscripts were retrieved. After removing 223 duplicates, 363 manuscripts were reviewed. All of the studies regarding the association between TMAO and inflammatory factors were included in the systematic review and eligible studies were included in to the meta-analysis. Accordingly, 13,783 number of participants were included and the results showed that being in the highest category of TMAO Accordingly was associated with 0.27 mg/L (weighted mean difference: 0.268; 95% confidence interval [CI]: 0.058-0.479; p = 0.013) increase in CRP concentrations compared with lowest category. The results of subgrouping and meta-regression revealed the location, CRP sample source, disease status, male percent, proportion of diabetes and smoking as the source of heterogeneity. Moreover, the dose-response meta-analysis revealed a non-linear association between increased TMAO concentrations and increased CRP concentrations (p for nonlinearity = 0.015). To our knowledge, this is first dose-response meta-analysis that summarized the results of studies about the association between circulating TMAO concentrations and inflammation risk.


Asunto(s)
Microbioma Gastrointestinal/fisiología , Inflamación/metabolismo , Metilaminas/metabolismo , Humanos , Factores de Riesgo
18.
Artículo en Inglés | MEDLINE | ID: mdl-32193004

RESUMEN

Short-chain fatty acids (SCFAs) are gut microbiota metabolites recognized for their beneficial effects on the host organism. In this study, a simple and rapid sample preparation method combined to SCFAs analysis by direct injection and gas chromatography coupled with flame ionization detection (GC-FID), for the determination and quantification of eight SCFAs (acetic, propionic, i-butyric, butyric, i-valeric, valeric, i-caproic and caproic acids) in rat, mice and human faeces and in fermentation fluids samples, has been developed and validated. The method consists of extraction of the SCFAs by ethyl ether after acidification of the samples. The effect of the number of extractions has been assessed in order to optimize the procedure and to obtain a satisfactory yield for all the analyzed SCFAs. The increase of the extracted analytes quantity was significant passing from 1 to 2 and from 2 to 3 extractions (P < 0.05), while no significant differences were found performing 3, 4 or 5 extractions (P > 0.05). The SCFAs extracted are directly analyzed by GC-FID without derivatization and separated on a polyethylene glycol nitroterephthalic acid modified coated capillary column, with a chromatographic run time of 13 min. The proposed method showed good sensitivity, with limits of quantifications in the range 0.14-0.48 µM for SCFAs from propionic to caproic acids and 2.12 µM for acetic acid; recovery was between 80.8 and 108.8% and intraday and interday repeatability in the range 0.6-5.0% of precision (RSD, %) The optimized method is suitable for the quantitative analysis of SCFAs in real samples of rat, mouse and human faeces and in fermentation fluids, and it can be applied also to very small amount of faecal sample (20 mg).


Asunto(s)
Mezclas Complejas/análisis , Ácidos Grasos Volátiles/análisis , Ácidos Grasos Volátiles/metabolismo , Heces/química , Animales , Cromatografía de Gases , Mezclas Complejas/metabolismo , Éter/química , Fermentación , Microbioma Gastrointestinal , Humanos , Límite de Detección , Metabolómica/métodos , Ratones , Polietilenglicoles/química , Ratas , Reproducibilidad de los Resultados
19.
Nutr Metab (Lond) ; 15: 81, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30479648

RESUMEN

BACKGROUND: Evidence has suggested a potential link exists between trimethylamine-N-oxide (TMAO), a choline-derived metabolite produced by gut microbiota, and some cancers, but little is known for primary liver cancer (PLC). METHODS: A case-control study was designed including 671 newly diagnosed PLC patients and 671 control subjects frequency-matched by age (±5 years) and sex, in Guangdong province, China. High-performance liquid chromatography with online electrospray ionization tandem mass spectrometry (HPLC-MS/MS) was used to measure serum TMAO and choline. The associations between these biomarkers and PLC risk were evaluated using logistic regression models. RESULTS: Serum TMAO concentrations were greater in the PLC group than the control group (P = 0.002). Logistic regression analysis showed that the sex- and age-adjusted odds ratio (OR) and (95% confidence interval [CI]) was 3.43 (2.42-4.86) when comparing the top and bottom quartiles (Q4 vs Q1). After further adjusting for more selected confounders, the OR (95% CI) remained significant but was attenuated to 2.85 (1.59-5.11) (Q4 vs Q1). The multivariable-adjusted ORs (95% CIs) across quartiles of choline were 0.35-0.15 (P -trend < 0.001). CONCLUSION: Higher serum levels of TMAO were associated with increased PLC risk. The association was stronger in those with lower serum levels of choline. Additional large prospective studies are required to confirm these findings. TRIAL REGISTRATION: This study was registered at clinicaltrials.gov as NCT 03297255.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA