Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 148
Filter
1.
Gut Microbes ; 16(1): 2356278, 2024.
Article in English | MEDLINE | ID: mdl-38825779

ABSTRACT

The gut microbiota has been shown to be associated with a range of illnesses and disorders, including hypertension, which is recognized as the primary factor contributing to the development of serious cardiovascular diseases. In this review, we conducted a comprehensive analysis of the progression of the research domain pertaining to gut microbiota and hypertension. Our primary emphasis was on the interplay between gut microbiota and blood pressure that are mediated by host and gut microbiota-derived metabolites. Additionally, we elaborate the reciprocal communication between gut microbiota and antihypertensive drugs, and its influence on the blood pressure of the host. The field of computer science has seen rapid progress with its great potential in the application in biomedical sciences, we prompt an exploration of the use of microbiome databases and artificial intelligence in the realm of high blood pressure prediction and prevention. We propose the use of gut microbiota as potential biomarkers in the context of hypertension prevention and therapy.


Subject(s)
Antihypertensive Agents , Blood Pressure , Gastrointestinal Microbiome , Hypertension , Gastrointestinal Microbiome/physiology , Humans , Hypertension/microbiology , Antihypertensive Agents/therapeutic use , Animals , Bacteria/classification , Bacteria/metabolism , Bacteria/genetics , Bacteria/isolation & purification
2.
Artif Cells Nanomed Biotechnol ; 52(1): 278-290, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38733373

ABSTRACT

Type 2 diabetes mellitus (T2DM), nonalcoholic fatty liver disease (NAFLD), obesity (OB) and hypertension (HT) are categorized as metabolic disorders (MDs), which develop independently without distinct borders. Herein, we examined the gut microbiota (GM) and Saururus chinensis (SC) to confirm their therapeutic effects via integrated pharmacology. The overlapping targets from the four diseases were determined to be key protein coding genes. The protein-protein interaction (PPI) networks, and the SC, GM, signalling pathway, target and metabolite (SGSTM) networks were analysed via RPackage. Additionally, molecular docking tests (MDTs) and density functional theory (DFT) analysis were conducted to determine the affinity and stability of the conformer(s). TNF was the main target in the PPI analysis, and equol derived from Lactobacillus paracasei JS1 was the most effective agent for the formation of the TNF complex. The SC agonism (PPAR signalling pathway), and antagonism (neurotrophin signalling pathway) by SC were identified as agonistic bioactives (aromadendrane, stigmasta-5,22-dien-3-ol, 3,6,6-trimethyl-3,4,5,7,8,9-hexahydro-1H-2-benzoxepine, 4α-5α-epoxycholestane and kinic acid), and antagonistic bioactives (STK734327 and piclamilast), respectively, via MDT. Finally, STK734327-MAPK1 was the most favourable conformer according to DFT. Overall, the seven bioactives from SC and equol that can be produced by Lactobacillus paracasei JS1 can exert synergistic effects on these four diseases.


Subject(s)
Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Hypertension , Non-alcoholic Fatty Liver Disease , Obesity , Saururaceae , Gastrointestinal Microbiome/drug effects , Non-alcoholic Fatty Liver Disease/microbiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Obesity/microbiology , Obesity/metabolism , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/drug therapy , Hypertension/microbiology , Hypertension/metabolism , Hypertension/drug therapy , Animals , Saururaceae/chemistry , Saururaceae/metabolism , Molecular Docking Simulation , Humans , Protein Interaction Maps
3.
J Hypertens ; 42(7): 1212-1225, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38690877

ABSTRACT

BACKGROUND: Resistant hypertension is a severe phenotype in hypertension that may be driven by interactions between genetic and environmental factors. Specific changes in gut microbiota and metabolites have been shown to influence cardiovascular disease progression. However, microbial and metabolomic changes associated with resistant hypertension remain elusive. METHODS: In this study, the gut microbiome of 30 participants with resistant hypertension, 30 with controlled hypertension, and 30 nonhypertension was characterized using 16S rRNA amplicon sequencing. In addition, the serum metabolome of the same population was assessed by untargeted metabolomics. RESULTS: The alpha diversity of microbiome in the resistant hypertension decreased, and changes were also observed in the composition of the gut microbiota. The resistant hypertension group was characterized by elevated levels of Actinobacteitia and Proteobacteria. Twenty-three genera were found to have significantly different abundances between resistant hypertension and controlled hypertension, as well as 55 genera with significantly different abundances between resistant hypertension and nonhypertension. Compared with the controlled hypertension group, the genera Rothia and Sharpea in resistant hypertension were more abundant. Compared with the nonhypertension group, the genera Escherichia-Shigella , Lactobacillus , Enterococcus were more abundant. Untargeted metabolomics provided distinctly different serum metabolic profiles for the three groups and identified a range of differential metabolites. These metabolites were mainly associated with the pathway of glycerophospholipid metabolism. Furthermore, correlation analysis provided evidence of new interactions between gut microbiota and metabolites in the resistant hypertension. CONCLUSION: In conclusion, our study provides a comprehensive understanding of the resistant hypertension gut microbiota and metabolites, suggesting that treatment resistance in resistant hypertension patients may be related to the gut microbiota and serum metabolites.


Subject(s)
Gastrointestinal Microbiome , Hypertension , Metabolome , Humans , Hypertension/microbiology , Male , Middle Aged , Female , Aged , Metabolomics , RNA, Ribosomal, 16S/genetics
4.
Front Cell Infect Microbiol ; 14: 1358684, 2024.
Article in English | MEDLINE | ID: mdl-38660493

ABSTRACT

The enrichment of oral taxa in the gut has recently been reported as a notable alteration in the microbial balance in patients with intestinal disorders. However, translocation in populations without such diseases remains controversial. In this study, we examined 49 pairs of tongue and rectal samples collected from orthopedic patients without a history of intestinal disorders to verify the presence of oral taxa in the rectal microbiota. The bacterial composition of each sample was determined using 16S rRNA gene sequencing and amplicon sequence variant (ASV) analysis. Although the bacterial compositions of the tongue and rectal microbiota were distinctly different, tongue ASVs were detected in 67.3% of the participants and accounted for 0.0%-9.37% of the rectal microbiota. Particularly, Streptococcus salivarius, Fusobacterium nucleatum, and Streptococcus parasanguinis were abundant in the rectal microbiota. According to the network analysis, tongue taxa, such as S. salivarius and S. parasanguinis, formed a cohabiting group with Klebsiella pneumoniae and Alistipes finegoldii in the rectal microbiota. The total abundance of tongue ASVs in the rectal microbiota was significantly higher in participants with older age, hypertension, and proton pump inhibitor (PPI) use. Our study presents an extensive translocation of oral taxa to the rectum of a population without intestinal disorders and suggests that aging, hypertension, and PPI use are associated with an increased abundance of oral taxa and potential pathogenic bacteria in the rectal microbiota.


Subject(s)
Bacteria , Gastrointestinal Microbiome , RNA, Ribosomal, 16S , Rectum , Tongue , Humans , Male , Female , RNA, Ribosomal, 16S/genetics , Middle Aged , Gastrointestinal Microbiome/genetics , Adult , Tongue/microbiology , Aged , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Rectum/microbiology , Mouth/microbiology , DNA, Bacterial/genetics , Young Adult , Proton Pump Inhibitors , Sequence Analysis, DNA , Hypertension/microbiology , Microbiota
5.
Am J Physiol Renal Physiol ; 326(6): F1032-F1038, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38634136

ABSTRACT

The gut microbiome regulates many important host physiological processes associated with cardiovascular health and disease; however, the impact of the gut microbiome on aldosterone is unclear. Investigating whether gut microbiota regulate aldosterone can offer novel insights into how the microbiome affects blood pressure. In this study, we aimed to determine whether gut microbiota regulate host aldosterone. We used enzyme-linked immunosorbent assays (ELISAs) to assess plasma aldosterone and plasma renin activity (PRA) in female and male mice in which gut microbiota are intact, suppressed, or absent. In addition, we examined urinary aldosterone. Our findings demonstrated that when the gut microbiota is suppressed following antibiotic treatment, there is an increase in plasma and urinary aldosterone in both female and male mice. In contrast, an increase in PRA is seen only in males. We also found that when gut microbiota are absent (germ-free mice), plasma aldosterone is significantly increased compared with conventional animals (in both females and males), but PRA is not. Understanding how gut microbiota influence aldosterone levels could provide valuable insights into the development and treatment of hypertension and/or primary aldosteronism. This knowledge may open new avenues for therapeutic interventions, such as probiotics or dietary modifications to help regulate blood pressure via microbiota-based changes to aldosterone.NEW & NOTEWORTHY We explore the role of the gut microbiome in regulating aldosterone, a hormone closely linked to blood pressure and cardiovascular disease. Despite the recognized importance of the gut microbiome in host physiology, the relationship with circulating aldosterone remains largely unexplored. We demonstrate that suppression of gut microbiota leads to increased levels of plasma and urinary aldosterone. These findings underscore the potential of the gut microbiota to influence aldosterone regulation, suggesting new possibilities for treating hypertension.


Subject(s)
Aldosterone , Gastrointestinal Microbiome , Mice, Inbred C57BL , Renin , Animals , Aldosterone/blood , Aldosterone/metabolism , Gastrointestinal Microbiome/drug effects , Female , Male , Renin/blood , Renin/metabolism , Blood Pressure/drug effects , Germ-Free Life , Mice , Anti-Bacterial Agents/pharmacology , Hypertension/microbiology , Hypertension/metabolism
6.
Animal Model Exp Med ; 5(6): 513-531, 2022 12.
Article in English | MEDLINE | ID: mdl-35880388

ABSTRACT

Hypertension is an important global public health issue because of its high morbidity as well as the increased risk of other diseases. Recent studies have indicated that the development of hypertension is related to the dysbiosis of the gut microbiota in both animals and humans. In this review, we outline the interaction between gut microbiota and hypertension, including gut microbial changes in hypertension, the effect of microbial dysbiosis on blood pressure (BP), indicators of gut microbial dysbiosis in hypertension, and the microbial genera that affect BP at the taxonomic level. For example, increases in Lactobacillus, Roseburia, Coprococcus, Akkermansia, and Bifidobacterium are associated with reduced BP, while increases in Streptococcus, Blautia, and Prevotella are associated with elevated BP. Furthermore, we describe the potential mechanisms involved in the regulation between gut microbiota and hypertension. Finally, we summarize the commonly used treatments of hypertension that are based on gut microbes, including fecal microbiota transfer, probiotics and prebiotics, antibiotics, and dietary supplements. This review aims to find novel potential genera for improving hypertension and give a direction for future studies on gut microbiota in hypertension.


Subject(s)
Gastrointestinal Microbiome , Hypertension , Probiotics , Animals , Humans , Blood Pressure , Dysbiosis/complications , Dysbiosis/therapy , Probiotics/therapeutic use , Hypertension/complications , Hypertension/microbiology
7.
Int J Mol Sci ; 23(7)2022 Mar 28.
Article in English | MEDLINE | ID: mdl-35409057

ABSTRACT

The maintenance of the physiological values of blood pressure is closely related to unchangeable factors (genetic predisposition or pathological alterations) but also to modifiable factors (dietary fat and salt, sedentary lifestyle, overweight, inappropriate combinations of drugs, alcohol abuse, smoking and use of psychogenic substances). Hypertension is usually characterized by the presence of a chronic increase in systemic blood pressure above the threshold value and is an important risk factor for cardiovascular disease, including myocardial infarction, stroke, micro- and macro-vascular diseases. Hypertension is closely related to functional changes in the endothelium, such as an altered production of vasoconstrictive and vasodilator substances, which lead to an increase in vascular resistance. These alterations make the endothelial tissue unresponsive to autocrine and paracrine stimuli, initially determining an adaptive response, which over time lead to an increase in risk or disease. The gut microbiota is composed of a highly diverse bacterial population of approximately 1014 bacteria. A balanced intestinal microbiota preserves the digestive and absorbent functions of the intestine, protecting from pathogens and toxic metabolites in the circulation and reducing the onset of various diseases. The gut microbiota has been shown to produce unique metabolites potentially important in the generation of hypertension and endothelial dysfunction. This review highlights the close connection between hypertension, endothelial dysfunction and gut microbiota.


Subject(s)
Gastrointestinal Microbiome , Hypertension , Animals , Bacteria , Blood Pressure , Dysbiosis/microbiology , Humans , Hypertension/microbiology , Intestines/microbiology , Models, Animal
8.
Circ Res ; 130(3): 401-417, 2022 02 04.
Article in English | MEDLINE | ID: mdl-35113664

ABSTRACT

Hypertension is a worldwide problem with major impacts on health including morbidity and mortality, as well as consumption of health care resources. Nearly 50% of American adults have high blood pressure, and this rate is rising. Even with multiple antihypertensive drugs and aggressive lifestyle modifications, blood pressure is inadequately controlled in about 1 of 5 hypertensive individuals. This review highlights a hypothesis for hypertension that suggests alternative mechanisms for blood pressure elevation and maintenance. A better understanding of these mechanisms could open avenues for more successful treatments. The hypothesis accounts for recent understandings of the involvement of gut physiology, gut microbiota, and neuroinflammation in hypertension. It includes bidirectional communication between gut microbiota and gut epithelium in the gut-brain axis that is involved in regulation of autonomic nervous system activity and blood pressure control. Dysfunction of this gut-brain axis, including dysbiosis of gut microbiota, gut epithelial dysfunction, and deranged input to the brain, contributes to hypertension via inflammatory mediators, metabolites, bacteria in the circulation, afferent information alterations, etc resulting in neuroinflammation and unbalanced autonomic nervous system activity that elevates blood pressure. This in turn negatively affects gut function and its microbiota exacerbating the problem. We focus this review on the gut-brain axis hypothesis for hypertension and possible contribution to racial disparities in hypertension. A novel idea, that immunoglobulin A-coated bacteria originating in the gut with access to the brain could be involved in hypertension, is raised. Finally, minocycline, with its anti-inflammatory and antimicrobial properties, is evaluated as a potential antihypertensive drug acting on this axis.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Hypertension/microbiology , Animals , Humans , Hypertension/physiopathology
9.
Kaohsiung J Med Sci ; 38(2): 165-173, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34672426

ABSTRACT

To explore the relationship between the severity of hypertension and the imbalanced intestinal flora, inflammatory factors, adiponectin (ADPN) and vascular endothelial function in primary hypertension patients. According to the grading criteria for hypertension, in total of 60 patients with primary hypertension in our hospital from April to July, 2020 were divided into Grade 1 group (n = 20), Grade 2 group (n = 20), and Grade 3 group (n = 20). The feces of the research subjects were collected to extract the deoxyribonucleic acid (DNA) and detect its composition of intestinal flora. Subsequently, the peripheral blood was collected to determine the changes in inflammatory factors interleukin-2 (IL-2), IL-4, tumor necrosis factor-α (TNF-α) and IL-1ß, serum immunoglobulin G (IgG) and IgM, ADPN and vascular endothelial function-related endothelin-1 (ET-1), nitric oxide (NO), vascular endothelial growth factor (VEGF), and intercellular adhesion molecule-1 (ICAM-1). There were no significant differences in the gender, age, and body mass index (BMI), the proportion of smokers, diet habit, probiotics and antihypertensive medication use, and number of diabetic cases among groups (p > 0.05). We found an inverse association between blood pressure measures and microbial diversity, in particular microbial richness (p < 0.05). Among the four major kinds of intestinal flora, the composition of firmicutes (p < 0.05) and bacteroidetes (p < 0.05) showed obvious differences among the three groups, and they had consistent trends with the changes in the abundance of firmicutes and bacteroidetes. Intestinal flora imbalance is closely related to the severity of hypertension, inflammatory factors, ADPN, and vascular endothelial function.


Subject(s)
Adiponectin/blood , Endothelium, Vascular/physiopathology , Gastrointestinal Microbiome , Hypertension/blood , Hypertension/microbiology , Hypertension/physiopathology , Female , Humans , Inflammation/microbiology , Inflammation/physiopathology , Male , Middle Aged , Retrospective Studies , Severity of Illness Index
10.
Dis Markers ; 2021: 1683981, 2021.
Article in English | MEDLINE | ID: mdl-34659587

ABSTRACT

A connection exists between hypertension (HTN) and cognitive impairment (CI) or gut microbiota (GM) and neuropsychiatric disease. However, the link between GM and HTNCI has not been illustrated. This study endeavoured to profile the landscape of GM in HTNCI patients and evaluate the value of GM as HTNCI biomarkers. We recruited 128 patients with hypertension and assigned them to two groups of different MoCA scores. Clinical and biological data were recorded. GM composition was illustrated with 16S ribosomal RNA sequencing, and the dominant species were identified by linear discriminant analysis Effect Size (LEfSe). It showed higher abundance of TM7 and lower abundances of Veillonella and Peptoniphilus in the HTNCI group than in the HTN without cognitive impairment (HTNnCI) group. We next clarified the link between GM and MoCA scores or HTNCI factors. KEGG analysis revealed the involvement of decreased bile secretion. An evident correlation showed up between HTNCI and Veillonella abundance (P = 0.0340). We concluded that some representative GM species, especially Veillonella, could predict cognitive impairment in hypertension patients, making them potential benchmarks of HTNCI.


Subject(s)
Cognitive Dysfunction/microbiology , Gastrointestinal Microbiome/physiology , Hypertension/psychology , Aged , Cognitive Dysfunction/psychology , Female , Gastrointestinal Microbiome/genetics , Humans , Hypertension/microbiology , Male , Neuropsychological Tests , Risk Factors , Veillonella
11.
Int J Mol Sci ; 22(18)2021 Sep 11.
Article in English | MEDLINE | ID: mdl-34575987

ABSTRACT

Food-derived oligosaccharides show promising therapeutic potential in lowering blood pressure (BP), but the mechanism is poorly understood. Recently, the potential role of gut microbiota (GM) in hypertension has been investigated, but the specific GM signature that may participate in hypertension remains unclear. To test the potassium alginate oligosaccharides (PAO) mechanism in lowering BP and specific microbial signature changes in altering GM, we administered various dosages of PAO in 40 spontaneously hypertensive rats for a duration of six weeks. We analyzed BP, sequenced the 16S ribosomal DNA gene in the cecum content, and gathered RNA-seq data in cardiac tissues. We showed that the oral administration of PAO could significantly decrease systolic BP and mean arterial pressure. Transcriptome analyses demonstrated that the protective effects of developing heart failure were accompanied by down-regulating of the Natriuretic Peptide A gene expression and by decreasing the concentrations of angiotensin II and atrial natriuretic peptide in plasma. In comparison to the Vehicle control, PAO could increase the microbial diversity by altering the composition of GM. PAO could also decrease the ratio of Firmicutes to Bacteroidetes by decreasing the abundance of Prevotella and Phascolarctobacterium bacteria. The favorable effect of PAO may be added to the positive influence of the abundance of major metabolites produced by Gram-negative bacteria in GM. We suggest that PAO caused changes in GM, and thus, they played an important role in preventing the development of cardiovascular disease.


Subject(s)
Alginates/pharmacology , Gastrointestinal Microbiome/drug effects , Heart Failure , Hypertension , Oligosaccharides/pharmacology , Animals , Heart Failure/blood , Heart Failure/microbiology , Heart Failure/physiopathology , Heart Failure/prevention & control , Hypertension/blood , Hypertension/microbiology , Hypertension/physiopathology , Hypertension/prevention & control , Male , Rats , Rats, Inbred SHR
12.
Toxicol Appl Pharmacol ; 429: 115701, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34453990

ABSTRACT

Gut dysbiosis and dysregulation of gut-brain communication have been identified in hypertensive patients and animal models. Previous studies have shown that probiotic or prebiotic treatments exert positive effects on the pathophysiology of hypertension. This study aimed to examine the hypothesis that the microbiota-gut-brain axis is involved in the antihypertensive effects of curcumin, a potential prebiotic obtained from Curcuma longa. Male 8- to 10-week-old spontaneously hypertensive rats (SHRs) and Wistar Kyoto (WKY) rats were divided into four groups: WKY rats and SHRs treated with vehicle and SHRs treated with curcumin in dosage of 100 or 300 mg/kg/day for 12 weeks. Our results show that the elevated blood pressure of SHRs was markedly decreased in both curcumin-treated groups. Curcumin treatment also altered the gut microbial composition and improved intestinal pathology and integrity. These factors were associated with reduced neuroinflammation and oxidative stress in the hypothalamus paraventricular nucleus (PVN). Moreover, curcumin treatment increased butyrate levels in the plasma, which may be the result of increased butyrate-producing gut microorganisms. In addition, curcumin treatment also activated G protein-coupled receptor 43 (GPR 43) in the PVN. These results indicate that curcumin reshapes the composition of the gut microbiota and ameliorates the dysregulation of the gut-brain communication to induce antihypertensive effects.


Subject(s)
Antihypertensive Agents/pharmacology , Bacteria/drug effects , Blood Pressure/drug effects , Brain-Gut Axis/drug effects , Curcumin/pharmacology , Gastrointestinal Microbiome/drug effects , Hypertension/drug therapy , Paraventricular Hypothalamic Nucleus/drug effects , Animals , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Bacteria/growth & development , Bacteria/metabolism , Butyrates/blood , Cardiomegaly/metabolism , Cardiomegaly/microbiology , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Disease Models, Animal , Dysbiosis , Hypertension/metabolism , Hypertension/microbiology , Hypertension/physiopathology , Inflammation Mediators/metabolism , Male , Oxidative Stress/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Rats, Inbred SHR , Rats, Inbred WKY , Receptors, G-Protein-Coupled/metabolism
13.
Cells ; 10(6)2021 06 17.
Article in English | MEDLINE | ID: mdl-34204247

ABSTRACT

Hypertension is associated with gut bacterial dysbiosis and gut pathology in animal models and people. Butyrate-producing gut bacteria are decreased in hypertension. RNA-seq analysis of gut colonic organoids prepared from spontaneously hypertensive rats (SHR) and normotensive Wistar Kyoto (WKY) rats was used to test the hypothesis that impaired interactions between the gut microbiome and gut epithelium are involved and that these would be remediated with butyrate supplementation. Gene expressions in immune responses including antigen presentation and antiviral pathways were decreased in the gut epithelium of the SHR in organoids and confirmed in vivo; these deficits were corrected by butyrate supplementation. Deficits in gene expression driving epithelial proliferation and differentiation were also observed in SHR. These findings highlight the importance of aligned interactions of the gut microbiome and gut immune responses to blood pressure homeostasis.


Subject(s)
Colon/microbiology , Dysbiosis , Gastrointestinal Microbiome/physiology , Hypertension/microbiology , Animals , Butyrates/pharmacology , Colon/drug effects , Gastrointestinal Microbiome/drug effects , Male , Organoids , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Transcriptome
14.
Biomed Pharmacother ; 140: 111736, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34034069

ABSTRACT

Children with Type 1 diabetes mellitus (T1DM) have an elevated risk of abnormal blood pressure (BP) measurements and patterns. Both hypertension and T1DM are well-known risk factors for cardiovascular disease and kidney failure. The human microbiome has been linked to both diabetes and hypertension, but the relationship between the gut microbiome and BP in children with T1DM is not well-understood. In this cross-sectional study, we examined the relationship between resting office BP and gut microbiota composition, diversity, and richness in children with T1DM and healthy controls. We recruited 29 pediatric subjects and divided them into three groups: healthy controls (HC, n = 5), T1DM with normal BP (T1DM-Normo, n = 17), and T1DM with elevated BP (T1DM-HBP, n = 7). We measured the BP, dietary and clinical parameters for each subject. We collected fecal samples to perform the 16s rDNA sequencing and to measure the short-chain fatty acids (SCFAs) level. The microbiome downstream analysis included the relative abundance of microbiota, alpha and beta diversity, microbial markers using Linear Discriminant effect size analysis (LEfSe), potential gut microbial metabolic pathways using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) and metabolic pathways validation using Statistical Inference of Associations between Microbial Communities And host phenotype (SIAMCAT) machine learning toolbox. Our study results showed that T1DM-HBP group had distinct gut microbial composition (at multiple taxonomic levels) and reduced diversity (richness and abundance) compared with T1DM-Normo and HC groups. Children with T1DM-HBP showed a significant reduction of Bifidobacterium levels (especially B. adolescentis, B. bifidum, and B. longum) compared to the T1DM-Normo group. We also observed unique gut-microbial metabolic pathways, such as elevated lipopolysaccharide synthesis and glutathione metabolism in children with T1DM-HBP compared to T1DM-Normo children. We can conclude that the reduction in the abundance of genus Bifidobacterium could play a significant role in elevating the BP in pediatric T1DM subjects. More studies are needed to corroborate our findings and further explore the potential contributing mechanisms we describe.


Subject(s)
Bifidobacterium , Diabetes Mellitus, Type 1/microbiology , Hypertension/microbiology , Child , DNA, Bacterial/analysis , DNA, Ribosomal/analysis , Fatty Acids, Volatile/analysis , Feces/chemistry , Feces/microbiology , Female , Gastrointestinal Microbiome/genetics , Humans , Male
15.
Int J Mol Sci ; 22(5)2021 Mar 06.
Article in English | MEDLINE | ID: mdl-33800916

ABSTRACT

Gut microbiota-derived metabolites, in particular short chain fatty acids (SCFAs) and their receptors, are linked to hypertension. Fructose and antibiotics are commonly used worldwide, and they have a negative impact on the gut microbiota. Our previous study revealed that maternal high-fructose (HF) diet-induced hypertension in adult offspring is relevant to altered gut microbiome and its metabolites. We, therefore, intended to examine whether minocycline administration during pregnancy and lactation may further affect blood pressure (BP) programmed by maternal HF intake via mediating gut microbiota and SCFAs. Pregnant Sprague-Dawley rats received a normal diet or diet containing 60% fructose throughout pregnancy and lactation periods. Additionally, pregnant dams received minocycline (50 mg/kg/day) via oral gavage or a vehicle during pregnancy and lactation periods. Four groups of male offspring were studied (n = 8 per group): normal diet (ND), high-fructose diet (HF), normal diet + minocycline (NDM), and HF + minocycline (HFM). Male offspring were killed at 12 weeks of age. We observed that the HF diet and minocycline administration, both individually and together, causes the elevation of BP in adult male offspring, while there is no synergistic effect between them. Four groups displayed distinct enterotypes. Minocycline treatment leads to an increase in the F/B ratio, but decreased abundance of genera Lactobacillus, Ruminococcus, and Odoribacter. Additionally, minocycline treatment decreases plasma acetic acid and butyric acid levels. Hypertension programmed by maternal HF diet plus minocycline exposure is related to the increased expression of several SCFA receptors. Moreover, minocycline- and HF-induced hypertension, individually or together, is associated with the aberrant activation of the renin-angiotensin system (RAS). Conclusively, our results provide a new insight into the support of gut microbiota and its metabolite SCAFs in the developmental programming of hypertension and cast new light on the role of RAS in this process, which will help prevent hypertension programmed by maternal high-fructose and antibiotic exposure.


Subject(s)
Anti-Bacterial Agents/toxicity , Fructose/toxicity , Gastrointestinal Microbiome/physiology , Hypertension/microbiology , Minocycline/toxicity , Prenatal Exposure Delayed Effects , Animals , Anti-Bacterial Agents/administration & dosage , Fatty Acids, Volatile/metabolism , Female , Gastrointestinal Microbiome/drug effects , Gram-Negative Bacteria/drug effects , Gram-Negative Bacteria/isolation & purification , Gram-Negative Bacteria/metabolism , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/isolation & purification , Gram-Positive Bacteria/metabolism , Hypertension/etiology , Kidney/drug effects , Kidney/metabolism , Lactation , Male , Minocycline/administration & dosage , Nitric Oxide/metabolism , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Renin-Angiotensin System/physiology
16.
BMC Microbiol ; 21(1): 128, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33902467

ABSTRACT

BACKGROUND: Hypertension (HTN) is one of the major cardiovascular risk factors, which contributes to increasing target organ damages and cardiovascular morbidity and mortality worldwide. Isolated systolic HTN (ISH) and isolated diastolic HTN (IDH) are two important subtypes of HTN. Previous researches have demonstrated the alteration of fecal bacteria in HTN, but not down to these two sub-types. In order to identify whether the composition of bacterial taxa and functional modules shift in ISH and IDH, we performed a metagenomic sequencing analysis of fecal samples from 15 controls, 14 ISH, and 11 IDH. RESULTS: Compared with control and ISH, IDH patients showed decreased gene number, bacterial richness, and evenness, although the bacterial alterations did not reach statistical significance in the Shannon index. Also, at the genus level, the ß-diversity for intestinal flora in IDH was distinguishable from those with ISH. Furthermore, the taxonomic composition of ISH or IDH was different from that of healthy control at genus and species levels. Patients with IDH or ISH were confirmed to be enriched with Rothia mucilaginosa, along with reduced Clostridium sp. ASBs410. Lastly, the altered KEGG modules were significantly decreased in IDH compared with the control group, such as sodium transport system; while for ISH, functions relevant to biotin biosynthesis were decreased. CONCLUSIONS: Overall, our results showed the disordered fecal bacteria profiles in subjects with ISH and especially IDH, emphasizing the significance of early intervention for IDH.


Subject(s)
Biodiversity , Feces/microbiology , Hypertension/microbiology , Microbiota/physiology , Genes, Bacterial/genetics , Humans
17.
Acta Diabetol ; 58(8): 1009-1022, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33754165

ABSTRACT

AIMS: To identify fecal microbiota profiles associated with metabolic abnormalities belonging to the metabolic syndrome (MS), high count of white blood cells (WBCs) and insulin resistance (IR). METHODS: Sixty-eight young patients with obesity were stratified for percentile distribution of MS abnormalities. A MS risk score was defined as low, medium, and high MS risk. High WBCs were defined as a count ≥ 7.0 103/µL; severe obesity as body mass index Z-score ≥ 2 standard deviations; IR as homeostatic assessment model algorithm of IR (HOMA) ≥ 3.7. Stool samples were analyzed by 16S rRNA-based metagenomics. RESULTS: We found reduced bacterial richness of fecal microbiota in patients with IR and high diastolic blood pressure (BP). Distinct microbial markers were associated to high BP (Clostridium and Clostridiaceae), low high-density lipoprotein cholesterol (Lachnospiraceae, Gemellaceae, Turicibacter), and high MS risk (Coriobacteriaceae), WBCs (Bacteroides caccae, Gemellaceae), severe obesity (Lachnospiraceae), and impaired glucose tolerance (Bacteroides ovatus and Enterobacteriaceae). Conversely, taxa such as Faecalibacterium prausnitzii, Parabacterodes, Bacteroides caccae, Oscillospira, Parabacterodes distasonis, Coprococcus, and Haemophilus parainfluenzae were associated to low MS risk score, triglycerides, fasting glucose and HOMA-IR, respectively. Supervised multilevel analysis grouped clearly "variable" patients based on the MS risk. CONCLUSIONS: This was a proof-of-concept study opening the way at the identification of fecal microbiota signatures, precisely associated with cardiometabolic risk factors in young patients with obesity. These evidences led us to infer, while some gut bacteria have a detrimental role in exacerbating metabolic risk factors some others are beneficial ameliorating cardiovascular host health.


Subject(s)
Feces/microbiology , Inflammation/microbiology , Insulin Resistance/physiology , Metabolic Syndrome/microbiology , Microbiota/physiology , Obesity , Adolescent , Bacteria/classification , Bacteria/genetics , Biomarkers/blood , Child , Female , Glucose Intolerance/microbiology , Humans , Hypertension/microbiology , Male , Metagenomics , Obesity/complications , Pilot Projects , RNA, Ribosomal, 16S/analysis , Risk Factors , Triglycerides/blood
18.
Nat Commun ; 12(1): 1970, 2021 03 30.
Article in English | MEDLINE | ID: mdl-33785752

ABSTRACT

Periods of fasting and refeeding may reduce cardiometabolic risk elevated by Western diet. Here we show in the substudy of NCT02099968, investigating the clinical parameters, the immunome and gut microbiome exploratory endpoints, that in hypertensive metabolic syndrome patients, a 5-day fast followed by a modified Dietary Approach to Stop Hypertension diet reduces systolic blood pressure, need for antihypertensive medications, body-mass index at three months post intervention compared to a modified Dietary Approach to Stop Hypertension diet alone. Fasting alters the gut microbiome, impacting bacterial taxa and gene modules associated with short-chain fatty acid production. Cross-system analyses reveal a positive correlation of circulating mucosa-associated invariant T cells, non-classical monocytes and CD4+ effector T cells with systolic blood pressure. Furthermore, regulatory T cells positively correlate with body-mass index and weight. Machine learning analysis of baseline immunome or microbiome data predicts sustained systolic blood pressure response within the fasting group, identifying CD8+ effector T cells, Th17 cells and regulatory T cells or Desulfovibrionaceae, Hydrogenoanaerobacterium, Akkermansia, and Ruminococcaceae as important contributors to the model. Here we report that the high-resolution multi-omics data highlight fasting as a promising non-pharmacological intervention for the treatment of high blood pressure in metabolic syndrome patients.


Subject(s)
Blood Pressure/physiology , Body Weight/physiology , Fasting/physiology , Gastrointestinal Microbiome/physiology , Metabolic Syndrome/physiopathology , Aged , Akkermansia/physiology , Body Mass Index , Desulfovibrionaceae/physiology , Diet , Feces/microbiology , Female , Humans , Hypertension/complications , Hypertension/microbiology , Hypertension/physiopathology , Male , Metabolic Syndrome/complications , Metabolic Syndrome/microbiology , Middle Aged , Ruminococcus/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/physiology
19.
Sci Rep ; 11(1): 6211, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737624

ABSTRACT

We aimed to determine whether puncture sites for blood sampling and topical disinfectants are associated with rates of contaminated blood cultures in the emergency department (ED) of a single institution. This single-center, prospective observational study of 249 consecutive patients aged ≥ 20 years proceeded in the ED of a university hospital in Japan during 6 months. Pairs of blood samples were collected for aerobic and anaerobic culture from all patients in the ED. Physicians selected puncture sites and topical disinfectants according to their personal preference. We found 50 (20.1%) patients with potentially contaminated blood cultures. Fifty-six (22.5%) patients were true bacteremia and 143 (57.4%) patients were true negatives. Multivariate analysis associated more frequent contamination when puncture sites were disinfected with povidone-iodine than with alcohol/chlorhexidine (adjusted risk difference, 12.9%; 95% confidence interval [CI] 8.8-16.9; P < 0.001). Sites of blood collection were also associated with contamination. Femoral and central venous with other sites were associated with contamination more frequently than venous sites (adjusted risk difference), 13.1% (95% CI 8.2-17.9; P < 0.001]) vs. 17.3% (95% CI 3.6-31.0; P = 0.013). Rates of contaminated blood cultures were significantly higher when blood was collected from femoral sites and when povidone-iodine was the topical antiseptic.


Subject(s)
Bacteremia/complications , Blood Culture/methods , Diabetes Complications/microbiology , Hypertension/complications , Neoplasms/complications , Aged , Aged, 80 and over , Bacteremia/microbiology , Bacteremia/pathology , Blood Specimen Collection/adverse effects , Chlorhexidine/adverse effects , Diabetes Complications/pathology , Diabetes Mellitus/microbiology , Diabetes Mellitus/pathology , Disinfectants/adverse effects , Emergency Service, Hospital , Ethanol/adverse effects , False Positive Reactions , Female , Femoral Vein , Hospitals, University , Humans , Hypertension/microbiology , Hypertension/pathology , Japan , Male , Middle Aged , Neoplasms/microbiology , Neoplasms/pathology , Povidone-Iodine/adverse effects , Prospective Studies
20.
Biomed Pharmacother ; 137: 111334, 2021 May.
Article in English | MEDLINE | ID: mdl-33556874

ABSTRACT

High blood pressure (BP) presents a significant public health challenge. Recent findings suggest that altered microbiota can exert a hypertensive effect on the host. One of the possible mechanisms involved is the chronic translocation of its components, mainly lipopolysaccharides (LPS) into systemic circulation leading to metabolic endotoxemia. In animal models, LPS has been commonly used to induce endothelial dysfunction and vascular inflammation. In human studies, plasma LPS concentration has been positively correlated with hypertension, however, the mechanistic link has not been fully elucidated. It is hypothesised here that the LPS-induced direct alterations to the vascular endothelium and resulting hypertension are possible targets for probiotic intervention. The methodology of this review involved a systematic search of the literature with critical appraisal of papers. Three tranches of search were performed: 1) existing review papers; 2) primary mechanistic animal, in vitro and human studies; and 3) primary intervention studies. A total of 70 peer-reviewed papers were included across the three tranches and critically appraised using SIGN50 for human studies and the ARRIVE guidelines for animal studies. The extracted information was coded into key themes and summarized in a narrative analysis. Results highlight the role of LPS in the activation of endothelial toll-like receptor 4 (TLR4) initiating a cascade of interrelated signalling pathways including: 1) Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase/ Reactive oxygen species (ROS)/ Endothelial nitric oxide synthase (eNOS) pathway leading to endothelial dysfunction; and 2) Mitogen-Activated Protein Kinase (MAPK) and Nuclear factor kappa B (NF-κB) pathways leading to vascular inflammation. Findings from animal intervention studies suggest an improvement in vasorelaxation, vascular inflammation and hypertension following probiotic supplementation, which was mediated by downregulation of LPS-induced pathways. Randomised controlled trials (RCTs) and systematic reviews provided some evidence for the anti-inflammatory effect of probiotics with statistically significant antihypertensive effect in clinical samples and may offer a viable intervention for the management of hypertension.


Subject(s)
Endothelium, Vascular/metabolism , Gastrointestinal Microbiome , Hypertension/etiology , Hypertension/therapy , Probiotics/therapeutic use , Signal Transduction , Toll-Like Receptor 4/metabolism , Animals , Endothelium, Vascular/microbiology , Humans , Hypertension/microbiology , Inflammation/metabolism , Inflammation/microbiology , Lipopolysaccharides/toxicity
SELECTION OF CITATIONS
SEARCH DETAIL
...