Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 833
Filter
1.
Gut Microbes ; 16(1): 2363011, 2024.
Article in English | MEDLINE | ID: mdl-38835220

ABSTRACT

The Mediterranean diet (MD) and its bioactive constituents have been advocated for their neuroprotective properties along with their capacity to affect gut microbiota speciation and metabolism. Mediated through the gut brain axis, this modulation of the microbiota may partly contribute to the neuroprotective properties of the MD. To explore this potential interaction, we evaluated the neuroprotective properties of a novel bioactive blend (Neurosyn240) resembling the Mediterranean diet in a rodent model of chronic low-grade inflammation. Behavioral tests of cognition, brain proteomic analysis, 16S rRNA sequencing, and 1H NMR metabolomic analyses were employed to develop an understanding of the gut-brain axis interactions involved. Recognition memory, as assessed by the novel object recognition task (NOR), decreased in response to LPS insult and was restored with Neurosyn240 supplementation. Although the open field task performance did not reach significance, it correlated with NOR performance indicating an element of anxiety related to this cognitive change. Behavioral changes associated with Neurosyn240 were accompanied by a shift in the microbiota composition which included the restoration of the Firmicutes: Bacteroidota ratio and an increase in Muribaculum, Rikenellaceae Alloprevotella, and most notably Akkermansia which significantly correlated with NOR performance. Akkermansia also correlated with the metabolites 5-aminovalerate, threonine, valine, uridine monophosphate, and adenosine monophosphate, which in turn significantly correlated with NOR performance. The proteomic profile within the brain was dramatically influenced by both interventions, with KEGG analysis highlighting oxidative phosphorylation and neurodegenerative disease-related pathways to be modulated. Intriguingly, a subset of these proteomic changes simultaneously correlated with Akkermansia abundance and predominantly related to oxidative phosphorylation, perhaps alluding to a protective gut-brain axis interaction. Collectively, our results suggest that the bioactive blend Neurosyn240 conferred cognitive and microbiota resilience in response to the deleterious effects of low-grade inflammation.


Subject(s)
Cognition , Diet, Mediterranean , Dietary Supplements , Disease Models, Animal , Gastrointestinal Microbiome , Inflammation , Animals , Gastrointestinal Microbiome/drug effects , Mice , Male , Cognition/drug effects , Inflammation/metabolism , Inflammation/diet therapy , Dietary Supplements/analysis , Mice, Inbred C57BL , Brain-Gut Axis/physiology , Brain/metabolism , Bacteria/classification , Bacteria/metabolism , Bacteria/isolation & purification , Bacteria/genetics
2.
J Affect Disord ; 360: 229-241, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38823591

ABSTRACT

A high-fat diet can modify the composition of gut microbiota, resulting in dysbiosis. Changes in gut microbiota composition can lead to increased permeability of the gut barrier, allowing bacterial products like lipopolysaccharides (LPS) to enter circulation. This process can initiate systemic inflammation and contribute to neuroinflammation. Empagliflozin (EF), an SGLT2 inhibitor-type hypoglycemic drug, has been reported to treat neuroinflammation. However, there is a lack of evidence showing that EF regulates the gut microbiota axis to control neuroinflammation in HFD models. In this study, we explored whether EF could improve neuroinflammation caused by an HFD via regulation of the gut microbiota and the mechanism underlying this phenomenon. Our data revealed that EF alleviates pathological brain injury, reduces the reactive proliferation of astrocytes, and increases the expression of synaptophysin. In addition, the levels of inflammatory factors in hippocampal tissue were significantly decreased after EF intervention. Subsequently, the results of 16S rRNA gene sequencing showed that EF could change the microbial community structure of mice, indicating that the abundance of Lactococcus, Ligilactobacillus and other microbial populations decreased dramatically. Therefore, EF alleviates neuroinflammation by inhibiting gut microbiota-mediated astrocyte activation in the brains of high-fat diet-fed mice. Our study focused on the gut-brain axis, and broader research on neuroinflammation can provide a more holistic understanding of the mechanisms driving neurodegenerative diseases and inform the development of effective strategies to mitigate their impact on brain health. The results provide strong evidence supporting the larger clinical application of EF.


Subject(s)
Astrocytes , Benzhydryl Compounds , Diet, High-Fat , Gastrointestinal Microbiome , Glucosides , Neuroinflammatory Diseases , Animals , Gastrointestinal Microbiome/drug effects , Diet, High-Fat/adverse effects , Astrocytes/drug effects , Glucosides/pharmacology , Mice , Benzhydryl Compounds/pharmacology , Neuroinflammatory Diseases/drug therapy , Male , Mice, Inbred C57BL , Brain/drug effects , Brain-Gut Axis/drug effects , Disease Models, Animal , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Hippocampus/drug effects , Hippocampus/metabolism , Dysbiosis
3.
Life Sci ; 350: 122748, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38843992

ABSTRACT

Neurodegenerative diseases (NDs) are a group of heterogeneous disorders with a high socioeconomic burden. Although pharmacotherapy is currently the principal therapeutic approach for the management of NDs, mounting evidence supports the notion that the protracted application of available drugs would abate their dopaminergic outcomes in the long run. The therapeutic application of microbiome-based modalities has received escalating attention in biomedical works. In-depth investigations of the bidirectional communication between the microbiome in the gut and the brain offer a multitude of targets for the treatment of NDs or maximizing the patient's quality of life. Probiotic administration is a well-known microbial-oriented approach to modulate the gut microbiota and potentially influence the process of neurodegeneration. Of note, there is a strong need for further investigation to map out the mechanistic prospects for the gut-brain axis and the clinical efficacy of probiotics. In this review, we discuss the importance of microbiome modulation and hemostasis via probiotics, prebiotics, postbiotics and synbiotics in ameliorating pathological neurodegenerative events. Also, we meticulously describe the underlying mechanism of action of probiotics and their metabolites on the gut-brain axis in different NDs. We suppose that the present work will provide a functional direction for the use of probiotic-based modalities in promoting current practical treatments for the management of neurodegenerative-related diseases.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Neurodegenerative Diseases , Probiotics , Probiotics/therapeutic use , Humans , Gastrointestinal Microbiome/physiology , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/therapy , Brain-Gut Axis/physiology , Animals , Brain/metabolism , Prebiotics/administration & dosage
4.
Nan Fang Yi Ke Da Xue Xue Bao ; 44(5): 876-884, 2024 May 20.
Article in Chinese | MEDLINE | ID: mdl-38862445

ABSTRACT

OBJECTIVE: To investigate the mechanisms that mediate the neuroprotective effect of the intestinal microbial metabolite sodium butyrate (NaB) in a mouse model of Parkinson's disease (PD) via the gut-brain axis. METHODS: Thirty-nine 7-week-old male C57BL/6J mice were randomized equally into control group, PD model group, and NaB treatment group. In the latter two groups, PD models were established by intraperitoneal injection of 30 mg/kg 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) once daily for 5 consecutive days, and normal saline was injected in the control group. After modeling, the mice received daily gavage of NaB (300 mg/kg) or an equal volume of saline for 14 days. Behavioral tests were carried out to assess the changes in motor function of the mice, and Western blotting was performed to detect the expressions of tyrosine hydroxylase (TH) and α-synuclein (α-syn) in the striatum and nuclear factor-κB (NF-κB), tumor necrosis factor (TNF-α), interleukin 6 (IL-6), and the tight junction proteins ZO-1, Occludin, and Claudinin the colon. HE staining was used to observe inflammatory cell infiltration in the colon of the mice. RNA sequencing analysis was performed to identify the differentially expressed genes in mouse colon tissues, and their expressions were verified using qRT-PCR and Western blotting. RESULTS: The mouse models of PD with NaB treatment showed significantly increased movement speed and pulling strength of the limbs with obviously upregulated expressions of TH, Occludin, and Claudin and downregulated expressions of α-syn, NF-κB, TNF-α, and IL-6 (all P < 0.05). HE staining showed that NaB treatment significantly ameliorated inflammatory cell infiltration in the colon of the PD mice. RNA sequencing suggested that Bmal1 gene probably mediated the neuroprotective effect of NaB in PD mice (P < 0.05). CONCLUSION: NaB can improve motor dysfunction, reduce dopaminergic neuron loss in the striatum, and ameliorate colonic inflammation in PD mice possibly through a mechanism involving Bmal1.


Subject(s)
Butyric Acid , Disease Models, Animal , Mice, Inbred C57BL , Neuroprotective Agents , Parkinson Disease , Animals , Mice , Butyric Acid/pharmacology , Butyric Acid/therapeutic use , Male , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Parkinson Disease/drug therapy , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Tumor Necrosis Factor-alpha/metabolism , NF-kappa B/metabolism , Interleukin-6/metabolism , Tyrosine 3-Monooxygenase/metabolism , Tyrosine 3-Monooxygenase/genetics , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Corpus Striatum/metabolism , Occludin/metabolism , Occludin/genetics , Brain-Gut Axis
5.
Biomed Pharmacother ; 175: 116601, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38749177

ABSTRACT

Alzheimer's disease (AD) comprises a group of neurodegenerative disorders with some changes in the brain, which could lead to the deposition of certain proteins and result in the degeneration and death of brain cells. Patients with AD manifest primarily as cognitive decline, psychiatric symptoms, and behavioural disorders. Short-chain fatty acids (SCFAs) are a class of saturated fatty acids (SFAs) produced by gut microorganisms through the fermentation of dietary fibre ingested. SCFAs, as a significant mediator of signalling, can have diverse physiological and pathological roles in the brain through the gut-brain axis, and play a positive effect on AD via multiple pathways. Firstly, differences in SCFAs and microbial changes have been stated in AD cases of humans and mice in this paper. And then, mechanisms of three main SCFAs in treating with AD have been summarized, as well as differences of gut bacteria. Finally, functions of SCFAs played in regulating intestinal flora homeostasis, modulating the immune system, and the metabolic system, which were considered to be beneficial for the treatment of AD, have been elucidated, and the key roles of gut bacteria and SCFAs were pointed out. All in all, this paper provides an overview of SCFAs and gut bacteria in AD, and can help people to understand the importance of gut-brain axis in AD.


Subject(s)
Alzheimer Disease , Brain-Gut Axis , Brain , Fatty Acids, Volatile , Gastrointestinal Microbiome , Humans , Fatty Acids, Volatile/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/microbiology , Animals , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Brain/metabolism
6.
Front Immunol ; 15: 1365673, 2024.
Article in English | MEDLINE | ID: mdl-38817603

ABSTRACT

Importance: Research is beginning to elucidate the sophisticated mechanisms underlying the microbiota-gut-brain-immune interface, moving from primarily animal models to human studies. Findings support the dynamic relationships between the gut microbiota as an ecosystem (microbiome) within an ecosystem (host) and its intersection with the host immune and nervous systems. Adding this to the effects on epigenetic regulation of gene expression further complicates and strengthens the response. At the heart is inflammation, which manifests in a variety of pathologies including neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis (MS). Observations: Generally, the research to date is limited and has focused on bacteria, likely due to the simplicity and cost-effectiveness of 16s rRNA sequencing, despite its lower resolution and inability to determine functional ability/alterations. However, this omits all other microbiota including fungi, viruses, and phages, which are emerging as key members of the human microbiome. Much of the research has been done in pre-clinical models and/or in small human studies in more developed parts of the world. The relationships observed are promising but cannot be considered reliable or generalizable at this time. Specifically, causal relationships cannot be determined currently. More research has been done in Alzheimer's disease, followed by Parkinson's disease, and then little in MS. The data for MS is encouraging despite this. Conclusions and relevance: While the research is still nascent, the microbiota-gut-brain-immune interface may be a missing link, which has hampered our progress on understanding, let alone preventing, managing, or putting into remission neurodegenerative diseases. Relationships must first be established in humans, as animal models have been shown to poorly translate to complex human physiology and environments, especially when investigating the human gut microbiome and its relationships where animal models are often overly simplistic. Only then can robust research be conducted in humans and using mechanistic model systems.


Subject(s)
Brain-Gut Axis , Brain , Gastrointestinal Microbiome , Neuroinflammatory Diseases , Humans , Gastrointestinal Microbiome/immunology , Animals , Brain-Gut Axis/immunology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/microbiology , Neuroinflammatory Diseases/etiology , Brain/immunology , Brain/microbiology
7.
Pharmacol Res ; 204: 107214, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38763328

ABSTRACT

Studies have shown that the microbiota-gut-brain axis is highly correlated with the pathogenesis of depression in humans. However, whether independent oral microbiome that do not depend on gut microbes could affect the progression of depression in human beings remains unclear, neither does the presence and underlying mechanisms of the microbiota-oral-brain axis in the development of the condition. Hence this study that encompasses clinical and animal experiments aims at investigating the correlation between oral microbiota and the onset of depression via mediating the microbiota-oral-brain axis. We compared the oral microbial compositions and metabolomes of 87 patients with depressive symptoms versus 70 healthy controls. We found that the oral microbial and metabolic signatures were significantly different between the two groups. Significantly, germ-free (GF) mice transplanted with saliva from mice exposing to chronic restraint stress (CRS) displayed depression-like behavior and oral microbial dysbiosis. This was characterized by a significant differential abundance of bacterial species, including the enrichment of Pseudomonas, Pasteurellaceae, and Muribacter, as well as the depletion of Streptococcus. Metabolomic analysis showed the alternation of metabolites in the plasma of CRS-exposed GF mice, especially Eicosapentaenoic Acid. Furthermore, oral and gut barrier dysfunction caused by CRS-induced oral microbiota dysbiosis may be associated with increased blood-brain barrier permeability. Pseudomonas aeruginosa supplementation exacerbated depression-like behavior, while Eicosapentaenoic Acid treatment conferred protection against depression-like states in mice. These results suggest that oral microbiome and metabolic function dysbiosis may be relevant to the pathogenesis and pathophysiology of depression. The proposed microbiota-oral-brain axis provides a new way and targets for us to study the pathogenesis of depression.


Subject(s)
Depression , Dysbiosis , Stress, Psychological , Animals , Dysbiosis/metabolism , Depression/metabolism , Depression/microbiology , Depression/psychology , Depression/etiology , Male , Humans , Stress, Psychological/metabolism , Stress, Psychological/microbiology , Stress, Psychological/psychology , Female , Adult , Mice , Restraint, Physical/psychology , Mice, Inbred C57BL , Gastrointestinal Microbiome , Brain-Gut Axis , Mouth/microbiology , Middle Aged , Saliva/metabolism , Saliva/microbiology , Behavior, Animal , Blood-Brain Barrier/metabolism
8.
Nat Commun ; 15(1): 4410, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38782979

ABSTRACT

Pancreatic ß cells secrete insulin in response to glucose elevation to maintain glucose homeostasis. A complex network of inter-organ communication operates to modulate insulin secretion and regulate glucose levels after a meal. Lipids obtained from diet or generated intracellularly are known to amplify glucose-stimulated insulin secretion, however, the underlying mechanisms are not completely understood. Here, we show that a Drosophila secretory lipase, Vaha (CG8093), is synthesized in the midgut and moves to the brain where it concentrates in the insulin-producing cells in a process requiring Lipid Transfer Particle, a lipoprotein originating in the fat body. In response to dietary fat, Vaha stimulates insulin-like peptide release (ILP), and Vaha deficiency results in reduced circulatory ILP and diabetic features including hyperglycemia and hyperlipidemia. Our findings suggest Vaha functions as a diacylglycerol lipase physiologically, by being a molecular link between dietary fat and lipid amplified insulin secretion in a gut-brain axis.


Subject(s)
Brain , Drosophila Proteins , Drosophila melanogaster , Insulin Secretion , Insulin , Animals , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Brain/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Brain-Gut Axis/physiology , Lipase/metabolism , Lipase/genetics , Dietary Fats/metabolism , Glucose/metabolism , Fat Body/metabolism , Lipoprotein Lipase/metabolism , Lipoprotein Lipase/genetics , Male
9.
Curr Obes Rep ; 13(2): 214-223, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38760652

ABSTRACT

PURPOSE OF REVIEW: Detail recent advancements in the science on ultra-processed food (UPF) addiction, focusing on estimated prevalence rates and emerging health disparities; progress towards identifying biological underpinnings and behavioral mechanisms; and implications for weight management. RECENT FINDINGS: Notable developments in the field have included: (1) estimating the global prevalence of UPF addiction at 14% of adults and 15% of youths; (2) revealing health disparities for persons of color and those with food insecurity; (3) observing altered functioning across the brain-gut-microbiome axis; (4) providing early evidence for UPF withdrawal; and (5) elucidating poorer weight management outcomes among persons with UPF addiction. The breadth of recent work on UPF addiction illustrates continued scientific and public interest in the construct and its implications for understanding and treating overeating behaviors and obesity. One pressing gap is the lack of targeted interventions for UPF addiction, which may result in more optimal clinical outcomes for this underserved population.


Subject(s)
Fast Foods , Food Addiction , Obesity , Humans , Prevalence , Gastrointestinal Microbiome , Brain-Gut Axis , Health Status Disparities , Food Handling , Food, Processed
10.
Brain Behav Immun ; 119: 878-897, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38710338

ABSTRACT

Metabolites and compounds derived from gut-associated bacteria can modulate numerous physiological processes in the host, including immunity and behavior. Using a model of oral bacterial infection, we previously demonstrated that gut-derived peptidoglycan (PGN), an essential constituent of the bacterial cell envelope, influences female fruit fly egg-laying behavior by activating the NF-κB cascade in a subset of brain neurons. These findings underscore PGN as a potential mediator of communication between gut bacteria and the brain in Drosophila, prompting further investigation into its impact on all brain cells. Through high-resolution mass spectrometry, we now show that PGN fragments produced by gut bacteria can rapidly reach the central nervous system. In Addition, by employing a combination of whole-genome transcriptome analyses, comprehensive genetic assays, and reporter gene systems, we reveal that gut bacterial infection triggers a PGN dose-dependent NF-κB immune response in perineurial glia, forming the continuous outer cell layer of the blood-brain barrier. Furthermore, we demonstrate that persistent PGN-dependent NF-κB activation in perineurial glial cells correlates with a reduction in lifespan and early neurological decline. Overall, our findings establish gut-derived PGN as a critical mediator of the gut-immune-brain axis in Drosophila.


Subject(s)
Brain-Gut Axis , Brain , Gastrointestinal Microbiome , NF-kappa B , Peptidoglycan , Animals , Peptidoglycan/metabolism , NF-kappa B/metabolism , Brain/metabolism , Brain/immunology , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Female , Drosophila , Neuroglia/metabolism , Neuroglia/immunology , Drosophila melanogaster/metabolism , Neurons/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/immunology , Drosophila Proteins/metabolism
11.
Environ Res ; 255: 119169, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38763277

ABSTRACT

Previous studies have identified the exposure to ubiquitous environmental endocrine disruptors may be a risk factor of neurological disorders. However, the effects of fluorene-9-bisphenol (BHPF) in environmental exposure concentrations associated with these disorders are poorly understood. In this study, classic light-dark and social behavior tests were performed on zebrafish larvae and adults exposed BHPF exposure to evaluate social behavioral disorders and the microbiota-gut-brain axis was assessed to reveal the potential mechanisms underlying the behavioral abnormalities observed. Our results demonstrated that zebrafish larvae exposed to an environmentally relevant concentration (0.1 nM) of BHPF for 7 days showed a diminished response to external environmental factors (light or dark). Zebrafish larvae exposed to BHPF for 7 days or adults exposed to BHPF for 30 days at 1 µM displayed significant behavioral inhibition and altered social behaviors, including social recognition, social preference, and social fear contagion, indicating autism-like behaviors were induced by the exposure. BHPF exposure reduced the distribution of Nissl bodies in midbrain neurons and significantly reduced 5-hydroxytryptamine signaling. Oxytocin (OXT) levels and expression of its receptor oxtra in the gut and brain were down-regulated by BHPF exposure. In addition, the expression levels of genes related to the excitation-inhibitory balance of synaptic transmission changed. Microbiomics revealed increased community diversity and altered abundance of some microflora, such as an elevation in Bacillota and Bacteroidota and a decline in Mycoplasmatota in zebrafish guts, which might contribute to the abnormal neural circuits and autism-like behaviors induced by BHPF. Finally, the rescue effect of exogenous OXT on social behavioral defects induced by BHPF exposure was verified in zebrafish, highlighting the crucial role of OXT signaling through gut-brain axis in the regulatory mechanisms of social behaviors affected by BHPF. This study contributes to understanding the effects of environmental BHPF exposure on neuropsychiatric disorders and attracts public attention to the health risks posed by chemicals in aquatic organisms. The potential mental disorders should be considered in the safety assessments of environmental pollutants.


Subject(s)
Brain-Gut Axis , Fluorenes , Oxytocin , Social Behavior , Zebrafish , Animals , Fluorenes/toxicity , Oxytocin/metabolism , Brain-Gut Axis/drug effects , Signal Transduction/drug effects , Endocrine Disruptors/toxicity , Water Pollutants, Chemical/toxicity , Behavior, Animal/drug effects , Larva/drug effects , Phenols/toxicity , Gastrointestinal Microbiome/drug effects
12.
Brain Behav Immun ; 119: 867-877, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38750700

ABSTRACT

The gastrointestinal tract is one of the main organs affected during systemic inflammation and disrupted gastrointestinal motility is a major clinical manifestation. Many studies have investigated the involvement of neuroimmune interactions in regulating colonic motility during localized colonic inflammation, i.e., colitis. However, little is known about how the enteric nervous system and intestinal macrophages contribute to dysregulated motility during systemic inflammation. Given that systemic inflammation commonly results from the innate immune response against bacterial infection, we mimicked bacterial infection by administering lipopolysaccharide (LPS) to rats and assessed colonic motility using ex vivo video imaging techniques. We utilized the Cx3cr1-Dtr rat model of transient depletion of macrophages to investigate the role of intestinal macrophages in regulating colonic motility during LPS infection. To investigate the role of inhibitory enteric neurotransmission on colonic motility following LPS, we applied the nitric oxide synthase inhibitor, Nω-nitro-L-arginine (NOLA). Our results confirmed an increase in colonic contraction frequency during LPS-induced systemic inflammation. However, neither the depletion of intestinal macrophages, nor the suppression of inhibitory enteric nervous system activity impacted colonic motility disruption during inflammation. This implies that the interplay between the enteric nervous system and intestinal macrophages is nuanced, and complex, and further investigation is needed to clarify their joint roles in colonic motility.


Subject(s)
Enteric Nervous System , Gastrointestinal Motility , Inflammation , Lipopolysaccharides , Macrophages , Animals , Lipopolysaccharides/pharmacology , Rats , Gastrointestinal Motility/physiology , Macrophages/metabolism , Inflammation/metabolism , Inflammation/physiopathology , Enteric Nervous System/physiopathology , Enteric Nervous System/metabolism , Male , Brain-Gut Axis/physiology , Colon/metabolism , Gastrointestinal Tract/metabolism , Colitis/physiopathology , Colitis/metabolism , Colitis/chemically induced , Brain/metabolism , Rats, Sprague-Dawley , Gastrointestinal Diseases/physiopathology , Gastrointestinal Diseases/metabolism
13.
Biosens Bioelectron ; 258: 116298, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-38701537

ABSTRACT

Wireless activation of the enteric nervous system (ENS) in freely moving animals with implantable optogenetic devices offers a unique and exciting opportunity to selectively control gastrointestinal (GI) transit in vivo, including the gut-brain axis. Programmed delivery of light to targeted locations in the GI-tract, however, poses many challenges not encountered within the central nervous system (CNS). We report here the development of a fully implantable, battery-free wireless device specifically designed for optogenetic control of the GI-tract, capable of generating sufficient light over large areas to robustly activate the ENS, potently inducing colonic motility ex vivo and increased propulsion in vivo. Use in in vivo studies reveals unique stimulation patterns that increase expulsion of colonic content, likely mediated in part by activation of an extrinsic brain-gut motor pathway, via pelvic nerves. This technology overcomes major limitations of conventional wireless optogenetic hardware designed for the CNS, providing targeted control of specific neurochemical classes of neurons in the ENS and brain-gut axis, for direct modulation of GI-transit and associated behaviours in freely moving animals.


Subject(s)
Enteric Nervous System , Optogenetics , Wireless Technology , Animals , Optogenetics/instrumentation , Enteric Nervous System/physiology , Mice , Wireless Technology/instrumentation , Brain-Gut Axis/physiology , Biosensing Techniques/instrumentation , Equipment Design , Brain/physiology , Mice, Inbred C57BL
14.
Front Immunol ; 15: 1365871, 2024.
Article in English | MEDLINE | ID: mdl-38756771

ABSTRACT

More than 20% of American adults live with a mental disorder, many of whom are treatment resistant or continue to experience symptoms. Other approaches are needed to improve mental health care, including prevention. The role of the microbiome has emerged as a central tenet in mental and physical health and their interconnectedness (well-being). Under normal conditions, a healthy microbiome promotes homeostasis within the host by maintaining intestinal and brain barrier integrity, thereby facilitating host well-being. Owing to the multidirectional crosstalk between the microbiome and neuro-endocrine-immune systems, dysbiosis within the microbiome is a main driver of immune-mediated systemic and neural inflammation that can promote disease progression and is detrimental to well-being broadly and mental health in particular. In predisposed individuals, immune dysregulation can shift to autoimmunity, especially in the presence of physical or psychological triggers. The chronic stress response involves the immune system, which is intimately involved with the gut microbiome, particularly in the process of immune education. This interconnection forms the microbiota-gut-immune-brain axis and promotes mental health or disorders. In this brief review, we aim to highlight the relationships between stress, mental health, and the gut microbiome, along with the ways in which dysbiosis and a dysregulated immune system can shift to an autoimmune response with concomitant neuropsychological consequences in the context of the microbiota-gut-immune-brain axis. Finally, we aim to review evidenced-based prevention strategies and potential therapeutic targets.


Subject(s)
Brain-Gut Axis , Brain , Dysbiosis , Gastrointestinal Microbiome , Mental Disorders , Mental Health , Stress, Psychological , Humans , Gastrointestinal Microbiome/immunology , Brain-Gut Axis/immunology , Stress, Psychological/immunology , Stress, Psychological/microbiology , Dysbiosis/immunology , Mental Disorders/immunology , Mental Disorders/microbiology , Brain/immunology , Animals , Neuroimmunomodulation
15.
Gut Microbes ; 16(1): 2351520, 2024.
Article in English | MEDLINE | ID: mdl-38717832

ABSTRACT

Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Host Microbial Interactions , Animals , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Humans , Brain/microbiology , Brain/metabolism , Brain/physiology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/metabolism , Models, Animal , Mice
16.
Phytomedicine ; 130: 155744, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38763011

ABSTRACT

BACKGROUND: Aging is associated with learning and memory disorder, affecting multiple brain areas, especially the hippocampus. Previous studies have demonstrated trilobatin (TLB), as a natural food additive, can extend the life of Caenorhabditis elegans and exhibit neuroprotection in Alzheimer's disease mice. However, the possible significance of TLB in anti-aging remains elusive. PURPOSE: This study aimed to delve into the physiological mechanism by which TLB ameliorated aging-induced cognitive impairment in senescence-accelerated mouse prone 8 (SAMP8) mice. METHODS: 6-month-old SAMP8 mice were administrated with TLB (5, 10, 20 mg/kg/day, i.g.) for 3 months. The therapeutic effect of TLB on aging-induced cognitive impairment was assessed in mice using behavioral tests and aging score. The gut microbiota composition in fecal samples was analyzed by metagenomic analysis. The protective effects of TLB on blood-brain barrier (BBB) and intestinal barrier were detected by transmission electron microscope, H&E staining and western blot (WB) assay. The inhibitive effects of TLB on inflammation in brain and intestine were assessed using immunofluorescence, WB and ELISA assay. Molecular docking and surface plasma resonance (SPR) assay were utilized to investigate interaction between TLB and sirtuin 2 (SIRT2). RESULTS: Herein, the findings exhibited TLB mitigated aging-induced cognitive impairment, neuron injury and neuroinflammation in hippocampus of aged SAMP8 mice. Moreover, TLB treatment repaired imbalance of gut microbiota in aged SAMP8 mice. Furthermore, TLB alleviated the damage to BBB and intestinal barrier, concomitant with reducing the expression of SIRT2, phosphorylated levels of c-Jun NH2 terminal kinases (JNK) and c-Jun, and expression of MMP9 protein in aged SAMP8 mice. Molecular docking and SPR unveiled TLB combined with SIRT2 and down-regulated SIRT2 protein expression. Mechanistically, the potential mechanism of SIRT2 in TLB that exerted anti-aging effect was validated in vitro. As expected, SIRT2 deficiency attenuated phosphorylated level of JNK in HT22 cells treated with d-galactose. CONCLUSION: These findings reveal, for the first time, SIRT2-mediated brain-gut barriers contribute to aging and aging-related diseases, and TLB can rescue aging-induced cognitive impairment by targeting SIRT2 and restoring gut microbiota disturbance to mediate the brain-gut axis. Overall, this work extends the potential application of TLB as a natural food additive in aging-related diseases.


Subject(s)
Aging , Brain-Gut Axis , Cognitive Dysfunction , Gastrointestinal Microbiome , Sirtuin 2 , Animals , Gastrointestinal Microbiome/drug effects , Cognitive Dysfunction/drug therapy , Mice , Aging/drug effects , Sirtuin 2/metabolism , Male , Brain-Gut Axis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Molecular Docking Simulation , Hippocampus/drug effects , Hippocampus/metabolism , Disease Models, Animal
17.
Asian J Psychiatr ; 97: 104068, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38776563

ABSTRACT

Mental illness is a hidden epidemic in modern science that has gradually spread worldwide. According to estimates from the World Health Organization (WHO), approximately 10% of the world's population suffers from various mental diseases each year. Worldwide, financial and health burdens on society are increasing annually. Therefore, understanding the different factors that can influence mental illness is required to formulate novel and effective treatments and interventions to combat mental illness. Gut microbiota, consisting of diverse microbial communities residing in the gastrointestinal tract, exert profound effects on the central nervous system through the gut-brain axis. The gut-brain axis serves as a conduit for bidirectional communication between the two systems, enabling the gut microbiota to affect emotional and cognitive functions. Dysbiosis, or an imbalance in the gut microbiota, is associated with an increased susceptibility to mental health disorders and psychiatric illnesses. Gut microbiota is one of the most diverse and abundant groups of microbes that have been found to interact with the central nervous system and play important physiological functions in the human gut, thus greatly affecting the development of mental illnesses. The interaction between gut microbiota and mental health-related illnesses is a multifaceted and promising field of study. This review explores the mechanisms by which gut microbiota influences mental health, encompassing the modulation of neurotransmitter production, neuroinflammation, and integrity of the gut barrier. In addition, it emphasizes a thorough understanding of how the gut microbiome affects various psychiatric conditions.


Subject(s)
Brain-Gut Axis , Dysbiosis , Gastrointestinal Microbiome , Mental Disorders , Humans , Gastrointestinal Microbiome/physiology , Mental Disorders/microbiology , Mental Disorders/physiopathology , Brain-Gut Axis/physiology
18.
Sheng Wu Gong Cheng Xue Bao ; 40(5): 1293-1308, 2024 May 25.
Article in Chinese | MEDLINE | ID: mdl-38783798

ABSTRACT

The intestinal microbiota exhibits a strong correlation with the function of the central nervous system, exerting influence on the host brain through neural pathways, immune pathways, and microbial metabolites along the gut-brain axis. Disorders in the composition of the intestinal microbial are closely associated with the onset and progression of neurological disorders, such as depression, Alzheimer's disease, and Parkinson's disease. It has been proven that fecal microbiota transplantation can improve symptoms in animal models of neurological diseases and clinical patients. This paper provides a comprehensive review of the composition and function of the human intestinal microbiota, as well as the intricate the relationship between the human intestinal microbiota and nervous system diseases through the gut-brain axis. Additionally, it delves into the research advancements and underlying mechanism of fecal microbiota transplantation in the treatment of nervous system diseases. These findings offer novel insights and potential avenues for clinical interventions targeting nervous system diseases.


Subject(s)
Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Nervous System Diseases , Humans , Animals , Nervous System Diseases/therapy , Nervous System Diseases/microbiology , Brain-Gut Axis , Parkinson Disease/therapy , Parkinson Disease/microbiology , Alzheimer Disease/therapy , Alzheimer Disease/microbiology , Depression/therapy , Depression/microbiology
19.
Exp Neurol ; 378: 114834, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38789022

ABSTRACT

The goal of this study is to investigate the role of microbiota-gut-brain axis involved in the protective effect of pair-housing on post-stroke depression (PSD). PSD model was induced by occluding the middle cerebral artery (MCAO) plus restraint stress for four weeks. At three days after MCAO, the mice were restrained 2 h per day. For pair-housing (PH), each mouse was pair housed with a healthy isosexual cohabitor for four weeks. While in the other PH group, their drinking water was replaced with antibiotic water. On day 35 to day 40, anxiety- and depression-like behaviors (sucrose consumption, open field test, forced swim test, and tail-suspension test) were conducted. Results showed pair-housed mice had better performance on anxiety- and depression-like behaviors than the PSD mice, and the richness and diversity of intestinal flora were also improved. However, drinking antibiotic water reversed the effects of pair-housing. Furthermore, pair-housing had an obvious improvement in gut barrier disorder and inflammation caused by PSD. Particularly, they showed significant decreases in CD8 lymphocytes and mRNA levels of pro-inflammatory cytokines (TNF-a, IL-1ß and IL-6), while IL-10 mRNA was upregulated. In addition, pair-housing significantly reduced activated microglia and increased Nissl's body in the hippocampus of PSD mice. However, all these improvements were worse in the pair-housed mice administrated with antibiotic water. We conclude that pair-housing significantly improves PSD in association with enhanced functions of microbiota-gut-brain axis, and homeostasis of gut microbiota is indispensable for the protective effect of pair-housing on PSD.


Subject(s)
Depression , Gastrointestinal Microbiome , Animals , Gastrointestinal Microbiome/physiology , Mice , Depression/etiology , Depression/microbiology , Male , Stroke/complications , Stroke/microbiology , Stroke/psychology , Brain-Gut Axis/physiology , Mice, Inbred C57BL , Housing, Animal , Infarction, Middle Cerebral Artery/complications , Infarction, Middle Cerebral Artery/psychology
20.
J Neuroimmunol ; 392: 578374, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38797060

ABSTRACT

We aimed to investigate ampicillin (AMP) mechanisms in microbiota-gut-brain axis. We evaluated its effect on two gut and brain regions and behavioral performances. We administred AMP (1 g/l) to BALB/c mice for 21 days. Then, we analyzed body weigth change, stool consistency scoring, gut length, intestinal microbiota composition, nitric oxide synthase 2 (NOS2) expression and tissue integrity. We subsequently evaluated NOS2, GFAP, CD68 and NFL cerebral expression and spatial memory.Interestingly, our data showed gut microbiota disruption, NOS2 upregulation and tissue damage, associated to cerebral NOS2, GFAP, CD68 and NFL over-expression and behavioral alteration. Antiobiotic therapy should be prescribed with great caution.


Subject(s)
Ampicillin , Brain-Gut Axis , Dysbiosis , Gastrointestinal Microbiome , Mice, Inbred BALB C , Nitric Oxide Synthase Type II , Animals , Mice , Ampicillin/pharmacology , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/physiology , Dysbiosis/chemically induced , Nitric Oxide Synthase Type II/metabolism , Male , Brain-Gut Axis/physiology , Brain-Gut Axis/drug effects , Neuroinflammatory Diseases/metabolism , Anti-Bacterial Agents/pharmacology , Spatial Memory/drug effects , Spatial Memory/physiology , Disease Models, Animal , Neurodegenerative Diseases/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL
...