Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 183
1.
Am Heart J ; 264: 40-48, 2023 10.
Article En | MEDLINE | ID: mdl-37301317

Heart failure (HF) is a leading cause of death worldwide despite recent advances in pharmacological treatments. Gut microbiota dysbiosis and gut barrier dysfunction with consequent bacterial translocation and increased blood endotoxemia has gained much attention as one of the key pathogenetic mechanisms contributing to increased mortality of patients at risk or with cardiovascular disease. Indeed, increased blood levels of lipopolysaccharide (LPS), a glycolipid of outer membrane of gut gram-negative bacteria, have been detected in patients with diabetes, obesity and nonalcoholic fatty liver disease or in patients with established coronary disease such as myocardial infarction or atrial fibrillation, suggesting endotoxemia as aggravating factor via systemic inflammation and eventually vascular damage. Upon interaction with its receptor Toll-like receptor 4 (TLR4) LPS may, in fact, act at different cellular levels so eliciting formation of proinflammatory cytokines or exerting a procoagulant activity. Increasing body of evidence pointed to endotoxemia as factor potentially deteriorating the clinical course of patients with HF, that, in fact, is associated with gut dysbiosis-derived changes of gut barrier functionality and eventually bacteria or bacterial product translocation into systemic circulation. The aim of this review is to summarize current experimental and clinical evidence on the mechanisms linking gut dysbiosis-related endotoxemia with HF, its potential negative impact with HF progression, and the therapeutic strategies that can counteract endotoxemia.


Endotoxemia , Heart Failure , Humans , Endotoxemia/complications , Endotoxemia/microbiology , Lipopolysaccharides/therapeutic use , Dysbiosis/complications , Obesity/complications , Heart Failure/complications
2.
Nutrients ; 13(8)2021 Aug 15.
Article En | MEDLINE | ID: mdl-34444955

Diet and dietary components have profound effects on the composition of the gut microbiota and are among the most important contributors to the alteration in bacterial flora. This review examines the effects the "Western", "plant-based", "high-fat", "medical ketogenic", and "Mediterranean" diets have on the composition of the gut microbiota in both mice and human subjects. We show that specific dietary components that are commonly found in the "plant-based" and "Mediterranean" diet play a role in shifting the microbial composition. This review further evaluates the bacterial metabolites that are associated with diet, and their role in systemic inflammation and metabolic endotoxemia. Furthermore, the associations between diet/dietary components and altering bacterial composition, may lead to potential therapeutic targets for type II diabetes, obesity, and inflammatory diseases.


Diet/adverse effects , Gastrointestinal Microbiome/physiology , Nutritional Physiological Phenomena , Animals , Diet/methods , Diet, High-Fat/adverse effects , Diet, Ketogenic/adverse effects , Diet, Mediterranean/adverse effects , Diet, Vegetarian/adverse effects , Diet, Western/adverse effects , Endotoxemia/etiology , Endotoxemia/microbiology , Humans , Inflammation/etiology , Inflammation/microbiology , Mice
3.
Biomed Res Int ; 2021: 5554991, 2021.
Article En | MEDLINE | ID: mdl-34337024

BACKGROUND: Obesity is a main contributing factor for the development of glucose intolerance and type 2 diabetes mellitus (T2D). Roux-en-Y gastric bypass (RYGB) is believed to be one of the most effective treatments to reduce body weight and improve glucose metabolism. In this study, we sought to explore the underlying mechanisms of weight reduction and insulin resistance improvement after RYGB. METHODS: This was a prospective observational study using consecutive samples of 14 obese subjects undergoing bariatric surgery. Main assessments were serum indexes (blood metabolites, glucose-lipid regulating hormones, trimethylamine-N-oxide (TMAO), and lipopolysaccharide-binding protein (LBP), fecal short-chain fatty acids (SCFAs), and gut microbiota. Correlation analysis of the factors changed by RYGB was used to indicate the potential mechanism by which surgery improves insulin resistance. RESULTS: The subjects showed significant improvement on indices of obesity and insulin resistance and a correlated change of gut microbiota components at 1 month, 3 months, and 6 months post-RYGB operation. In particular, the abundance of a counterobese strain, Akkemansia muciniphila, had gradually increased with the postoperative time. Moreover, these changes were negatively correlated to serum levels of LBP and positively correlated to serum TMAO and fecal SCFAs. CONCLUSIONS: Our findings uncovered links between intestinal microbiota alterations, circulating endotoxemia, and insulin resistance. This suggests that the underlying mechanism of protection of the intestine by RYGB in obesity may be through changing the gut microbiota.


Endotoxemia/microbiology , Endotoxemia/surgery , Gastric Bypass , Gastrointestinal Microbiome , Insulin Resistance , Acute-Phase Proteins/metabolism , Carrier Proteins/metabolism , Down-Regulation , Humans , Membrane Glycoproteins/metabolism , Metabolome , Methylamines/metabolism , Obesity/microbiology , Obesity/surgery
4.
Eur J Pharmacol ; 909: 174438, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34437885

Increasing evidence indicates that patients or experimental animals exposure to endotoxin (lipopolysaccharides, LPS) exert deleterious cardiac functions that greatly contribute to morbidity and mortality. The pathophysiologic processes, including NLRP3 inflammasome overactivation and cardiac inflammatory injury, are complicated. Sodium tanshinone IIA sulfonate (STS), a water-soluble derivative of tanshinone IIA, is a naturally occurring compound extracted from Salvia miltiorrhiza and has anti-inflammatory and cardioprotective properties. In this study we examined the effect of STS on endotoxin-induced cardiomyopathy and investigated the underlying mechanisms. An endotoxemic mouse model was established by injecting LPS (10 mg/kg). Different doses of STS were administered intraperitoneally (5, 10, or 50 mg/kg) at different time points (2/12 h, 4/12 h, and 8/12 h) after LPS challenge to assess its effect on survival of mice with endotoxemia. In parallel, cardiac function, myocardial inflammatory cytokines, cardiomyocyte pyroptosis and autophagy were evaluated to determine the extent of myocardial damage due to sepsis in the presence and absence of STS at the optimal dose (10 mg/kg) and time-point (2/12 h). The results demonstrated that STS increased the survival rates, improved the compromised cardiac function and reduced myocardial inflammatory injury associated with enhanced autophagy and mitigated NLRP3 inflammasome activation. Moreover, inhibiting of autophagy or blocking the AMPK pathway reversed STS-elicited prevention of cardiomyopathy and activated the NLRP3 inflammasome in endotoxemic mice. Collectively, our study demonstrates that STS attenuates endotoxemia-induced mortality and cardiomyopathy, which may be associated with promotion of autophagy and inhibition of NLRP3 inflammasome overactivation.


Cardiomyopathies/prevention & control , Endotoxemia/drug therapy , Inflammasomes/antagonists & inhibitors , Phenanthrenes/pharmacology , Animals , Autophagy/drug effects , Autophagy/immunology , Cardiomyopathies/diagnosis , Cardiomyopathies/immunology , Cardiomyopathies/microbiology , Disease Models, Animal , Echocardiography , Endotoxemia/complications , Endotoxemia/immunology , Endotoxemia/microbiology , Endotoxins/blood , Endotoxins/immunology , Heart Ventricles/diagnostic imaging , Heart Ventricles/drug effects , Heart Ventricles/immunology , Heart Ventricles/pathology , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Male , Mice , Myocytes, Cardiac , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Phenanthrenes/therapeutic use , Pyroptosis/drug effects , Pyroptosis/immunology
5.
Nutr Res ; 91: 26-35, 2021 07.
Article En | MEDLINE | ID: mdl-34130208

Current evidence suggests that high fructose intake results in gut dysbiosis, leading to endotoxemia and NAFLD onset. Thus, the hypothesis of the study was that an enhanced Proteobacteria proportion in the cecal microbiota could be the most prominent trigger of NAFLD through enhanced endotoxin (LPS) in adult high-fructose-fed C57BL/6 mice. Male C57BL/6 mice received a control diet (n = 10, C: 76% of energy as carbohydrates, 0% as fructose) or high-fructose diet (n = 10, HFRU: 76% of energy as carbohydrate, 50% as fructose) for 12 weeks. Outcomes included biochemical analyses, 16S rDNA PCR amplification, hepatic stereology, and RT-qPCR. The groups showed similar body masses during the whole experiment. However, the HFRU group showed greater water intake and blood pressure than the C group. The HFRU group showed a significantly lower amount of Bacteroidetes and a predominant rise in Proteobacteria, implying increased LPS. The HFRU group also showed enhanced de novo lipogenesis (Chrebp expression), while beta-oxidation was decreased (Ppar-alpha expression). These results agree with the deposition of fat droplets within hepatocytes and the enhanced hepatic triacylglycerol concentrations, as observed in the photomicrographs, where the HFRU group had a higher volume density of steatosis than the C group. Thus, we confirmed that a rise in the Proteobacteria phylum proportion was the most prominent alteration in gut-liver axis-induced hepatic steatosis in HFRU-fed C57BL/6 mice. Gut dysbiosis and fatty liver were observed even in the absence of overweight in this dietary adult mouse model.


Diet/adverse effects , Dysbiosis/microbiology , Fructose/adverse effects , Gastrointestinal Microbiome , Liver , Non-alcoholic Fatty Liver Disease/microbiology , Proteobacteria/growth & development , Animals , Body Weight , Cecum/microbiology , Dietary Sugars/adverse effects , Disease Models, Animal , Dysbiosis/etiology , Endotoxemia/etiology , Endotoxemia/microbiology , Feeding Behavior , Lipid Metabolism , Lipopolysaccharides , Liver/metabolism , Male , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Triglycerides/metabolism
6.
J Zoo Wildl Med ; 52(2): 755-762, 2021 Jun.
Article En | MEDLINE | ID: mdl-34130423

A multiparous pygmy hippopotamus (Choeropsis liberiensis) dam produced three consecutive calves that died acutely at 13-15 wk of age from bacterial sepsis, for which diagnostic and therapeutic intervention was not possible. Streptococcus iniae (Cases 1 and 3), Escherichia coli (Case 2), and an unidentified member of the family Pasteurellaceae (Case 1) were identified in postmortem tissues through bacterial culture followed by standard and molecular identification methods. After the loss of two calves, a series of vaccinations were administered to the dam during the third pregnancy to enhance transplacental and colostral transfer of antibodies to the calf. The third calf did not survive, and the source of the bacterial infection in these three calves was undetermined. Prior to and after the birth of the fourth calf, nutritional and nutraceutical supplements were provided to the dam and calf. Additionally, pest control around the barn was enhanced. The fourth calf survived. Pygmy hippopotamus calves at the age of 13-15 wk may have increased susceptibility to bacterial infection, possibly due to waning maternally derived immunity. The findings in these cases, combined with a previous association of S. iniae in pygmy hippopotamus deaths, suggest that this bacterium is an especially important pathogen of the endangered pygmy hippopotamus.


Artiodactyla , Bacterial Infections/veterinary , Endotoxemia/veterinary , Escherichia coli Infections/veterinary , Sepsis/veterinary , Streptococcal Infections/veterinary , Animal Husbandry , Animals , Animals, Zoo , Bacterial Infections/prevention & control , Bacterial Vaccines/immunology , Endotoxemia/microbiology , Escherichia coli , Escherichia coli Infections/pathology , Female , Male , Sepsis/microbiology , Streptococcal Infections/microbiology , Streptococcal Infections/pathology , Streptococcus iniae
7.
Int J Mol Sci ; 22(7)2021 Mar 28.
Article En | MEDLINE | ID: mdl-33800707

Over unimaginable expanses of evolutionary time, our gut microbiota have co-evolved with us, creating a symbiotic relationship in which each is utterly dependent upon the other. Far from confined to the recesses of the alimentary tract, our gut microbiota engage in complex and bi-directional communication with their host, which have far-reaching implications for overall health, wellbeing and normal physiological functioning. Amongst such communication streams, the microbiota-gut-brain axis predominates. Numerous complex mechanisms involve direct effects of the microbiota, or indirect effects through the release and absorption of the metabolic by-products of the gut microbiota. Proposed mechanisms implicate mitochondrial function, the hypothalamus-pituitary-adrenal axis, and autonomic, neuro-humeral, entero-endocrine and immunomodulatory pathways. Furthermore, dietary composition influences the relative abundance of gut microbiota species. Recent human-based data reveal that dietary effects on the gut microbiota can occur rapidly, and that our gut microbiota reflect our diet at any given time, although much inter-individual variation pertains. Although most studies on the effects of dietary macronutrients on the gut microbiota report on associations with relative changes in the abundance of particular species of bacteria, in broad terms, our modern-day animal-based Westernized diets are relatively high in fats and proteins and impoverished in fibres. This creates a perfect storm within the gut in which dysbiosis promotes localized inflammation, enhanced gut wall permeability, increased production of lipopolysaccharides, chronic endotoxemia and a resultant low-grade systemic inflammatory milieu, a harbinger of metabolic dysfunction and many modern-day chronic illnesses. Research should further focus on the colony effects of the gut microbiota on health and wellbeing, and dysbiotic effects on pathogenic pathways. Finally, we should revise our view of the gut microbiota from that of a seething mass of microbes to one of organ-status, on which our health and wellbeing utterly depends. Future guidelines on lifestyle strategies for wellbeing should integrate advice on the optimal establishment and maintenance of a healthy gut microbiota through dietary and other means. Although we are what we eat, perhaps more importantly, we are what our gut microbiota thrive on and they thrive on what we eat.


Brain/physiology , Diet , Gastrointestinal Microbiome , Intestines/innervation , Intestines/physiology , Animals , Appetite , Autonomic Nervous System/embryology , Brain/metabolism , Diet, High-Fat , Dietary Fats , Dysbiosis/microbiology , Endotoxemia/microbiology , Humans , Incretins/metabolism , Inflammation , Lipopolysaccharides , Mice , Mitochondria/metabolism , Oligosaccharides/chemistry , Permeability
8.
Mol Nutr Food Res ; 65(1): e1900481, 2021 01.
Article En | MEDLINE | ID: mdl-33111450

SCOPE: More than a decade ago, the concept of "metabolic endotoxemia" is elaborated on the fact that some bacterial components, classified as microbial associated membrane pathogens (MAMPs) can pass through the gut barrier and create a systemic low tone inflammation. METHODS AND RESULTS: The translocation of lipopolysaccharides and its contribution to systemic inflammation are largely studied in murine models of obesity, allowing to unravel the molecular pathways involved in the process. Many different pathological contexts evoke the loss of gut barrier as an event contributing to inflammation and thereby driving metabolic and behavioral alterations. CONCLUSION: This review describes the role of nutrition as a modulator of metabolic regulation and focuses on the contribution of the gut microbiota in the process of the production of a large diversity of bioactive metabolites. The two first sections of the review will be dedicated to the impact of nutritional disorders on both the gut microbiota composition and on metabolic inflammation. The last and more prominent section will describe the role of different nutrient-derived gut metabolites on the gut barrier integrity, metabolic inflammation, and peripheral tissue alterations during obesity or associated complications.


Gastrointestinal Microbiome/physiology , Inflammation/etiology , Nutrition Disorders/microbiology , Obesity/microbiology , Animals , Cholesterol/metabolism , Dietary Carbohydrates/pharmacokinetics , Dysbiosis/etiology , Endocannabinoids/metabolism , Endotoxemia/microbiology , Humans , Lipopolysaccharides/metabolism , Mice , Nutrition Disorders/etiology , Obesity/complications , Polyphenols/pharmacokinetics , Toll-Like Receptors/metabolism
9.
Artif Organs ; 45(6): E187-E194, 2021 Jun.
Article En | MEDLINE | ID: mdl-33377184

The coronavirus disease 2019 (COVID-19) has been shown to involve the gastrointestinal tract, which implies bacterial translocation and endotoxemia. The aim of this study was to evaluate the role of extracorporeal endotoxin removal by Polymyxin B hemoperfusion (PMX-HP), in the treatment of patients with COVID-19 and secondary bacterial infection. We conducted a subgroup analysis of a multicenter, multinational, prospective, and observational web-based database (EUPHAS2 registry). We included 12 patients with severe acute respiratory syndrome coronavirus 2 infection confirmed by real-time reverse transcriptase-polymerase chain reaction from nasal/oral swab, admitted to the intensive care unit between February and May 2020, who were affected by septic shock and received PMX-HP as per clinical indication of the attending physician. Septic shock was diagnosed in nine patients (75%), with a median time between symptoms onset and PMX-HP treatment of 16 (14-22) days. We identified Gram-negative bacteria in most of the microbiological cultures (N = 17, 65%), followed by Gram-positive bacteria in (N = 4, 15%), fungi (N = 3, 12%) and no growth (N = 2, 8%). Sequential Organ Failure Assessment (SOFA) score progressively improved over the next 120 hours following PMX-HP and it was associated with median endotoxin activity assay (EAA) decrease from 0.78 [0.70-0.92] at T0 to 0.60 [0.44-0.72] at T120 (P = .245). A direct correlation was observed between SOFA score and EAA. Lung Injury Score decreased and was associated with hemodynamic improvement over the same period. No statistically significant difference was observed for RIFLE score at each time point. Nine out of 12 patients (75%) required continuous renal replacement therapy because of acute kidney injury. In a series of consecutive COVID-19 patients with endotoxic shock, PMX-HP was associated with organ function recovery, hemodynamic improvement, and contemporary EAA level reduction. No PMX-HP-related complications were observed.


Anti-Bacterial Agents/therapeutic use , COVID-19/complications , Endotoxemia/drug therapy , Endotoxemia/microbiology , Polymyxin B/therapeutic use , Shock, Septic/drug therapy , Shock, Septic/microbiology , Anti-Bacterial Agents/administration & dosage , Biomarkers/blood , COVID-19/mortality , Critical Illness , Endotoxemia/mortality , Female , Hospital Mortality , Humans , Male , Middle Aged , Organ Dysfunction Scores , Prospective Studies , Registries , SARS-CoV-2 , Shock, Septic/mortality
10.
J Agric Food Chem ; 68(42): 11710-11725, 2020 Oct 21.
Article En | MEDLINE | ID: mdl-33034193

The responses of gut microbiota to dietary proteins have been studied previously. However, the effects of dietary proteins supplemented with a high-fat diet (HFD) on the metabolite biomarkers associated with non-alcoholic fatty liver disease (NAFLD) are not well understood. To understand the underlying mechanisms, C57BL/6J mice were fed with either a low-fat diet with casein (LFC) or an HFD with casein (HFC), fish (HFF), or mutton proteins (HFM), and their cecal microbiota and liver metabolites were analyzed. At the phylum level, the HFD group had a relatively higher abundance of Firmicutes compared to the LFC-diet group. At the genus level, the HFF-diet group had the highest abundance of Lactobacillus and Akkermansia compared to the HFC- and HFM-diet groups. Furthermore, mice fed with the HFF diet had significantly reduced levels of hepatic metabolites involved in oxidative stress and bile acid metabolism. Thus, meat proteins in HFD interact in the host to create distinct responses in the gut microbiota and its metabolites.


Dietary Proteins/adverse effects , Endotoxemia/immunology , Gastrointestinal Microbiome , NF-kappa B/immunology , Toll-Like Receptor 4/immunology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/growth & development , Bacteria/isolation & purification , Bile Acids and Salts/metabolism , Diet, High-Fat/adverse effects , Dietary Proteins/metabolism , Endotoxemia/etiology , Endotoxemia/metabolism , Endotoxemia/microbiology , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/genetics , Oxidative Stress , Toll-Like Receptor 4/genetics
11.
Gut Microbes ; 12(1): 1801301, 2020 11 09.
Article En | MEDLINE | ID: mdl-32804018

Fat and sweeteners contribute to obesity. However, it is unknown whether specific bacteria are selectively modified by different caloric and noncaloric sweeteners with or without a high-fat diet (HFD). Here, we combined extensive host phenotyping and shotgun metagenomics of the gut microbiota to investigate this question. We found that the type of sweetener and its combination with an HFD selectively modified the gut microbiota. Sucralose and steviol glycosides led to the lowest α-diversity of the gut microbiota. Sucralose increased the abundance of B. fragilis in particular, resulting in a decrease in the abundance of occludin and an increase in proinflammatory cytokines, glucose intolerance, fatty acid oxidation and ketone bodies. Sucrose+HFD showed the highest metabolic endotoxemia, weight gain, body fat, total short chain fatty acids (SCFAs), serum TNFα concentration and glucose intolerance. Consumption of sucralose or sucrose resulted in enrichment of the bacterial genes involved in the synthesis of LPS and SCFAs. Notably, brown sugar and honey were associated with the absence of metabolic endotoxemia, increases in bacterial gene diversity and anti-inflammatory markers such as IL-10 and sIgA, the maintenance of glucose tolerance and energy expenditure, similar to the control group, despite the consumption of an HFD. These findings indicate that the type of sweetener and an HFD selectively modify the gut microbiota, bacterial gene enrichment of metabolic pathways involved in LPS and SCFA synthesis, and metabolic endotoxemia associated with different metabolic profiles.


Endotoxemia/etiology , Fatty Acids/adverse effects , Sweetening Agents/adverse effects , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Bacteria/metabolism , Diet, High-Fat/adverse effects , Endotoxemia/metabolism , Endotoxemia/microbiology , Fatty Acids/metabolism , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome , Humans , Male , Rats , Rats, Wistar , Sweetening Agents/metabolism
12.
Pharmacol Res ; 161: 105135, 2020 11.
Article En | MEDLINE | ID: mdl-32814166

The intestinal epithelial layer serves as a physical and functional barrier between the microbe-rich lumen and immunologically active submucosa; it prevents systemic translocation of microbial pyrogenic products (e.g. endotoxin) that elicits immune activation upon translocation to the systemic circulation. Loss of barrier function has been associated with chronic 'low-grade' systemic inflammation which underlies pathogenesis of numerous no-communicable chronic inflammatory disease. Thus, targeting gut barrier dysfunction is an effective strategy for the prevention and/or treatment of chronic disease. This review intends to emphasize on the beneficial effects of herbal formulations, phytochemicals and traditional phytomedicines in attenuating intestinal barrier dysfunction. It also aims to provide a comprehensive understanding of intestinal-level events leading to a 'leaky-gut' and systemic complications mediated by endotoxemia. Additionally, a variety of detectable markers and diagnostic criteria utilized to evaluate barrier improving capacities of experimental therapeutics has been discussed. Collectively, this review provides rationale for targeting gut barrier dysfunction by phytotherapies for treating chronic diseases that are associated with endotoxemia-induced systemic inflammation.


Anti-Inflammatory Agents/therapeutic use , Endotoxemia/drug therapy , Gastrointestinal Agents/therapeutic use , Inflammatory Bowel Diseases/drug therapy , Intestinal Mucosa/drug effects , Phytotherapy , Plant Extracts/therapeutic use , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/isolation & purification , Bacteria/immunology , Bacteria/metabolism , Chronic Disease , Dysbiosis , Endotoxemia/metabolism , Endotoxemia/microbiology , Endotoxemia/pathology , Endotoxins/metabolism , Gastrointestinal Agents/adverse effects , Gastrointestinal Agents/isolation & purification , Gastrointestinal Microbiome , Host-Pathogen Interactions , Humans , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Intestinal Mucosa/pathology , Permeability , Plant Extracts/adverse effects , Plant Extracts/isolation & purification
13.
Am J Hypertens ; 33(10): 890-901, 2020 10 21.
Article En | MEDLINE | ID: mdl-32614942

There are numerous studies indicating a direct association between hypertension and gut microbiota in both animal models and humans. In this review, we focused on the imbalance in the gut microbiota composition relative to healthy state or homeostasis, termed dysbiosis, associated with hypertension and discuss the current knowledge regarding how microbiota regulates blood pressure (BP), involving the sympathetic nervous system and the immune system. The profile of ecological parameters and bacterial genera composition of gut dysbiosis in hypertension varies according to the experimental model of hypertension. Recent evidence supports that gut microbiota can protect or promote the development of hypertension by interacting with gut secondary lymph organs and altering T helper 17/regulatory T cells polarization, with subsequent changes in T cells infiltration in vascular tissues. Here, we also describe the bidirectional communication between the microbiome and the host via the sympathetic nervous system and its role in BP regulation. Dysbiosis in hypertension is mainly associated with reduced proportions of short-chain fatty acid-producing bacteria, mainly acetate- and butyrate-producing bacteria, and an increased enrichment of the genes for lipopolysaccharide biosynthesis and export, lending to moderate endotoxemia. The role of these metabolic and structural products in both immune and sympathetic system regulation and vascular inflammation was also analyzed. Overall, gut microbiota is now recognized as a well-established target to dietary interventions with prebiotics or probiotics to reduce BP.


Dysbiosis/immunology , Gastrointestinal Microbiome/immunology , Hypertension/immunology , Inflammation/immunology , Sympathetic Nervous System/physiopathology , Animals , Dysbiosis/physiopathology , Endotoxemia/immunology , Endotoxemia/microbiology , Endotoxemia/physiopathology , Gene Expression Regulation/immunology , Humans , Hypertension/microbiology , Hypertension/physiopathology , Inflammation/microbiology , Inflammation/physiopathology , Lipopolysaccharides/biosynthesis , Lipopolysaccharides/metabolism , Sympathetic Nervous System/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology
14.
Acta Vet Hung ; 68(1): 53-58, 2020 03.
Article En | MEDLINE | ID: mdl-32384071

The objective of this study was to examine whether serum iron (Fe) concentration is useful as a prognostic biomarker for cows with acute coliform mastitis (ACM). Our study was composed of determining the reproducibility of serum Fe concentration as a prognostic criterion in cows with ACM (Study 1) and clarifying the sequential changes in serum Fe concentration in cattle that received endotoxin (Study 2). Seventy-seven cows with (n = 47) or without (n = 30) ACM were enrolled in Study 1. The proposed diagnostic cut-off value of serum Fe concentration indicating a poor prognosis of ACM based on the analysis of the receiver operating characteristic curves was < 31.5 µg/dL. Ten young cattle aged 176.8 ± 23.7 days were enrolled in Study 2. Five young cattle received endotoxin (LPS group) and the remaining five received physiological saline (control group). Blood collections were carried out before endotoxin challenge (pre), and 0.5, 1, 2, 4, 8, 12, 24, and 48 h after the challenge. As a result, a significant decrease in serum Fe concentration was not observed until 24 h after endotoxin challenge. Because in cows with clinical ACM it is difficult to know the time course after infection, the alteration in serum Fe concentrations alone may be an insufficient prognostic criterion.


Endotoxemia/veterinary , Escherichia coli Infections/veterinary , Iron/blood , Klebsiella Infections/veterinary , Mastitis, Bovine/diagnosis , Acute Disease , Animals , Biomarkers/blood , Cattle , Endotoxemia/diagnosis , Endotoxemia/microbiology , Endotoxins/administration & dosage , Escherichia coli/physiology , Escherichia coli Infections/complications , Escherichia coli Infections/diagnosis , Escherichia coli Infections/microbiology , Female , Klebsiella Infections/complications , Klebsiella Infections/diagnosis , Klebsiella Infections/microbiology , Klebsiella pneumoniae/physiology , Mastitis, Bovine/microbiology , Prognosis , Reproducibility of Results
15.
Bull Exp Biol Med ; 168(4): 435-438, 2020 Feb.
Article En | MEDLINE | ID: mdl-32146625

We studied the levels endotoxin and microbial markers in the blood of female rats with experimental heart failure and the effects of preliminary treatment with a prebiotic complex based on fermented wheat bran and inactivated Saccharomyces cerevisiae culture on these parameters. The concentrations of endotoxin, markers of lactobacilli, and opportunistic microorganisms were found to increase in rats with experimental heart failure and significantly decreased against the background of treatment with prebiotic complex. The dynamics of markers of bifidobacteria, eubacteria, and propionibacteria were reciprocal. The observed effect of the prebiotic complex effect on gut microbiota in rats with experimental heart failure suggests that this complex can be used for the correction of intestinal dysbiosis and endotoxemia in this clinical condition.


Dysbiosis/diet therapy , Endotoxemia/diet therapy , Heart Failure/diet therapy , Prebiotics/administration & dosage , Animals , Animals, Outbred Strains , Bacteria/growth & development , Bifidobacterium/growth & development , Disease Models, Animal , Dysbiosis/microbiology , Dysbiosis/physiopathology , Endotoxemia/microbiology , Endotoxemia/physiopathology , Endotoxins/biosynthesis , Female , Gastrointestinal Microbiome/drug effects , Heart Failure/microbiology , Heart Failure/physiopathology , Phenylephrine/administration & dosage , Physical Exertion , Propionibacterium/growth & development , Rats
16.
Sci Rep ; 10(1): 978, 2020 01 22.
Article En | MEDLINE | ID: mdl-31969646

Societal lifestyle changes, especially increased consumption of a high-fat diet lacking dietary fibers, lead to gut microbiota dysbiosis and enhance the incidence of adiposity and chronic inflammatory disease. We aimed to investigate the metabolic effects of inulin with different degrees of polymerization on high-fat diet-fed C57BL/6 J mice and to evaluate whether different health outcomes are related to regulation of the gut microbiota. Short-chain and long-chain inulins exert beneficial effects through alleviating endotoxemia and inflammation. Antiinflammation was associated with a proportional increase in short-chain fatty acid-producing bacteria and an increase in the concentration of short-chain fatty acids. Inulin might decrease endotoxemia by increasing the proportion of Bifidobacterium and Lactobacillus, and their inhibition of endotoxin secretion may also contribute to antiinflammation. Interestingly, the beneficial health effects of long-chain inulin were more pronounced than those of short-chain inulin. Long-chain inulin was more dependent than short-chain inulin on species capable of processing complex polysaccharides, such as Bacteroides. A good understanding of inulin-gut microbiota-host interactions helps to provide a dietary strategy that could target and prevent high-fat diet-induced endotoxemia and inflammation through a prebiotic effect.


Diet, High-Fat/adverse effects , Endotoxemia/prevention & control , Gastrointestinal Microbiome/drug effects , Inflammation/prevention & control , Inulin/therapeutic use , Protective Agents/therapeutic use , Animals , Bifidobacterium/drug effects , Body Weight/drug effects , Eating/drug effects , Endotoxemia/etiology , Endotoxemia/microbiology , Inflammation/etiology , Inflammation/microbiology , Inulin/pharmacology , Lactobacillus/drug effects , Male , Mice , Protective Agents/pharmacology
17.
Gut ; 69(9): 1608-1619, 2020 09.
Article En | MEDLINE | ID: mdl-31900292

OBJECTIVE: High-fat diet (HFD)-induced metabolic disorders can lead to impaired sperm production. We aim to investigate if HFD-induced gut microbiota dysbiosis can functionally influence spermatogenesis and sperm motility. DESIGN: Faecal microbes derived from the HFD-fed or normal diet (ND)-fed male mice were transplanted to the mice maintained on ND. The gut microbes, sperm count and motility were analysed. Human faecal/semen/blood samples were collected to assess microbiota, sperm quality and endotoxin. RESULTS: Transplantation of the HFD gut microbes into the ND-maintained (HFD-FMT) mice resulted in a significant decrease in spermatogenesis and sperm motility, whereas similar transplantation with the microbes from the ND-fed mice failed to do so. Analysis of the microbiota showed a profound increase in genus Bacteroides and Prevotella, both of which likely contributed to the metabolic endotoxaemia in the HFD-FMT mice. Interestingly, the gut microbes from clinical subjects revealed a strong negative correlation between the abundance of Bacteroides-Prevotella and sperm motility, and a positive correlation between blood endotoxin and Bacteroides abundance. Transplantation with HFD microbes also led to intestinal infiltration of T cells and macrophages as well as a significant increase of pro-inflammatory cytokines in the epididymis, suggesting that epididymal inflammation have likely contributed to the impairment of sperm motility. RNA-sequencing revealed significant reduction in the expression of those genes involved in gamete meiosis and testicular mitochondrial functions in the HFD-FMT mice. CONCLUSION: We revealed an intimate linkage between HFD-induced microbiota dysbiosis and defect in spermatogenesis with elevated endotoxin, dysregulation of testicular gene expression and localised epididymal inflammation as the potential causes. TRIAL REGISTRATION NUMBER: NCT03634644.


Bacteroides/isolation & purification , Diet, High-Fat/adverse effects , Dysbiosis , Prevotella/isolation & purification , Sperm Motility/immunology , Spermatogenesis/immunology , Animals , Correlation of Data , Cytokines/analysis , Dysbiosis/etiology , Dysbiosis/microbiology , Endotoxemia/microbiology , Epididymis/immunology , Epididymis/pathology , Feces/microbiology , Gastrointestinal Microbiome/immunology , Humans , Macrophages/immunology , Male , Mice , T-Lymphocytes/immunology
18.
Clin Exp Pharmacol Physiol ; 47(6): 927-939, 2020 06.
Article En | MEDLINE | ID: mdl-31894861

Translocation of microbiome-derived lipopolysaccharide (LPS) to the bloodstream (metabolic endotoxaemia) is associated with a significantly increased risk of cardiovascular diseases (CVD); however, the direction of this association is not fully understood. It has been revealed by some studies that alterations in the intestinal microbiota (dysbiosis) lead to increased intestinal permeability and translocation of LPS to the blood circulation. LPS may trigger toll-like receptor 4- (TLR-4) mediated inflammatory responses; this could lead to a chronic low-grade pro-inflammatory condition named metabolic endotoxaemia (ME), which is typically observed in CVD patients. ME is promoted by increased intestinal permeability. Moreover, dysbiosis leads to production of trimethylamine-N-oxide (TMAO), a gut bacterial metabolite suggested as a new risk factor in CVD development. Probiotics, extensively reviewed for decades, are live microorganisms which, when taken in adequate amounts, have beneficial effects on the host metabolism. Prebiotics are a type of dietary fibre that act as nourishment for the good bacteria in the gut and decrease the population of pathogen bacteria that produce greater amounts of endotoxins. Although an association has been postulated between ME and CVD, the results of studies investigating the role of antibiotic therapy in preventing the disease have been inconsistent. In this review, we discuss how prebiotics and probiotics modulate gut microbiota and consequently might help with prevention and/or treatment of CVD associated with ME.


Bacteria/metabolism , Cardiovascular Diseases/therapy , Endotoxemia/therapy , Gastrointestinal Microbiome , Intestines/microbiology , Prebiotics , Probiotics/therapeutic use , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/microbiology , Dysbiosis , Endotoxemia/metabolism , Endotoxemia/microbiology , Heart Disease Risk Factors , Host-Pathogen Interactions , Humans , Methylamines/metabolism , Prebiotics/adverse effects , Probiotics/adverse effects , Prognosis , Risk Assessment
19.
PLoS One ; 14(12): e0224838, 2019.
Article En | MEDLINE | ID: mdl-31790417

The gut plays a vital role in critical illness, and alterations in the gut structure and function have been reported in endotoxemia and sepsis models. Previously, we have demonstrated a novel link between the diet-induced alteration of the gut microbiome with cellulose and improved outcomes in sepsis. As compared to mice receiving basal fiber (BF) diet, mice that were fed a non-fermentable high fiber (HF) diet demonstrated significant improvement in survival and decreased organ injury in both cecal-ligation and puncture (CLP) and endotoxin sepsis models. To understand if the benefit conferred by HF diet extends to the gut structure and function, we hypothesized that HF diet would be associated with a reduction in sepsis-induced gut epithelial loss and permeability in mice. We demonstrate that the use of dietary cellulose decreased LPS-mediated intestinal hyperpermeability and protected the gut from apoptosis. Furthermore, we noted a significant increase in epithelial cell proliferation, as evidenced by an increase in the percentage of bromodeoxyuridine-positive cells in HF fed mice as compared to BF fed mice. Thus, the use of HF diet is a simple and effective tool that confers benefit in a murine model of sepsis, and understanding the intricate relationship between the epithelial barrier, gut microbiota, and diet will open-up additional therapeutic avenues for the treatment of gut dysfunction in critical illness.


Apoptosis/drug effects , Cellulose/pharmacology , Dietary Supplements , Endotoxemia/metabolism , Endotoxemia/pathology , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Animals , Cell Proliferation/drug effects , Disease Models, Animal , Endotoxemia/microbiology , Gastrointestinal Microbiome/drug effects , Gene Expression Regulation/drug effects , Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Male , Mice , Mice, Inbred C57BL , Permeability/drug effects , Tight Junction Proteins/metabolism
20.
Cell Rep ; 29(12): 3933-3945.e3, 2019 12 17.
Article En | MEDLINE | ID: mdl-31851924

Natural killer (NK) cells are unique players in innate immunity and, as such, an attractive target for immunotherapy. NK cells display immune memory properties in certain models, but the long-term status of NK cells following systemic inflammation is unknown. Here we show that following LPS-induced endotoxemia in mice, NK cells acquire cell-intrinsic memory-like properties, showing increased production of IFNγ upon specific secondary stimulation. The NK cell memory response is detectable for at least 9 weeks and contributes to protection from E. coli infection upon adoptive transfer. Importantly, we reveal a mechanism essential for NK cell memory, whereby an H3K4me1-marked latent enhancer is uncovered at the ifng locus. Chemical inhibition of histone methyltransferase activity erases the enhancer and abolishes NK cell memory. Thus, NK cell memory develops after endotoxemia in a histone methylation-dependent manner, ensuring a heightened response to secondary stimulation.


Endotoxemia/immunology , Escherichia coli Infections/immunology , Histones/metabolism , Immunity, Innate/immunology , Immunologic Memory/immunology , Inflammation/immunology , Killer Cells, Natural/immunology , Animals , Endotoxemia/metabolism , Endotoxemia/microbiology , Endotoxemia/pathology , Enhancer Elements, Genetic , Escherichia coli/immunology , Escherichia coli Infections/microbiology , Histones/genetics , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Interferon-gamma/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/microbiology , Killer Cells, Natural/pathology , Male , Mice
...