Bidirectional communication between the brain and gut microbiota in Shudderer, a Drosophila Nav channel mutant

2018 ◽  
Author(s):  
Patrick Arthur Lansdon
2020 ◽  
Vol 57 (12) ◽  
pp. 5026-5043 ◽  
Author(s):  
Shan Liu ◽  
Jiguo Gao ◽  
Mingqin Zhu ◽  
Kangding Liu ◽  
Hong-Liang Zhang

Abstract Understanding how gut flora influences gut-brain communications has been the subject of significant research over the past decade. The broadening of the term “microbiota-gut-brain axis” from “gut-brain axis” underscores a bidirectional communication system between the gut and the brain. The microbiota-gut-brain axis involves metabolic, endocrine, neural, and immune pathways which are crucial for the maintenance of brain homeostasis. Alterations in the composition of gut microbiota are associated with multiple neuropsychiatric disorders. Although a causal relationship between gut dysbiosis and neural dysfunction remains elusive, emerging evidence indicates that gut dysbiosis may promote amyloid-beta aggregation, neuroinflammation, oxidative stress, and insulin resistance in the pathogenesis of Alzheimer’s disease (AD). Illustration of the mechanisms underlying the regulation by gut microbiota may pave the way for developing novel therapeutic strategies for AD. In this narrative review, we provide an overview of gut microbiota and their dysregulation in the pathogenesis of AD. Novel insights into the modification of gut microbiota composition as a preventive or therapeutic approach for AD are highlighted.


Biomolecules ◽  
2021 ◽  
Vol 11 (7) ◽  
pp. 1000
Author(s):  
Agata Chudzik ◽  
Anna Orzyłowska ◽  
Radosław Rola ◽  
Greg J. Stanisz

The brain–gut–microbiome axis is a bidirectional communication pathway between the gut microbiota and the central nervous system. The growing interest in the gut microbiota and mechanisms of its interaction with the brain has contributed to the considerable attention given to the potential use of probiotics, prebiotics and postbiotics in the prevention and treatment of depressive disorders. This review discusses the up-to-date findings in preclinical and clinical trials regarding the use of pro-, pre- and postbiotics in depressive disorders. Studies in rodent models of depression show that some of them inhibit inflammation, decrease corticosterone level and change the level of neurometabolites, which consequently lead to mitigation of the symptoms of depression. Moreover, certain clinical studies have indicated improvement in mood as well as changes in biochemical parameters in patients suffering from depressive disorders.


Nutrients ◽  
2018 ◽  
Vol 10 (11) ◽  
pp. 1765 ◽  
Author(s):  
Vo Giau ◽  
Si Wu ◽  
Angelo Jamerlan ◽  
Seong An ◽  
SangYun Kim ◽  
...  

The bidirectional communication between the central nervous system (CNS) and the gut microbiota plays a pivotal role in human health. Increasing numbers of studies suggest that the gut microbiota can influence the brain and behavior of patients. Various metabolites secreted by the gut microbiota can affect the cognitive ability of patients diagnosed with neurodegenerative diseases. Nearly one in every ten Korean senior citizens suffers from Alzheimer’s disease (AD), the most common form of dementia. This review highlights the impact of metabolites from the gut microbiota on communication pathways between the brain and gut, as well as the neuroinflammatory roles they may have in AD patients. The objectives of this review are as follows: (1) to examine the role of the intestinal microbiota in homeostatic communication between the gut microbiota and the brain, termed the microbiota–gut–brain (MGB) axis; (2) to determine the underlying mechanisms of signal dysfunction; and (3) to assess the impact of signal dysfunction induced by the microbiota on AD. This review will aid in understanding the microbiota of elderly people and the neuroinflammatory roles they may have in AD.


2018 ◽  
Vol 15 (13) ◽  
pp. 1179-1190 ◽  
Author(s):  
Vilma M. Junges ◽  
Vera E. Closs ◽  
Guilherme M. Nogueira ◽  
Maria G.V. Gottlieb

The role of diet and gut microbiota in the pathophysiology of neurodegenerative diseases, such as Alzheimer's, has recently come under intense investigation. Studies suggest that human gut microbiota may contribute to the modulation of several neurochemical and neurometabolic pathways, through complex systems that interact and interconnect with the central nervous system. The brain and intestine form a bidirectional communication axis, or vice versa, they form an axis through bi-directional communication between endocrine and complex immune systems, involving neurotransmitters and hormones. Above all, studies suggest that dysbiotic and poorly diversified microbiota may interfere with the synthesis and secretion of neurotrophic factors, such as brain-derived neurotrophic factor, gammaaminobutyric acid and N-methyl D-Aspartate receptors, widely associated with cognitive decline and dementia. In this context, the present article provides a review of the literature on the role of the gutbrain axis in Alzheimer's disease.


2021 ◽  
Vol 15 ◽  
Author(s):  
Amaryllis E. Hill ◽  
Richard Wade-Martins ◽  
Philip W. J. Burnet

Microbiota have increasingly become implicated in predisposition to human diseases, including neurodegenerative disorders such as Parkinson’s disease (PD). Traditionally, a central nervous system (CNS)-centric approach to understanding PD has predominated; however, an association of the gut with PD has existed since Parkinson himself reported the disease. The gut–brain axis refers to the bidirectional communication between the gastrointestinal tract (GIT) and the brain. Gut microbiota dysbiosis, reported in PD patients, may extend this to a microbiota–gut–brain axis. To date, mainly the bacteriome has been investigated. The change in abundance of bacterial products which accompanies dysbiosis is hypothesised to influence PD pathophysiology via multiple mechanisms which broadly centre on inflammation, a cause of alpha-synuclein (a-syn) misfolding. Two main routes are hypothesised by which gut microbiota can influence PD pathophysiology, the neural and humoral routes. The neural route involves a-syn misfolding peripherally in the enteric nerves which can then be transported to the brain via the vagus nerve. The humoral route involves transportation of bacterial products and proinflammatory cytokines from the gut via the circulation which can cause central a-syn misfolding by inducing neuroinflammation. This article will assess whether the current literature supports gut bacteria influencing PD pathophysiology via both routes.


Cells ◽  
2021 ◽  
Vol 10 (8) ◽  
pp. 2084
Author(s):  
Julia Murciano-Brea ◽  
Martin Garcia-Montes ◽  
Stefano Geuna ◽  
Celia Herrera-Rincon

The accumulating evidence linking bacteria in the gut and neurons in the brain (the microbiota–gut–brain axis) has led to a paradigm shift in the neurosciences. Understanding the neurobiological mechanisms supporting the relevance of actions mediated by the gut microbiota for brain physiology and neuronal functioning is a key research area. In this review, we discuss the literature showing how the microbiota is emerging as a key regulator of the brain’s function and behavior, as increasing amounts of evidence on the importance of the bidirectional communication between the intestinal bacteria and the brain have accumulated. Based on recent discoveries, we suggest that the interaction between diet and the gut microbiota, which might ultimately affect the brain, represents an unprecedented stimulus for conducting new research that links food and mood. We also review the limited work in the clinical arena to date, and we propose novel approaches for deciphering the gut microbiota–brain axis and, eventually, for manipulating this relationship to boost mental wellness.


2021 ◽  
Vol 22 (18) ◽  
pp. 10052
Author(s):  
Lucía N. Peralta-Marzal ◽  
Naika Prince ◽  
Djordje Bajic ◽  
Léa Roussin ◽  
Laurent Naudon ◽  
...  

Autism Spectrum Disorder (ASD) is a set of neurodevelopmental disorders characterised by behavioural impairment and deficiencies in social interaction and communication. A recent study estimated that 1 in 89 children have developed some form of ASD in European countries. Moreover, there is no specific treatment and since ASD is not a single clinical entity, the identification of molecular biomarkers for diagnosis remains challenging. Besides behavioural deficiencies, individuals with ASD often develop comorbid medical conditions including intestinal problems, which may reflect aberrations in the bidirectional communication between the brain and the gut. The impact of faecal microbial composition in brain development and behavioural functions has been repeatedly linked to ASD, as well as changes in the metabolic profile of individuals affected by ASD. Since metabolism is one of the major drivers of microbiome–host interactions, this review aims to report emerging literature showing shifts in gut microbiota metabolic function in ASD. Additionally, we discuss how these changes may be involved in and/or perpetuate ASD pathology. These valuable insights can help us to better comprehend ASD pathogenesis and may provide relevant biomarkers for improving diagnosis and identifying new therapeutic targets.


2021 ◽  
Vol 8 ◽  
Author(s):  
Jie-Yu Chuang

Microbiota inhabit nearly every part of our body with the gut microbiota representing the greatest density and absolute abundance. The gut-microbiota-brain axis facilitates bidirectional communication between gut microbiota and the brain. For instance, romantic relationship not only brings joy, it is also associated with increased gut microbiota diversity and health benefits whereas reduced microbiota diversity is related to obesity, cardiac disease, type 2 diabetes, and inflammatory disorders. Research has shown that dietary fibers may increase microbiota diversity and exert antidepressant effect. Among a plethora of life stressors, romantic relationship dissolution is a relatively common and painful experience that people encounter from time to time. Depressed mood, social isolation and poor intake are all associated with romantic relationship dissolution. In this article, it is hypothesized that romantic relationship dissolution is accompanied by decreased gut microbiota diversity which could be corrected with the ingestion of dietary fibers with an additional antidepressant benefit.


Molecules ◽  
2020 ◽  
Vol 25 (10) ◽  
pp. 2444
Author(s):  
Kenjiro Ono ◽  
Mayumi Tsuji ◽  
Tritia R. Yamasaki ◽  
Giulio M. Pasinetti

The aggregation and deposition of α-synuclein (αS) are major pathologic features of Parkinson’s disease, dementia with Lewy bodies, and other α-synucleinopathies. The propagation of αS pathology in the brain plays a key role in the onset and progression of clinical phenotypes. Thus, there is increasing interest in developing strategies that attenuate αS aggregation and propagation. Based on cumulative evidence that αS oligomers are neurotoxic and critical species in the pathogenesis of α-synucleinopathies, we and other groups reported that phenolic compounds inhibit αS aggregation including oligomerization, thereby ameliorating αS oligomer-induced cellular and synaptic toxicities. Heterogeneity in gut microbiota may influence the efficacy of dietary polyphenol metabolism. Our recent studies on the brain-penetrating polyphenolic acids 3-hydroxybenzoic acid (3-HBA), 3,4-dihydroxybenzoic acid (3,4-diHBA), and 3-hydroxyphenylacetic acid (3-HPPA), which are derived from gut microbiota-based metabolism of dietary polyphenols, demonstrated an in vitro ability to inhibit αS oligomerization and mediate aggregated αS-induced neurotoxicity. Additionally, 3-HPPA, 3,4-diHBA, 3-HBA, and 4-hydroxybenzoic acid significantly attenuated intracellular αS seeding aggregation in a cell-based system. This review focuses on recent research developments regarding neuroprotective properties, especially anti-αS aggregation effects, of phenolic compounds and their metabolites by the gut microbiome, including our findings in the pathogenesis of α-synucleinopathies.


2020 ◽  
Vol 6 (12) ◽  
pp. 124-154
Author(s):  
S. Bulgakova ◽  
N. Romanchuk ◽  
O. Pomazanova

The new competencies of psychoneuroimmunoendocrinology and psychoneuroimmunology play a strategic role in interdisciplinary science and interdisciplinary planning and decision-making. The introduction of multi-vector neurotechnologies of artificial intelligence and the principles of digital health care will contribute to the development of modern neuroscience and neuromarketing. The availability of innovative technologies, such as next-generation sequencing and correlated bioinformatics tools, allows deeper investigation of the cross-network relationships between the microbiota and human immune responses. Immune homeostasis is the balance between immunological tolerance and inflammatory immune responses — a key feature in the outcome of health or disease. A healthy microbiota is the qualitative and quantitative ratio of diverse microbes of individual organs and systems, maintaining the biochemical, metabolic and immune equilibrium of the macroorganism necessary to preserve human health. Functional foods, healthy biomicrobiota, healthy lifestyle and controlled protective environmental effects, artificial intelligence and electromagnetic information load/overload are responsible for the work of the human immune system and its ability to respond to pandemic attacks in a timely manner. Obesity continues to be one of the main problems of modern health care due to its high prevalence and polymorbidity. In addition to cardiometabolic diseases, lesions of the musculoskeletal system, obese individuals show impaired cognitive functions, have a high risk of developing depression and anxiety. The gut microbiota mediates between environmental influences (food, lifestyle) and the physiology of the host, and its change may partially explain the cross-link between the above pathologies. It is known that Western eating patterns are the main cause of the obesity epidemic, which also contributes to dysbiotic drift of the gut microbiota, which in turn contributes to the development of complications associated with obesity. Experimental studies in animal models and, to a lesser extent in humans, show that microbiota is associated with obesity and may contribute to the endocrine, neurochemical and development of systemic inflammation underlying obesity itself and related diseases. Nevertheless, a number of questions remain at present. Modeling the microbiota-gut-brain axis, provides the brain with information from the gut not only through the nervous system but also through a continuous stream of microbial, endocrine, metabolic and immune messages. The communication network provides important keys to understanding how obesity and diabetes can affect the brain by provoking neuropsychiatric diseases. The literature review is devoted to the analysis of data on the relationship of the gut-brain axis, obesity and cognitive functions, immune homeostasis and new competencies: psychoneuroimmunology and psychoneuroimmunoendocrinology.


Sign in / Sign up

Export Citation Format

Share Document