Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 916
Filter
1.
BMC Microbiol ; 24(1): 289, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095715

ABSTRACT

BACKGROUND: Chronic stress induces cognitive deficits. There is a well-established connection between the enteric and central nervous systems through the microbiota-gut-brain (MGB) axis. However, the effects of the gut microbiota on cognitive deficits remain unclear. The present study aimed to elucidate the microbiota composition in cognitive deficits and explore its potential in predicting chronic stress-induced cognitive deficits. METHODS: Mice were randomly divided into control and chronic restraint stress (CRS) groups. The mice subjected to CRS were further divided into cognitive deficit (CRS-CD) and non-cognitive deficit (CRS-NCD) groups using hierarchical cluster analysis of novel object recognition test results. The composition and diversity of the gut microbiota were analyzed. RESULTS: After being subjected to chronic restraint distress, the CRS-CD mice travelled shorter movement distances (p = 0.034 vs. CRS-NCD; p < 0.001 vs. control) and had a lower recognition index than the CRS-NCD (p < 0.0001 vs. CRS-NCD; p < 0.0001 vs. control) and control mice. The results revealed that 5 gut bacteria at genus levels were significantly different in the fecal samples of mice in the three groups. Further analyses demonstrated that Muricomes were not only significantly enriched in the CRS-CD group but also correlated with a decreased cognitive index. The area under the receiver operating curve of Muricomes for CRS-induced cognitive deficits was 0.96. CONCLUSIONS: Our study indicates that the composition of the gut microbiota is involved in the development of cognitive deficits induced by chronic restraint stress. Further analysis revealed that Muricomes have the potential to predict the development of chronic stress-induced cognitive deficits in mice.


Subject(s)
Cognitive Dysfunction , Feces , Gastrointestinal Microbiome , Restraint, Physical , Stress, Psychological , Animals , Mice , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/physiopathology , Cognitive Dysfunction/etiology , Male , Stress, Psychological/microbiology , Stress, Psychological/complications , Stress, Psychological/psychology , Feces/microbiology , Bacteria/classification , Bacteria/isolation & purification , Bacteria/genetics , Mice, Inbred C57BL , Disease Models, Animal , Brain-Gut Axis/physiology
2.
J Neuroimmune Pharmacol ; 19(1): 37, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-39052165

ABSTRACT

Recombinant interleukin-22 (rIL-22) has been reported as a protective agent in murine models of diseases driven by epithelial injury. Parasites have a circadian rhythm and their sensitivity to a certain drug may vary during the day. Therefore, this work aimed to investigate the effect of rIL-22 administration at different times of the day on the inflammation, oxidative status, and neurotransmitter release in the gut-brain axis of the Schistosoma mansoni-infected mice. Sixty male BALB/c mice aged six weeks weighing 25-30 g were divided into a control group (injected intraperitoneally with PBS), mice infected with 80 ± 10 cercariae of S. mansoni (infected group) then injected intraperitoneally with PBS, and rIL-22 treated groups. rIL-22 was administrated intraperitoneally (400 ng/kg) either at the onset or offset of the light phase for 14 days. IL-22 administration reduced the levels of IL-1ß, tumor necrosis factor-alpha (TNF-α), nuclear factor kappa beta (NF-κß), and enhanced the production of IL-22 and IL-17. The treatment with IL-22 increased glutathione (GSH) and reduced malondialdehyde (MDA) and nitric oxide (NO) levels both in the ileum and brain. The B-cell lymphoma 2 (BCL2) protein level in the ileum was diminished after IL-22 administration. Brain-derived neurotrophic factor (BDNF) and neurotransmitter release (serotonin, 5HT, norepinephrine, NE, dopamine, DA, Glutamate, Glu, and -amino butyric acid, GABA) were improved by rIL-22. In conclusion, rIL-22 showed promising immunotherapy for inflammation, oxidative damage, and neuropathological signs associated with schistosomiasis. The efficacy of IL-22 increased significantly upon its administration at the time of light offset.


Subject(s)
Brain-Gut Axis , Interleukin-22 , Interleukins , Mice, Inbred BALB C , Neurotransmitter Agents , Recombinant Proteins , Schistosomiasis mansoni , Animals , Mice , Male , Neurotransmitter Agents/metabolism , Neurotransmitter Agents/pharmacology , Interleukins/metabolism , Schistosomiasis mansoni/drug therapy , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/metabolism , Recombinant Proteins/pharmacology , Recombinant Proteins/administration & dosage , Brain-Gut Axis/drug effects , Brain-Gut Axis/physiology , Immunotherapy/methods , Biogenic Monoamines/metabolism , Inflammation/metabolism , Inflammation/drug therapy
3.
Alcohol Alcohol ; 59(5)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39042929

ABSTRACT

The human intestine is colonized by a variety of microorganisms that influence the immune system, the metabolic response, and the nervous system, with consequences for brain function and behavior. Unbalance in this microbial ecosystem has been shown to be associated with psychiatric disorders, and altered gut microbiome composition related to bacteria, viruses, and fungi has been well established in patients with alcohol use disorder. This review describes the gut microbiome-brain communication pathways, including the ones related to the vagus nerve, the inflammatory cytokines, and the gut-derived metabolites. Finally, the potential benefits of microbiota-based therapies for the management of alcohol use disorder, such as probiotics, prebiotics, and fecal microbiota transplantation, are also discussed.


Subject(s)
Alcoholism , Brain-Gut Axis , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Alcoholism/microbiology , Alcoholism/therapy , Probiotics/therapeutic use , Prebiotics , Animals , Brain/metabolism
4.
Discov Med ; 36(186): 1354-1362, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39054706

ABSTRACT

Major depressive disorder (MDD) is a clinical condition that significantly impacts patients' physical and mental well-being, quality of life, and social functioning. The pathogenesis of MDD remains unclear, but accumulating evidence suggests a close relationship between gut microbiota and the occurrence and progression of MDD. Gut microbiota refers to the microbial community in the human intestine, which engages in bidirectional communication with the host via the "gut-brain axis" and plays a pivotal role in influencing the host's metabolism, immune system, endocrine system, and nervous system. Modulating gut microbiota entails restoring the balance and function of the intestinal flora through methods such as probiotic intake, fecal transplantation, and dietary intervention. Such modulation has been shown to effectively alleviate depressive symptoms in the host. This review synthesizes recent advancements in research on gut microbiota modulation for ameliorating depressive symptoms and can serve as a foundation for further exploration of the gut microbiota's role in MDD and its potential therapeutic benefits.


Subject(s)
Depressive Disorder, Major , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Probiotics , Humans , Gastrointestinal Microbiome/physiology , Depressive Disorder, Major/microbiology , Depressive Disorder, Major/therapy , Depressive Disorder, Major/immunology , Probiotics/therapeutic use , Brain-Gut Axis/physiology , Animals
5.
CNS Neurosci Ther ; 30(7): e14840, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38973202

ABSTRACT

BACKGROUND: Heat stress (HS) commonly occurs as a severe pathological response when the body's sensible temperature exceeds its thermoregulatory capacity, leading to the development of chronic brain inflammation, known as neuroinflammation. Emerging evidence suggests that HS leads to the disruption of the gut microbiota, whereas abnormalities in the gut microbiota have been demonstrated to affect neuroinflammation. However, the mechanisms underlying the effects of HS on neuroinflammation are poorly studied. Meanwhile, effective interventions have been unclear. ß-Hydroxybutyric acid (BHBA) has been found to have neuroprotective and anti-inflammatory properties in previous studies. This study aims to explore the modulatory effects of BHBA on neuroinflammation induced by HS and elucidate the underlying molecular mechanisms. METHODS: An in vivo and in vitro model of HS was constructed under the precondition of BHBA pretreatment. The modulatory effects of BHBA on HS-induced neuroinflammation were explored and the underlying molecular mechanisms were elucidated by flow cytometry, WB, qPCR, immunofluorescence staining, DCFH-DA fluorescent probe assay, and 16S rRNA gene sequencing of colonic contents. RESULTS: Heat stress was found to cause gut microbiota disruption in HS mouse models, and TM7 and [Previotella] spp. may be the best potential biomarkers for assessing the occurrence of HS. Fecal microbiota transplantation associated with BHBA effectively reversed the disruption of gut microbiota in HS mice. Moreover, BHBA may inhibit microglia hyperactivation, suppress neuroinflammation (TNF-α, IL-1ß, and IL-6), and reduce the expression of cortical endoplasmic reticulum stress (ERS) markers (GRP78 and CHOP) mainly through its modulatory effects on the gut microbiota (TM7, Lactobacillus spp., Ruminalococcus spp., and Prevotella spp.). In vitro experiments revealed that BHBA (1 mM) raised the expression of the ERS marker GRP78, enhanced cellular activity, and increased the generation of reactive oxygen species (ROS) and anti-inflammatory cytokines (IL-10), while also inhibiting HS-induced apoptosis, ROS production, and excessive release of inflammatory cytokines (TNF-α and IL-1ß) in mouse BV2 cells. CONCLUSION: ß-Hydroxybutyric acid may be an effective agent for preventing neuroinflammation in HS mice, possibly due to its ability to inhibit ERS and subsequent microglia neuroinflammation via the gut-brain axis. These findings lay the groundwork for future research and development of BHBA as a preventive drug for HS and provide fresh insights into techniques for treating neurological illnesses by modifying the gut microbiota.


Subject(s)
3-Hydroxybutyric Acid , Brain-Gut Axis , Disease Models, Animal , Endoplasmic Reticulum Stress , Gastrointestinal Microbiome , Mice, Inbred C57BL , Neuroinflammatory Diseases , Animals , Mice , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Brain-Gut Axis/physiology , Brain-Gut Axis/drug effects , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/drug therapy , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Male , 3-Hydroxybutyric Acid/pharmacology , Heat Stress Disorders/metabolism , Endoplasmic Reticulum Chaperone BiP , Neuroprotective Agents/pharmacology , Heat-Shock Response/physiology , Heat-Shock Response/drug effects
6.
Front Cell Infect Microbiol ; 14: 1324794, 2024.
Article in English | MEDLINE | ID: mdl-39015337

ABSTRACT

Background: Despite mounting evidence of gut-brain involvement in psychiatric conditions, functional data remain limited, and analyses of other microbial niches, such as the vaginal microbiota, are lacking in relation to mental health. This aim of this study was to investigate if the connections between the gut microbiome and mental health observed in populations with a clinical diagnosis of mental illness extend to healthy women experiencing stress and depressive symptoms. Additionally, this study examined the functional pathways of the gut microbiota according to the levels of psychological symptoms. Furthermore, the study aimed to explore potential correlations between the vaginal microbiome and mental health parameters in young women without psychiatric diagnoses. Methods: In this cross-sectional study, 160 healthy Danish women (aged 18-40 years) filled out questionnaires with validated scales measuring symptoms of stress and depression and frequency of dietary intake. Fecal and vaginal microbiota samples were collected at the beginning of the menstrual cycle and vaginal samples were also collected at cycle day 8-12 and 18-22. Shotgun metagenomic profiling of the gut and vaginal microbiome was performed. The Kyoto Encyclopedia of Genes and Genomes (KEGG) was used for functional profiling and 56 Gut Brain Modules were analyzed in the fecal samples. Results: The relative abundance in the gut of the genera Escherichia, Parabacteroides, and Shigella was higher in women with elevated depressive symptoms. Women with high perceived stress showed a tendency of increased abundance of Escherichia, Shigella, and Blautia. Amongst others, the potentially pathogenic genera, Escherichia and Shigella correlate with alterations in the neuroactive pathways such as the glutamatergic, GABAeric, dopaminergic, and Kynurenine pathways. Vaginosis symptoms were more prevalent in women reporting high levels of stress and depressive symptoms. Conclusions: The findings of this study support the concept of a microbiota-associated effect on the neuroactive pathways even in healthy young women. This suggest, that targeting the gut microbiome could be a promising approach for future psychiatric interventions.


Subject(s)
Depression , Feces , Gastrointestinal Microbiome , Stress, Psychological , Vagina , Humans , Female , Adult , Young Adult , Cross-Sectional Studies , Adolescent , Depression/microbiology , Vagina/microbiology , Feces/microbiology , Stress, Psychological/microbiology , Microbiota , Denmark , Healthy Volunteers , Brain-Gut Axis/physiology , Surveys and Questionnaires , Metagenomics/methods , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification
7.
Nutrients ; 16(13)2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38999791

ABSTRACT

With the recognition of the importance of the gut-brain axis in Parkinson's disease (PD) etiology, there is increased interest in developing therapeutic strategies that target α-synuclein, the hallmark abhorrent protein of PD pathogenesis, which may originate in the gut. Research has demonstrated that inhibiting the aggregation, oligomerization, and fibrillation of α-synuclein are key strategies for disease modification. Polyphenols, which are rich in fruits and vegetables, are drawing attention for their potential role in this context. In this paper, we reviewed how polyphenols influence the composition and functional capabilities of the gut microbiota and how the resulting microbial metabolites of polyphenols may potentially enhance the modulation of α-synuclein aggregation. Understanding the interaction between polyphenols and gut microbiota and identifying which specific microbes may enhance the efficacy of polyphenols is crucial for developing therapeutic strategies and precision nutrition based on the microbiome.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Parkinson Disease , Polyphenols , alpha-Synuclein , Parkinson Disease/metabolism , Parkinson Disease/microbiology , Parkinson Disease/drug therapy , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Polyphenols/pharmacology , Humans , alpha-Synuclein/metabolism , Brain-Gut Axis/physiology , Animals , Brain/metabolism , Brain/drug effects
8.
J Neuroimmune Pharmacol ; 19(1): 36, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39042202

ABSTRACT

Newly conducted research suggests that metabolic disorders, like diabetes and obesity, play a significant role as risk factors for psychiatric disorders. This connection presents a potential avenue for creating novel antidepressant medications by repurposing drugs originally developed to address antidiabetic conditions. Earlier investigations have shown that GLP-1 (Glucagon-like Peptide-1) analogs exhibit neuroprotective qualities in various models of neurological diseases, encompassing conditions such as Alzheimer's disease, Parkinson's disease, and stroke. Moreover, GLP-1 analogs have demonstrated the capability to enhance neurogenesis, a process recognized for its significance in memory formation and the cognitive and emotional aspects of information processing. Nonetheless, whether semaglutide holds efficacy as both an antidepressant and anxiolytic agent remains uncertain. To address this, our study focused on a mouse model of depression linked to type 2 diabetes induced by a High Fat Diet (HFD). In this model, we administered semaglutide (0.05 mg/Kg intraperitoneally) on a weekly basis to evaluate its potential as a therapeutic option for depression and anxiety. Diabetic mice had higher blood glucose, lipidic profile, and insulin resistance. Moreover, mice fed HFD showed higher serum interleukin (IL)-1ß and lipopolysaccharide (LPS) associated with impaired humor and cognition. The analysis of behavioral responses revealed that the administration of semaglutide effectively mitigated depressive- and anxiety-like behaviors, concurrently demonstrating an enhancement in cognitive function. Additionally, semaglutide treatment protected synaptic plasticity and reversed the hippocampal neuroinflammation induced by HFD fed, improving activation of the insulin pathway, demonstrating the protective effects of semaglutide. We also found that semaglutide treatment decreased astrogliosis and microgliosis in the dentate gyrus region of the hippocampus. In addition, semaglutide prevented the DM2-induced impairments of pro-opiomelanocortin (POMC), and G-protein-coupled receptor 43 (GPR43) and simultaneously increased the NeuN + and Glucagon-like Peptide-1 receptor (GLP-1R+) neurons in the hippocampus. Our data also showed that semaglutide increased the serotonin (5-HT) and serotonin transporter (5-HTT) and glutamatergic receptors in the hippocampus. At last, semaglutide changed the gut microbiota profile (increasing Bacterioidetes, Bacteroides acidifaciens, and Blautia coccoides) and decreased leaky gut, improving the gut-brain axis. Taken together, semaglutide has the potential to act as a therapeutic tool for depression and anxiety.


Subject(s)
Anxiety , Brain-Gut Axis , Cognitive Dysfunction , Depression , Diabetes Mellitus, Type 2 , Gastrointestinal Microbiome , Glucagon-Like Peptides , Mice, Inbred C57BL , Animals , Glucagon-Like Peptides/pharmacology , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/psychology , Diabetes Mellitus, Type 2/metabolism , Mice , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/prevention & control , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Depression/drug therapy , Depression/psychology , Depression/metabolism , Male , Anxiety/drug therapy , Anxiety/psychology , Anxiety/etiology , Gastrointestinal Microbiome/drug effects , Brain-Gut Axis/drug effects , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/psychology , Diabetes Mellitus, Experimental/metabolism , Disease Models, Animal , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
10.
Gut Microbes ; 16(1): 2371950, 2024.
Article in English | MEDLINE | ID: mdl-39008552

ABSTRACT

The gut microbial ecosystem communicates bidirectionally with the brain in what is known as the gut-microbiome-brain axis. Bidirectional signaling occurs through several pathways including signaling via the vagus nerve, circulation of microbial metabolites, and immune activation. Alterations in the gut microbiota are implicated in Alzheimer's disease (AD), a progressive neurodegenerative disease. Perturbations in gut microbial communities may affect pathways within the gut-microbiome-brain axis through altered production of microbial metabolites including ɣ-aminobutyric acid (GABA), the primary inhibitory mammalian neurotransmitter. GABA has been shown to act on gut integrity through modulation of gut mucins and tight junction proteins and may be involved in vagus nerve signal inhibition. The GABAergic signaling pathway has been shown to be dysregulated in AD, and may be responsive to interventions. Gut microbial production of GABA is of recent interest in neurological disorders, including AD. Bacteroides and Lactic Acid Bacteria (LAB), including Lactobacillus, are predominant producers of GABA. This review highlights how temporal alterations in gut microbial communities associated with AD may affect the GABAergic signaling pathway, intestinal barrier integrity, and AD-associated inflammation.


Subject(s)
Alzheimer Disease , Brain-Gut Axis , Gastrointestinal Microbiome , Signal Transduction , gamma-Aminobutyric Acid , Alzheimer Disease/metabolism , Alzheimer Disease/microbiology , Humans , gamma-Aminobutyric Acid/metabolism , Animals , Brain-Gut Axis/physiology , Brain/metabolism , Bacteria/metabolism , Bacteria/classification
11.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(4): 508-515, 2024 Apr 28.
Article in English, Chinese | MEDLINE | ID: mdl-39019779

ABSTRACT

Gut microbiota refers to the vast and diverse community of microorganisms residing in the intestines. Factors such as genetics, environmental influences (e.g., exercise, diet), and early life experiences (e.g., infant feeding methods) can all affect the ecological balance of gut microbiota within the body. Dysbiosis of the gut microbiota is associated with extra-intestinal diseases such as Parkinson's syndrome, osteoporosis, and autoimmune diseases, suggesting that disturbances in gut microbiota may be one of the causes of these diseases. Exercise benefits various diseases, with gut microbiota playing a role in regulating the nervous, musculoskeletal, and immune systems. Gut microbiota can impact the body's health status through the gut-brain axis, gut-muscle axis, and immune pathways. Moderate-intensity aerobic exercise can increase the quantity of gut microbiota and change microbial abundance, although short-term exercise does not significantly affect the alpha diversity of the microbiota. Resistance exercise also does not have a significant regulatory effect on gut microbiota.


Subject(s)
Exercise , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Exercise/physiology , Dysbiosis/microbiology , Brain-Gut Axis/physiology , Parkinson Disease/microbiology , Osteoporosis/microbiology , Osteoporosis/prevention & control
12.
Gut Microbes ; 16(1): 2360233, 2024.
Article in English | MEDLINE | ID: mdl-38949979

ABSTRACT

Functional gastrointestinal disorders (FGIDs), chronic disorders characterized by either abdominal pain, altered intestinal motility, or their combination, have a worldwide prevalence of more than 40% and impose a high socioeconomic burden with a significant decline in quality of life. Recently, FGIDs have been reclassified as disorders of gut-brain interaction (DGBI), reflecting the key role of the gut-brain bidirectional communication in these disorders and their impact on psychological comorbidities. Although, during the past decades, the field of DGBIs has advanced significantly, the molecular mechanisms underlying DGBIs pathogenesis and pathophysiology, and the role of the gut microbiome in these processes are not fully understood. This review aims to discuss the latest body of literature on the complex microbiota-gut-brain interactions and their implications in the pathogenesis of DGBIs. A better understanding of the existing communication pathways between the gut microbiome and the brain holds promise in developing effective therapeutic interventions for DGBIs.


Subject(s)
Brain-Gut Axis , Brain , Gastrointestinal Diseases , Gastrointestinal Microbiome , Gastrointestinal Microbiome/physiology , Humans , Brain-Gut Axis/physiology , Gastrointestinal Diseases/microbiology , Gastrointestinal Diseases/physiopathology , Brain/microbiology , Brain/physiopathology , Animals , Gastrointestinal Tract/microbiology
14.
Nutrients ; 16(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39064664

ABSTRACT

This review summarizes the relationship between diet, the gut microbiome, and migraine. Key findings reveal that certain dietary factors, such as caffeine and alcohol, can trigger migraine, while nutrients like magnesium and riboflavin may help alleviate migraine symptoms. The gut microbiome, through its influence on neuroinflammation (e.g., vagus nerve and cytokines), gut-brain signaling (e.g., gamma-aminobutyric acid), and metabolic function (e.g., short-chain fatty acids), plays a crucial role in migraine susceptibility. Migraine can also alter eating behaviors, leading to poor nutritional choices and further exacerbating the condition. Individual variability in diet and microbiome composition highlights the need for personalized dietary and prebiotic interventions. Epidemiological and clinical data support the effectiveness of tailored nutritional approaches, such as elimination diets and the inclusion of beneficial nutrients, in managing migraine. More work is needed to confirm the role of prebiotics, probiotics, and potentially fecal microbiome translation in the management of migraine. Future research should focus on large-scale studies to elucidate the underlying mechanisms of bidirectional interaction between diet and migraine and develop evidence-based clinical guidelines. Integrating dietary management, gut health optimization, and lifestyle modifications can potentially offer a holistic approach to reducing migraine frequency and severity, ultimately improving patient outcomes and quality of life.


Subject(s)
Brain-Gut Axis , Diet , Gastrointestinal Microbiome , Migraine Disorders , Humans , Gastrointestinal Microbiome/physiology , Migraine Disorders/microbiology , Migraine Disorders/therapy , Brain-Gut Axis/physiology , Brain , Feeding Behavior/physiology , Prebiotics/administration & dosage , Probiotics/therapeutic use
15.
Nutrients ; 16(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39064675

ABSTRACT

Schizophrenia is a disease with a complex etiology that significantly impairs the functioning of patients. In recent years, there has been increasing focus on the importance of the gut microbiota in the context of the gut-brain axis. In our study, we analyzed data on the gut-brain axis in relation to schizophrenia, as well as the impacts of eating habits, the use of various supplements, and diets on schizophrenia. Additionally, the study investigated the impact of antipsychotics on the development of metabolic disorders, such as diabetes, dyslipidemia, and obesity. There may be significant clinical benefits to be gained from therapies supported by supplements such as omega-3 fatty acids, B vitamins, and probiotics. The results suggest the need for a holistic approach to the treatment of schizophrenia, incorporating both drug therapy and dietary interventions.


Subject(s)
Brain-Gut Axis , Diet , Dietary Supplements , Gastrointestinal Microbiome , Probiotics , Schizophrenia , Humans , Gastrointestinal Microbiome/drug effects , Probiotics/administration & dosage , Antipsychotic Agents/administration & dosage , Fatty Acids, Omega-3/administration & dosage , Feeding Behavior
16.
Nutrients ; 16(14)2024 Jul 21.
Article in English | MEDLINE | ID: mdl-39064809

ABSTRACT

Alzheimer's disease is the most common cause of dementia globally. The pathogenesis is multifactorial and includes deposition of amyloid-ß in the central nervous system, presence of intraneuronal neurofibrillary tangles and a decreased amount of synapses. It remains uncertain what causes the progression of the disease. Nowadays, it is suggested that the brain is connected to the gastrointestinal tract, especially the enteric nervous system and gut microbiome. Studies have found a positive association between AD and gastrointestinal diseases such as periodontitis, Helicobacter pylori infection, inflammatory bowel disease and microbiome disorders. H. pylori and its metabolites can enter the CNS via the oropharyngeal olfactory pathway and may predispose to the onset and progression of AD. Periodontitis may cause systemic inflammation of low severity with high levels of pro-inflammatory cytokines and neutrophils. Moreover, lipopolysaccharide from oral bacteria accompanies beta-amyloid in plaques that form in the brain. Increased intestinal permeability in IBS leads to neuronal inflammation from transference. Chronic inflammation may lead to beta-amyloid plaque formation in the intestinal tract that spreads to the brain via the vagus nerve. The microbiome plays an important role in many bodily functions, such as nutrient absorption and vitamin production, but it is also an important factor in the development of many diseases, including Alzheimer's disease. Both the quantity and diversity of the microbiome change significantly in patients with AD and even in people in the preclinical stage of the disease, when symptoms are not yet present. The microbiome influences the functioning of the central nervous system through, among other things, the microbiota-gut-brain axis. Given the involvement of the microbiome in the pathogenesis of AD, antibiotic therapy, probiotics and prebiotics, and faecal transplantation are being considered as possible therapeutic options.


Subject(s)
Alzheimer Disease , Gastrointestinal Diseases , Gastrointestinal Microbiome , Humans , Alzheimer Disease/microbiology , Gastrointestinal Diseases/microbiology , Brain-Gut Axis/physiology , Helicobacter Infections/complications , Helicobacter Infections/microbiology , Periodontitis/microbiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Helicobacter pylori , Inflammatory Bowel Diseases/microbiology , Inflammatory Bowel Diseases/metabolism , Brain/metabolism
17.
Exp Gerontol ; 194: 112507, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38971546

ABSTRACT

Gut microbiota and metabolites are considered key factors in the pathogenesis of perioperative neurocognitive disorders (PND), and the brain-gut axis may be a promising target for PND treatment. Electroacupuncture has been shown to improve a wide range of neurological disorders and to restore function to the gastrointestinal tract. Thus, we hypothesized whether electroacupuncture could remodel gut microbiota and neuroinflammation induced by anesthesia/surgery. First, we observed electroacupuncture at acupoints GV20, LI4 and PC6 significantly improved memory in behavioral tests. Next, we found electroacupuncture decreased the levels of inflammatory factors (NSE, S-100ß, IL-6, etc.) in the hippocampus, indicating that nerve inflammation was blocked by electroacupuncture. Furthermore, via 16S rRNA sequence analysis and LC-MS analysis, the gut microbiota and its metabolites were appropriately restored after electroacupuncture treatment. Additionally, we further confirmed the restorative effect of electroacupuncture on PND by fecal transplantation. In conclusion, the role of electroacupuncture in improving cognitive function and protecting neurons may be related to the modulation of gut microbiota and their metabolite dysregulation, thereby inhibiting neuroinflammation in PND mice.


Subject(s)
Electroacupuncture , Gastrointestinal Microbiome , Hippocampus , Animals , Electroacupuncture/methods , Gastrointestinal Microbiome/physiology , Male , Mice , Hippocampus/metabolism , Mice, Inbred C57BL , Brain-Gut Axis/physiology , Fecal Microbiota Transplantation , Disease Models, Animal , Neurocognitive Disorders/therapy , Neurocognitive Disorders/metabolism , Neuroinflammatory Diseases/metabolism , Memory , Cognition
18.
Poult Sci ; 103(8): 103966, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959642

ABSTRACT

Leg disorders have become increasingly common in broilers, leading to lower meat quality and major economic losses. This study evaluated the effects of dietary supplementation with Clostridium butyricum (C. butyricum) and 25-hydroxyvitamin D3 (25-OH-D3) on bone development by comparing growth performance, tibial parameters, Ca and P contents of tibial ash, bone development-related indicators' level, and cecal short-chain fatty acids in Cobb broilers. All birds were divided into four treatment groups, which birds fed either a basal diet (Con), basal diet + 75 mg chlortetracycline/kg (Anti), basal diet + C. butyricum at 109 CFU/kg (Cb), basal diet + C. butyricum at 109 CFU/kg and 25-OH-D3 at 25 µg/kg (CbD), or basal diet + 25-OH-D3 at 25 µg/kg (CD). Our results suggest that the dietary supplementation in Cb, CbD, and CD significantly increased the body weight (BW) and average daily gain (ADG), and reduced the feed-to-weight ratio (F/G) at different stages of growth (P < 0.05). Dietary supplementation in Cb, CbD, and CD prolonged (P < 0.05) the behavioral responses latency-to-lie (LTL) time, reduced (P < 0.05) the levels of osteocalcin (BGP) and peptide tyrosine (PYY), and increased (P < 0.05) serotonin (5-HT) and dopamine (DA). Treatment with Cb increased (P < 0.05) the levels of acetic acid, isobutyric acid, butyric acid, and isovaleric acid compared with those in Con group. The cecal metagenome showed that Alistipes spp. were significantly more abundant in Cb, CbD, and CD groups (P < 0.05). A total of 12 metabolic pathways were significantly affected by supplementation, including the signaling pathways of glucagon, insulin, and PI3K-AKT; primary and secondary bile acid biosynthesis; and P-type Ca 2+ transporters (P < 0.05). Hence, the CbD supplementation modulates bone metabolism by regulating the mediators of gut-brain axis, which may inform strategies to prevent leg diseases and improve meat quality in broilers.


Subject(s)
Animal Feed , Calcifediol , Chickens , Clostridium butyricum , Diet , Dietary Supplements , Animals , Chickens/physiology , Clostridium butyricum/physiology , Animal Feed/analysis , Diet/veterinary , Calcifediol/administration & dosage , Calcifediol/pharmacology , Dietary Supplements/analysis , Brain-Gut Axis/physiology , Brain-Gut Axis/drug effects , Probiotics/pharmacology , Probiotics/administration & dosage , Male , Bone and Bones/drug effects , Random Allocation , Gastrointestinal Microbiome/drug effects
19.
Food Funct ; 15(15): 7865-7882, 2024 Jul 29.
Article in English | MEDLINE | ID: mdl-38967039

ABSTRACT

Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized primarily by cognitive impairment. Recent investigations have highlighted the potential of nutritional interventions that target the gut-brain axis, such as probiotics and prebiotics, in forestalling the onset of AD. In this study, whole-genome sequencing was employed to identify xylan as the optimal carbon source for the tryptophan metabolism regulating probiotic Clostridium sporogenes (C. sporogenes). Subsequent in vivo studies demonstrated that administration of a synbiotic formulation comprising C. sporogenes (1 × 1010 CFU per day) and xylan (1%, w/w) over a duration of 30 days markedly enhanced cognitive performance and spatial memory faculties in the 5xFAD transgenic AD mouse model. The synbiotic treatment significantly reduced amyloid-ß (Aß) accumulation in the cortex and hippocampus of the brain. Importantly, synbiotic therapy substantially restored the synaptic ultrastructure in AD mice and suppressed neuroinflammatory responses. Moreover, the intervention escalated levels of the microbial metabolite indole-3-propionic acid (IPA) and augmented the relative prevalence of IPA-synthesizing bacteria, Lachnospira and Clostridium, while reducing the dominant bacteria in AD, such as Aquabacterium, Corynebacterium, and Romboutsia. Notably, synbiotic treatment also prevented the disruption of gut barrier integrity. Correlation analysis indicated a strong positive association between gut microbiota-generated IPA levels and behavioral changes. In conclusion, this study demonstrates that synbiotic supplementation significantly improves cognitive and intellectual deficits in 5xFAD mice, which could be partly attributed to enhanced IPA production by gut microbiota. These findings provide a theoretical basis for considering synbiotic therapy as a novel microbiota-targeted approach for the treatment of metabolic and neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Clostridium , Cognitive Dysfunction , Disease Models, Animal , Gastrointestinal Microbiome , Indoles , Mice, Transgenic , Synbiotics , Xylans , Animals , Alzheimer Disease/therapy , Alzheimer Disease/metabolism , Mice , Synbiotics/administration & dosage , Indoles/metabolism , Cognitive Dysfunction/therapy , Cognitive Dysfunction/metabolism , Xylans/metabolism , Xylans/pharmacology , Clostridium/metabolism , Male , Amyloid beta-Peptides/metabolism , Humans , Propionates/metabolism , Brain-Gut Axis/physiology
20.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062876

ABSTRACT

Functional gastrointestinal disorders (FGIDs) are characterized by chronic gastrointestinal symptoms in the absence of overt pathology and affect a significant percentage of the worldwide population. They are commonly accompanied by co-morbid psychiatric symptomatology and are associated with significant suffering and great healthcare services utilization. There is growing evidence that dysregulation of the gut-brain axis and disturbances in the processing of afferent interoceptive signals lie at the heart of these disorders. In this context, the aim of the current review was to detect and critically review original articles focusing on the role of interoception in the pathophysiology of FGIDs. Our search yielded 38 relevant studies. FGID patients displayed increased visceral sensitivity, enhanced attention to gastrointestinal interoceptive cues, and greater emotional arousal when coping with gut-derived sensations. Neuroimaging studies have shown significant structural and functional changes in regions of the interoceptive network, while molecular and genetic studies have revealed significant associations between interoceptive signaling and deficits in excitatory neurotransmission, altered endocrine and immune physiological pathways, and aberrant expression of transient receptor potential channel genes. Finally, there were emerging data suggesting that interoception-based interventions may reduce physical symptoms and improve quality of life and should be integrated into FGID clinical management practices.


Subject(s)
Gastrointestinal Diseases , Interoception , Humans , Interoception/physiology , Gastrointestinal Diseases/physiopathology , Brain-Gut Axis
SELECTION OF CITATIONS
SEARCH DETAIL