Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 71
Filter
1.
J Prev Alzheimers Dis ; 11(3): 759-768, 2024.
Article in English | MEDLINE | ID: mdl-38706292

ABSTRACT

BACKGROUND: With differences apparent in the gut microbiome in mild cognitive impairment (MCI) and dementia, and risk factors of dementia linked to alterations of the gut microbiome, the question remains if gut microbiome characteristics may mediate associations of education with MCI. OBJECTIVES: We sought to examine potential mediation of the association of education and MCI by gut microbiome diversity or composition. DESIGN: Cross-sectional study. SETTING: Luxembourg, the Greater Region (surrounding areas in Belgium, France, Germany). PARTICIPANTS: Control participants of the Luxembourg Parkinson's Study. MEASUREMENTS: Gut microbiome composition, ascertained with 16S rRNA gene amplicon sequencing. Differential abundance, assessed across education groups (0-10, 11-16, 16+ years of education). Alpha diversity (Chao1, Shannon and inverse Simpson indices). Mediation analysis with effect decomposition was conducted with education as exposure, MCI as outcome and gut microbiome metrics as mediators. RESULTS: After exclusion of participants below 50, or with missing data, n=258 participants (n=58 MCI) were included (M [SD] Age=64.6 [8.3] years). Higher education (16+ years) was associated with MCI (Odds ratio natural direct effect=0.35 [95% CI 0.15-0.81]. Streptococcus and Lachnospiraceae-UCG-001 genera were more abundant in higher education. CONCLUSIONS: Education is associated with gut microbiome composition and MCI risk without clear evidence for mediation. However, our results suggest signatures of the gut microbiome that have been identified previously in AD and MCI to be reflected in lower education and suggest education as important covariate in microbiome studies.


Subject(s)
Cognitive Dysfunction , Educational Status , Gastrointestinal Microbiome , Humans , Cognitive Dysfunction/microbiology , Male , Risk Factors , Female , Cross-Sectional Studies , Aged , Middle Aged , Luxembourg/epidemiology , RNA, Ribosomal, 16S/genetics
2.
Front Cell Infect Microbiol ; 14: 1334581, 2024.
Article in English | MEDLINE | ID: mdl-38644963

ABSTRACT

Ischemic stroke (IS) is a serious central nervous system disease. Post-IS complications, such as post-stroke cognitive impairment (PSCI), post-stroke depression (PSD), hemorrhagic transformation (HT), gastrointestinal dysfunction, cardiovascular events, and post-stroke infection (PSI), result in neurological deficits. The microbiota-gut-brain axis (MGBA) facilitates bidirectional signal transduction and communication between the intestines and the brain. Recent studies have reported alterations in gut microbiota diversity post-IS, suggesting the involvement of gut microbiota in post-IS complications through various mechanisms such as bacterial translocation, immune regulation, and production of gut bacterial metabolites, thereby affecting disease prognosis. In this review, to provide insights into the prevention and treatment of post-IS complications and improvement of the long-term prognosis of IS, we summarize the interaction between the gut microbiota and IS, along with the effects of the gut microbiota on post-IS complications.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Ischemic Stroke , Humans , Ischemic Stroke/complications , Ischemic Stroke/microbiology , Brain-Gut Axis/physiology , Animals , Dysbiosis , Brain/microbiology , Bacterial Translocation , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/etiology
3.
BMC Microbiol ; 24(1): 134, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38654189

ABSTRACT

BACKGROUND: The incidence of exertional heat stroke (EHS) escalates during periods of elevated temperatures, potentially leading to persistent cognitive impairment postrecovery. Currently, effective prophylactic or therapeutic measures against EHS are nonexistent. METHODS: The selection of days 14 and 23 postinduction for detailed examination was guided by TEM of neuronal cells and HE staining of intestinal villi and the hippocampal regions. Fecal specimens from the ileum and cecum at these designated times were analyzed for changes in gut microbiota and metabolic products. Bioinformatic analyses facilitated the identification of pivotal microbial species and metabolites. The influence of supplementing these identified microorganisms on behavioral outcomes and the expression of functional proteins within the hippocampus was subsequently assessed. RESULTS: TEM analyses of neurons, coupled with HE staining of intestinal villi and the hippocampal region, indicated substantial recovery in intestinal morphology and neuronal injury on Day 14, indicating this time point for subsequent microbial and metabolomic analyses. Notably, a reduction in the Lactobacillaceae family, particularly Lactobacillus murinus, was observed. Functional annotation of 16S rDNA sequences suggested diminished lipid metabolism and glycan biosynthesis and metabolism in EHS models. Mice receiving this intervention (EHS + probiotics group) exhibited markedly reduced cognitive impairment and increased expression of BDNF/TrKB pathway molecules in the hippocampus during behavioral assessment on Day 28. CONCLUSION: Probiotic supplementation, specifically with Lactobacillus spp., appears to mitigate EHS-induced cognitive impairment, potentially through the modulation of the BDNF/TrKB signaling pathway within the hippocampus, illustrating the therapeutic potential of targeting the gut-brain axis.


Subject(s)
Cognitive Dysfunction , Gastrointestinal Microbiome , Heat Stroke , Animals , Female , Male , Mice , Brain-Gut Axis , Cognitive Dysfunction/diet therapy , Cognitive Dysfunction/etiology , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/psychology , Gastrointestinal Microbiome/physiology , Heat Stroke/complications , Heat Stroke/metabolism , Heat Stroke/physiopathology , Hippocampus/cytology , Hippocampus/physiopathology , Lactobacillus/metabolism , Neurons/ultrastructure , Probiotics , Behavior, Animal , Fatty Acids, Volatile/metabolism
4.
Cells ; 12(13)2023 06 28.
Article in English | MEDLINE | ID: mdl-37443769

ABSTRACT

The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.


Subject(s)
Brain-Gut Axis , Brain , Cognitive Dysfunction , Maternal Exposure , Prenatal Exposure Delayed Effects , Stress, Psychological , Cognitive Dysfunction/immunology , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/prevention & control , Humans , Female , Animals , Prenatal Exposure Delayed Effects/etiology , Melatonin/administration & dosage , Brain/growth & development , Neurogenesis , Antioxidants/administration & dosage , Probiotics/administration & dosage
5.
J Agric Food Chem ; 71(11): 4646-4655, 2023 Mar 22.
Article in English | MEDLINE | ID: mdl-36888896

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease, pathological markers of which are amyloid plaques and neurofibrillary tangles. As a key node of gut-brain axis, gut microbiota is increasingly associated with changes in cognitive behaviors and brain function. Psychobiotics are known to benefit patients with neurodegenerative diseases by the production and deliberation of neuroactive substances. However, psychobiotics are strain-specific probiotics, and their neuroprotective effects on the brain and modulation effects on the gut microbiome are not generalizable. In this study, we investigated the effects of Bifidobacterium breve HNXY26M4 in APP/PS1 mice. By assessing the alterations associated with brain function, we found that B. breve HNXY26M4 attenuated cognitive deficits and suppressed neuroinflammation and synaptic dysfunction in APP/PS1 mice. Moreover, by determining the modulation effects of B. breve HNXY26M4 on gut homeostasis, we identified that B. breve HNXY26M4 supplementation restored the composition of gut microbiota and short-chain fatty acids, as well as enhanced the function of the intestinal barrier. These findings indicate that microbiome-derived acetate and butyrate modulated by B. breve HNXY26M4 administration may be transported to the brain through the blood-brain barrier, and thus confer neuroprotective effects against AD-associated brain deficits and inflammation via the gut-brain axis.


Subject(s)
Alzheimer Disease , Bifidobacterium breve , Cognitive Dysfunction , Neurodegenerative Diseases , Neuroprotective Agents , Animals , Mice , Alzheimer Disease/genetics , Alzheimer Disease/microbiology , Bifidobacterium breve/genetics , Brain-Gut Axis , Cognition , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/microbiology , Mice, Transgenic , Neuroinflammatory Diseases
6.
Arch Microbiol ; 205(4): 118, 2023 Mar 16.
Article in English | MEDLINE | ID: mdl-36928985

ABSTRACT

Alzheimer's disease (AD), the most prevalent neurodegenerative disease, has a significant relationship with alteration of the gut microbiota (GM), and the GM-gut-brain axis has been explored to find novel therapeutic approaches for AD. The present study aimed to evaluate the effect of human Lactobacillaceae (HLL) on cognitive function in APP/PS1 mice. The results showed that HLL treatment significantly improved the cognitive function of mice via MWM and NOR tests. Furthermore, the expression of Aß plaques, tau phosphorylation and neuroinflammation were markedly reduced in the hippocampus. Meanwhile, HLL treatment significantly increased the activity of GSH-PX and decreased the expression levels of IL-6 and MDA in the brain, and simultaneously increased the abundance of beneficial bacteria and restrained pathogenic bacteria in the intestine. Interestingly, significant correlations were observed between significant changes in abundance of GMs and AD-related markers. Collectively, these findings reveal that HLL is a promising therapeutic agent and potential probiotics, which might improve the cognitive function and AD pathologies.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Gastrointestinal Microbiome , Lactobacillaceae , Neurodegenerative Diseases , Animals , Humans , Mice , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/therapeutic use , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/therapy , Disease Models, Animal , Mice, Transgenic , Neurodegenerative Diseases/microbiology , Neuroinflammatory Diseases
7.
PLoS One ; 18(2): e0280211, 2023.
Article in English | MEDLINE | ID: mdl-36827280

ABSTRACT

Those with mild cognitive impairment (MCI), a precursor to dementia, have a gut microbiome distinct from healthy individuals, but this has only been shown in healthy individuals, not in those exhibiting several risk factors for dementia. Using amplicon 16S rRNA gene sequencing in a case-control study of 60 older (ages 55-76), obese, predominately female, African American adults, those with MCI (cases) had different gut microbiota profiles than controls. While microbial community diversity was similar between cases and controls, the abundances of specific microbial taxa weren't, such as Parabacteroides distasonis (lower in cases) and Dialister invisus (higher in cases). These differences disappeared after adjusting for markers of oxidative stress and systemic inflammation. Cognitive scores were positively correlated with levels of Akkermansia muciniphila, a bacterium associated with reduced inflammation. Our study shows that gut microbial composition may be associated with inflammation, oxidative stress, and MCI in those at high risk for dementia.


Subject(s)
Cognitive Dysfunction , Gastrointestinal Microbiome , Obesity , Aged , Female , Humans , Middle Aged , Black or African American , Case-Control Studies , Cognitive Dysfunction/microbiology , Dementia , Gastrointestinal Microbiome/genetics , Inflammation , Obesity/microbiology , RNA, Ribosomal, 16S/genetics , Male
8.
Nutrients ; 14(19)2022 Sep 23.
Article in English | MEDLINE | ID: mdl-36235612

ABSTRACT

Altered gut microbiota has been reported in individuals with mild cognitive impairment (MCI) and Alzheimer's disease (AD). Previous research has suggested that specific bacterial species might be associated with the decline of cognitive function. However, the evidence was insufficient, and the results were inconsistent. To determine whether there is an alteration of gut microbiota in patients with MCI and AD and to investigate its correlation with clinical characteristics, the fecal samples from 94 cognitively normal controls (NC), 125 participants with MCI, and 83 patients with AD were collected and analyzed by 16S ribosomal RNA sequencing. The overall microbial compositions and specific taxa were compared. The clinical relevance was analyzed. There was no significant overall difference in the alpha and beta diversity among the three groups. Patients with AD or MCI had increased bacterial taxa including Erysipelatoclostridiaceae, Erysipelotrichales, Patescibacteria, Saccharimonadales, and Saccharimonadia, compared with NC group (p < 0.05), which were positively correlated with APOE 4 carrier status and Clinical Dementia Rating (correlation coefficient: 0.11~0.31, p < 0.05), and negatively associated with memory (correlation coefficient: −0.19~−0.16, p < 0.01). Our results supported the hypothesis that intestinal microorganisms change in MCI and AD. The alteration in specific taxa correlated closely with clinical manifestations, indicating the potential role in AD pathogenesis.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Gastrointestinal Microbiome , Aging , Alzheimer Disease/pathology , Apolipoprotein E4 , China , Cognitive Dysfunction/microbiology , Gastrointestinal Microbiome/genetics , Humans , RNA, Ribosomal, 16S/genetics
9.
Microbiol Spectr ; 10(1): e0235821, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35138147

ABSTRACT

Cognitive impairment (CI) is among the most common non-motor symptoms of Parkinson's disease (PD), with a substantially negative impact on patient management and outcome. The development and progression of CI exhibits high interindividual variability, which requires better diagnostic and monitoring strategies. PD patients often display sweating disorders resulting from autonomic dysfunction, which has been associated with CI. Because the axillary microbiota is known to change with humidity level and sweat composition, we hypothesized that the axillary microbiota of PD patients shifts in association with CI progression, and thus can be used as a proxy for classification of CI stages in PD. We compared the axillary microbiota compositions of 103 PD patients (55 PD patients with dementia [PDD] and 48 PD patients with mild cognitive impairment [PD-MCI]) and 26 cognitively normal healthy controls (HC). We found that axillary microbiota profiles differentiate HC, PD-MCI, and PDD groups based on differential ranking analysis, and detected an increasing trend in the log ratio of Corynebacterium to Anaerococcus in progression from HC to PDD. In addition, phylogenetic factorization revealed that the depletion of the Anaerococcus, Peptoniphilus, and W5053 genera is associated with PD-MCI and PDD. Moreover, functional predictions suggested significant increases in myo-inositol degradation, ergothioneine biosynthesis, propionate biosynthesis, menaquinone biosynthesis, and the proportion of aerobic bacteria and biofilm formation capacity, in parallel to increasing CI. Our results suggest that alterations in axillary microbiota are associated with CI in PD. Thus, axillary microbiota has the potential to be exploited as a noninvasive tool in the development of novel strategies. IMPORTANCE Parkinson's disease (PD) is the second most common neurodegenerative disease. Cognitive impairment (CI) in PD has significant negative impacts on life quality of patients. The emergence and progression of cognitive impairment shows high variability among PD patients, and thus requires better diagnostic and monitoring strategies. Recent findings indicate a close link between autonomic dysfunction and cognitive impairment. Since thermoregulatory dysfunction and skin changes are among the main manifestations of autonomic dysfunction in PD, we hypothesized that alterations in the axillary microbiota may be useful for tracking cognitive impairment stages in PD. To our knowledge, this the first study characterizing the axillary microbiota of PD patients and exploring its association with cognitive impairment stages in PD. Future studies should include larger cohorts and multicenter studies to validate our results and investigate potential biological mechanisms.


Subject(s)
Axilla/microbiology , Bacteria/isolation & purification , Cognitive Dysfunction/microbiology , Microbiota , Parkinson Disease/complications , Aged , Bacteria/classification , Bacteria/genetics , Cognitive Dysfunction/etiology , Female , Humans , Male , Middle Aged , Parkinson Disease/microbiology , Parkinson Disease/psychology , Phylogeny
10.
J Biol Chem ; 298(1): 101482, 2022 01.
Article in English | MEDLINE | ID: mdl-34896150

ABSTRACT

Patients who recover from nosocomial pneumonia oftentimes exhibit long-lasting cognitive impairment comparable with what is observed in Alzheimer's disease patients. We previously hypothesized that the lung endothelium contributes to infection-related neurocognitive dysfunction, because bacteria-exposed endothelial cells release a form(s) of cytotoxic tau that is sufficient to impair long-term potentiation in the hippocampus. However, the full-length lung and endothelial tau isoform(s) have yet to be resolved and it remains unclear whether the infection-induced endothelial cytotoxic tau triggers neuronal tau aggregation. Here, we demonstrate that lung endothelial cells express a big tau isoform and three additional tau isoforms that are similar to neuronal tau, each containing four microtubule-binding repeat domains, and that tau is expressed in lung capillaries in vivo. To test whether infection elicits endothelial tau capable of causing transmissible tau aggregation, the cells were infected with Pseudomonas aeruginosa. The infection-induced tau released from endothelium into the medium-induced neuronal tau aggregation in reporter cells, including reporter cells that express either the four microtubule-binding repeat domains or the full-length tau. Infection-induced release of pathological tau variant(s) from endothelium, and the ability of the endothelial-derived tau to cause neuronal tau aggregation, was abolished in tau knockout cells. After bacterial lung infection, brain homogenates from WT mice, but not from tau knockout mice, initiated tau aggregation. Thus, we conclude that bacterial pneumonia initiates the release of lung endothelial-derived cytotoxic tau, which is capable of propagating a neuronal tauopathy.


Subject(s)
Lung Diseases , Pneumonia, Bacterial , Tauopathies , tau Proteins , Animals , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/pathology , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Endothelial Cells/pathology , Humans , Lung/blood supply , Lung Diseases/metabolism , Lung Diseases/microbiology , Lung Diseases/pathology , Mice , Pneumonia, Bacterial/metabolism , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/pathology , Protein Isoforms , Pseudomonas aeruginosa , Tauopathies/genetics , Tauopathies/metabolism , Tauopathies/pathology , tau Proteins/chemistry , tau Proteins/genetics , tau Proteins/metabolism
11.
Int J Mol Sci ; 24(1)2022 Dec 31.
Article in English | MEDLINE | ID: mdl-36614151

ABSTRACT

Metabolites produced by an altered gut microbiota might mediate the effects in the brain. Among metabolites, the fecal volatile organic compounds (VOCs) are considered to be potential biomarkers. In this study, we examined both the VOCs and bacterial taxa in the feces from healthy subjects and Alzheimer's disease (AD) patients at early and middle stages. Remarkably, 29 fecal VOCs and 13 bacterial genera were differentiated from the healthy subjects and the AD patients. In general, higher amounts of acids and esters were found in in the feces of the AD patients and terpenes, sulfur compounds and aldehydes in the healthy subjects. At the early stage of AD, the most relevant VOCs with a higher abundance were short-chain fatty acids and their producing bacteria, Faecalibacterium and Lachnoclostridium. Coinciding with the development of dementia in the AD patients, parallel rises of heptanoic acid and Peptococcus were observed. At a more advanced stage of AD, the microbiota and volatiles shifted towards a profile in the feces with increases in hexanoic acid, Ruminococcus and Blautia. The most remarkable VOCs that were associated with the healthy subjects were 4-ethyl-phenol and dodecanol, together with their possible producers Clostridium and Coprococcus. Our results revealed a VOCs and microbiota crosstalk in AD development and their profiles in the feces were specific depending on the stage of AD. Additionally, some of the most significant fecal VOCs identified in our study could be used as potential biomarkers for the initiation and progression of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Microbiota , Volatile Organic Compounds , Humans , Volatile Organic Compounds/metabolism , Alzheimer Disease/metabolism , Cognitive Dysfunction/microbiology , Feces/microbiology , Fatty Acids, Volatile/metabolism , Bacteria/metabolism , Biomarkers/metabolism
12.
J Immunol Res ; 2021: 5578958, 2021.
Article in English | MEDLINE | ID: mdl-34869782

ABSTRACT

BACKGROUND: As a transitional state between normal aging and Alzheimer's disease (AD), mild cognitive impairment (MCI) is characterized by a worse cognitive decline than that of natural aging. The association between AD and gut microbiota has been reported in a number of studies; however, microbial research regarding MCI remains limited. METHODS: This study examined 48 participants, of whom 22 were MCI cases and 26 were normal control cases. Fecal samples were collected for 16S ribosomal RNA (rRNA) quantitative arrays and bioinformatics analysis. RESULTS: A principal coordinates analysis (PCoA) and nonmetric multidimensional scaling (NMDS) both demonstrated that the microbial composition of participants with MCI deviated from that of healthy control participants. Multiple bacterial species were significantly increased (e.g., Staphylococcus intermedius) or decreased (e.g., Bacteroides salyersiae) in samples from the MCI group. CONCLUSION: The composition of gut microbiota differed between normal control and MCI cases. This is the first study to identify a signature series of species in the gut microbiota of individuals with MCI. The results provide a new direction for the future development of an early diagnosis and probiotic regimen.


Subject(s)
Aging/immunology , Cognitive Dysfunction/immunology , Dysbiosis/complications , Gastrointestinal Microbiome/immunology , Aged , Case-Control Studies , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/prevention & control , Dysbiosis/diet therapy , Dysbiosis/immunology , Dysbiosis/microbiology , Feces/microbiology , Female , Healthy Volunteers , Humans , Male , Middle Aged , Probiotics/administration & dosage
13.
Cell Rep Med ; 2(9): 100398, 2021 09 21.
Article in English | MEDLINE | ID: mdl-34622235

ABSTRACT

Evidence linking the gut-brain axis to Alzheimer's disease (AD) is accumulating, but the characteristics of causally important microbes are poorly understood. We perform a fecal microbiome analysis in healthy subjects and those with mild cognitive impairment (MCI) and AD. We find that Faecalibacterium prausnitzii (F. prausnitzii) correlates with cognitive scores and decreases in the MCI group compared with the healthy group. Two isolated strains from the healthy group, live Fp360 and pasteurized Fp14, improve cognitive impairment in an AD mouse model. Whole-genome comparison of isolated strains reveals specific orthologs that are found only in the effective strains and are more abundant in the healthy group compared with the MCI group. Metabolome and RNA sequencing analyses of mouse brains provides mechanistic insights into the relationship between the efficacy of pasteurized Fp14, oxidative stress, and mitochondrial function. We conclude that F. prausnitzii strains with these specific orthologs are candidates for gut microbiome-based intervention in Alzheimer's-type dementia.


Subject(s)
Alzheimer Disease/microbiology , Dementia/microbiology , Faecalibacterium prausnitzii/physiology , Gastrointestinal Microbiome , Aged , Amyloid beta-Peptides/metabolism , Brain/microbiology , Brain/pathology , Cognition , Cognitive Dysfunction/microbiology , Faecalibacterium prausnitzii/genetics , Faecalibacterium prausnitzii/isolation & purification , Female , Genome, Bacterial , Humans , Male , Metabolome/genetics , Metagenome , Pasteurization , Principal Component Analysis , RNA-Seq
14.
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
15.
Sci Rep ; 11(1): 20659, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34667205

ABSTRACT

Gut lactobacilli and bifidobacteria on the immune homeostasis. Therefore, to understand the mechanism in vivo, we selected human fecal Lactobacillus rhamnosus NK210 and Bifidobacterium longum NK219, which strongly suppressed the IFN-γ to IL-10 expression (IIE) ratio in lipopolysaccharide-stimulated macrophages. Thereafter, we examined their effects on the endotoxin, antibiotics, or antitumor drug-stimulated immune imbalance in mice. Intraperitoneal injection of lipopolysaccharide and oral gavage of ampicillin increased IFN-γ and TNF-α expression in the spleen, colon, and hippocampus, while IL-10 expression decreased. However, intraperitoneal injection of cyclophosphamide suppressed IFN-γ, TNF-α, and IL-10 expression. LPS exposure induced splenic natural killer cell cytotoxicity against YAC-1 cells (sNK-C) and peritoneal macrophage phagocytosis against Candida albicans (pMA-P) activities, while cyclophosphamide and ampicillin treatments suppressed sNK-C and pMA-P activities. However, LPS, ampicillin, cyclophosphamide all increased IIE and TNF-α to IL-10 expression (TIE) ratios. Oral administration of NK210 and/or NK219 significantly reduced LPS-induced sNK-C, pMA-P, and IFN-γ expression, while cyclophosphamide- or ampicillin-suppressed sNK-C and pMA-P activities, cyclophosphamide-suppressed IFN-γ, TNF-α, and IL-10 expression, and ampicillin-suppressed IL-10 expression increased. Nevertheless, they suppressed LPS-, ampicillin-, or cyclophosphamide-induced IIE and TIE ratios, cognitive impairment, and gut dysbiosis. In particular, NK219, but not NK210, increased the IIE expression ratio in vitro and in vivo, and enhanced sNK-C and pMA-P activities in normal control mice, while cognitive function and gut microbiota composition were not significantly affected. These findings suggest that NK210, Lactobacillus sp, and NK219, Bifidobacterium additively or synergistically alleviate gut dysbiosis, inflammation, and cognitive impairment with immune imbalance by controlling IIE and TIE ratios.


Subject(s)
Bifidobacterium longum/metabolism , Dysbiosis/therapy , Lacticaseibacillus rhamnosus/metabolism , Animals , Bifidobacterium/metabolism , Bifidobacterium longum/pathogenicity , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/therapy , Colitis/microbiology , Colitis/therapy , Feces/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Inflammation/metabolism , Interferon-gamma/antagonists & inhibitors , Interferon-gamma/metabolism , Interleukin-10/metabolism , Lactobacillus/metabolism , Lacticaseibacillus rhamnosus/pathogenicity , Male , Mice , Mice, Inbred C57BL , Probiotics/administration & dosage , Tumor Necrosis Factor-alpha/metabolism
16.
J Leukoc Biol ; 110(3): 511-524, 2021 09.
Article in English | MEDLINE | ID: mdl-34342041

ABSTRACT

Periodontitis is one of the most common oral diseases worldwide, and it is associated with various systemic diseases, including cognitive diseases. STAT3 regulates the inflammatory cascade and influences adaptive immunity by modulating Th17/Treg cell differentiation. In this study, we aimed to explore the effect of adaptive immunity inside and outside the brain on the association between periodontitis and cognitive impairment and understand the role of the STAT3 signaling pathway. We established Porphyromonas gingivalis LPS-induced periodontitis mice models by injecting P. gingivalis LPS into the gingival sulcus of mice. Behavioral tests showed that learning and memory abilities were impaired. The flow cytometry data showed an imbalance in the Th17/Treg ratio in the blood and brain samples of the mice. The expression of Th17-related cytokines (IL-1ß, IL-17A, IL-21, and IL-22) increased, whereas that of Treg-related cytokines (IL-2 and IL-10) decreased in both the blood and the brain. The level of LPS increased and the STAT3 signaling pathway was activated during this process. These effects were reversed by C188-9, a STAT3 inhibitor. In conclusion, P. gingivalis LPS-induced periodontitis may promote the occurrence and progression of cognitive impairment by modulating the Th17/Treg balance inside and outside the brain. The STAT3 signaling pathway may have immunoregulatory effects on the mouth-to-brain axis.


Subject(s)
Cognitive Dysfunction/immunology , Cognitive Dysfunction/microbiology , Periodontitis/immunology , Periodontitis/microbiology , Porphyromonas gingivalis/physiology , STAT3 Transcription Factor/metabolism , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Alveolar Process/pathology , Animals , Astrocytes/pathology , Bone Resorption/complications , Bone Resorption/immunology , Bone Resorption/microbiology , Bone Resorption/pathology , Cognitive Dysfunction/complications , Cognitive Dysfunction/diagnostic imaging , Cytokines/metabolism , Gingiva/pathology , Lipopolysaccharides , Memory , Mice , Microglia/pathology , Periodontitis/complications , Periodontitis/diagnostic imaging , Signal Transduction , Spatial Learning
17.
Nutrients ; 13(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34444674

ABSTRACT

Changes in the composition and proportions of the gut microbiota may be associated with numerous diseases, including cognitive impairment. Over the recent years, the growing interest in this relation is observed, but there are still many unknowns, especially in the elderly. To the best of our knowledge, this is the first work that synthesizes and critically evaluates existing evidence on the possible association between human gut microbiota and cognitive function in the elderly. For this purpose, comprehensive literature searches were conducted using the electronic databases PubMed, Google Scholar, and ScienceDirect. The gut microbiota of cognitively healthy and impaired elderly people may differ in the diversity and abundance of individual taxes, but specific taxes cannot be identified. However, some tendencies to changing the Firmicutes/Bacteroidetes ratio can be identified. Currently, clinical trials involving probiotics, prebiotics, and synbiotics supplementation have shown that there are premises for the claim that these factors can improve cognitive functions, however there is no single intervention beneficial to the elderly population. More reliable evidence from large-scale, long-period RCT is needed. Despite proposing several potential mechanisms of the gut microbiota's influence on the cognitive function impairment, prospective research on this topic is extremely difficult to conduct due to numerous confounding factors that may affect the gut microbiota. Heterogeneity of research outcomes impairs insight into these relations.


Subject(s)
Aging/physiology , Cognition/physiology , Gastrointestinal Microbiome/physiology , Probiotics/administration & dosage , Aged , Aged, 80 and over , Bacteroidetes/physiology , Cognitive Dysfunction/microbiology , Diet , Firmicutes/physiology , Humans , Prebiotics/administration & dosage
18.
Aging (Albany NY) ; 13(12): 16733-16748, 2021 06 28.
Article in English | MEDLINE | ID: mdl-34182544

ABSTRACT

Multiple exposures to anesthesia may increase the risk of cognitive impairment in young children. However, the mechanisms underlying this neurodevelopmental disorder remain elusive. In this study, we investigated alteration of the gut microbiota after multiple neonatal exposures to the anesthetic sevoflurane and the potential role of microbiota alteration on cognitive impairment using a young mice model. Multiple neonatal sevoflurane exposures resulted in obvious cognitive impairment symptoms and altered gut microbiota composition. Fecal transplantation experiments confirmed that alteration of the microbiota was responsible for the cognitive disorders in young mice. Microbiota profiling analysis identified microbial taxa that showed consistent differential abundance before and after fecal microbiota transplantation. Several of the differentially abundant taxa are associated with memory and/or health of the host, such as species of Streptococcus, Lachnospiraceae, and Pseudoflavonifractor. The results reveal that abnormal composition of the gut microbiota is a risk factor for cognitive impairment in young mice after multiple neonatal exposures to sevoflurane and provide insight into a potential therapeutic strategy for sevoflurane-related neurotoxicity.


Subject(s)
Cognitive Dysfunction/microbiology , Gastrointestinal Microbiome , Prenatal Exposure Delayed Effects/microbiology , Sevoflurane/adverse effects , Anesthesia , Animals , Anti-Bacterial Agents/pharmacology , Behavior, Animal , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Germ-Free Life , Mice, Inbred C57BL , Morris Water Maze Test , Pregnancy
19.
Aging (Albany NY) ; 13(11): 15320-15335, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34081627

ABSTRACT

Recent evidence suggests alterations in the gut microbiota-brain axis may drive cognitive impairment with aging. In the present study, we observed that prolonged administration of D-galactose to mice induced cognitive decline, gut microbial dysbiosis, peripheral inflammation, and oxidative stress. In this model of age-related cognitive decline, Cistanche deserticola polysaccharides (CDPS) improved cognitive function in D-galactose-treated mice by restoring gut microbial homeostasis, thereby reducing oxidative stress and peripheral inflammation. The beneficial effects of CDPS in these aging model mice were abolished through ablation of gut microbiota with antibiotics or immunosuppression with cyclophosphamide. Serum metabolomic profiling showed that levels of creatinine, valine, L-methionine, o-Toluidine, N-ethylaniline, uric acid and proline were all altered in the aging model mice, but were restored by CDPS. These findings demonstrated that CDPS improves cognitive function in a D-galactose-induced aging model in mice by restoring homeostasis of the gut microbiota-brain axis, which alleviated an amino acid imbalance, peripheral inflammation, and oxidative stress. CDPS thus shows therapeutic potential for patients with memory and learning disorders, especially those related to gut microbial dysbiosis.


Subject(s)
Aging/pathology , Brain/pathology , Cistanche/chemistry , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/microbiology , Gastrointestinal Microbiome/drug effects , Models, Biological , Polysaccharides/therapeutic use , Amino Acids/metabolism , Animals , Cognitive Dysfunction/complications , Cognitive Dysfunction/immunology , Cytokines/metabolism , Dysbiosis/complications , Dysbiosis/microbiology , Galactose , Homeostasis , Inflammation/pathology , Inflammation Mediators/metabolism , Male , Memory Disorders/complications , Mice , Nerve Degeneration/complications , Nerve Degeneration/pathology , Oxidative Stress/drug effects , Polysaccharides/pharmacology , Purines/metabolism
20.
J Alzheimers Dis ; 82(2): 513-526, 2021.
Article in English | MEDLINE | ID: mdl-34024839

ABSTRACT

BACKGROUND: Subjective cognitive decline (SCD) is the earliest symptomatic manifestation of preclinical Alzheimer's disease (AD). Gut microbiota may serve as a susceptibility factor for AD. Altered gut microbiota has been reported in patients with mild cognitive impairment (MCI) and AD dementia. However, whether gut microbial compositions changed in SCD remains largely unknown. OBJECTIVE: To characterize the gut microbiota in SCD. METHODS: In this study, a total of 105 participants including 38 normal controls (NC), 53 individuals with SCD, and 14 patients with cognitive impairment (CI) were recruited. Gut microbiota of all participants isolated from fecal samples were investigated using 16S ribosomal RNA (rRNA) Illumina Miseq sequencing technique. The gut microbial compositions were compared among the three groups, and the association between altered gut microbiota and cognitive performance was analyzed. To validate the alteration of gut microbiota in SCD, we conducted amyloid positron emission tomography (PET) in selected participants and further compared the gut microbiota among subgroups. RESULTS: The abundance of phylum Firmicutes, class Clostridia, order Clostridiales, family Ruminococcaceae, and genus Faecalibacterium showed a trend toward a progressive decline from NC to SCD and CI. Specifically, the abundance of the anti-inflammatory genus Faecalibacterium was significantly decreased in SCD compared with NC. In addition, altered bacterial taxa among the three groups were associated with cognitive performance. The findings were validated in SCD participants with positive amyloid evidence. CONCLUSION: The composition of gut microbiota is altered in individuals with SCD. This preliminary study will provide novel insights into the pathophysiological mechanism of AD.


Subject(s)
Alzheimer Disease , Cognitive Dysfunction , Diagnostic Self Evaluation , Feces/microbiology , Gastrointestinal Microbiome/genetics , Aged , Alzheimer Disease/diagnosis , Alzheimer Disease/epidemiology , Brain/diagnostic imaging , Brain/metabolism , China/epidemiology , Cognition/physiology , Cognitive Dysfunction/diagnosis , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/psychology , Early Diagnosis , Female , Humans , Male , Neuropsychological Tests , Plaque, Amyloid/diagnostic imaging , Positron-Emission Tomography/methods , RNA, Ribosomal, 16S/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL
...