Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Physiol Rev ; 99(4): 1877-2013, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31460832

ABSTRACT

The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson's disease, and Alzheimer's disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.


Subject(s)
Bacteria/metabolism , Brain Diseases/microbiology , Brain/microbiology , Gastrointestinal Microbiome , Intestines/microbiology , Age Factors , Aging , Animals , Bacteria/immunology , Bacteria/pathogenicity , Behavior , Brain/immunology , Brain/metabolism , Brain/physiopathology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Brain Diseases/psychology , Dysbiosis , Enteric Nervous System/metabolism , Enteric Nervous System/microbiology , Enteric Nervous System/physiopathology , Host-Pathogen Interactions , Humans , Intestines/immunology , Neuroimmunomodulation , Neuronal Plasticity , Risk Factors
2.
Pharmacol Rev ; 71(2): 198-224, 2019 04.
Article in English | MEDLINE | ID: mdl-30890566

ABSTRACT

The microbiome plays a key role in health and disease, and there has been considerable interest in therapeutic targeting of the microbiome as well as mining this rich resource in drug discovery efforts. However, a growing body of evidence suggests that the gut microbiota can itself influence the actions of a range of xenobiotics, in both beneficial and potentially harmful ways. Traditionally, clinical studies evaluating the pharmacokinetics of new drugs have mostly ignored the important direct and indirect effects of the gut microbiome on drug metabolism and efficacy. Despite some important observations from xenobiotic metabolism in general, there is only an incomplete understanding of the scope of influence of the microbiome specifically on drug metabolism and absorption, and how this might influence systemic concentrations of parent compounds and toxic metabolites. The significance of both microbial metabolism of xenobiotics and the impact of the gut microbiome on host hepatic enzyme systems is nonetheless gaining traction and presents a further challenge in drug discovery efforts, with implications for improving treatment outcomes or counteracting adverse drug reactions. Microbial factors must now be considered when determining drug pharmacokinetics and the impact that an evolving and dynamic microbiome could have in this regard. In this review, we aim to integrate the contribution of the gut microbiome in health and disease to xenobiotic metabolism focusing on therapeutic interventions, pharmacological drug action, and chemical biotransformations that collectively will have implications for the future practice of precision medicine.


Subject(s)
Gastrointestinal Microbiome/physiology , Microbiota , Xenobiotics/metabolism , Animals , Drug Discovery/methods , Drug-Related Side Effects and Adverse Reactions/epidemiology , Humans , Precision Medicine/methods , Xenobiotics/adverse effects , Xenobiotics/pharmacology
3.
Mol Psychiatry ; 25(10): 2567-2583, 2020 10.
Article in English | MEDLINE | ID: mdl-31092898

ABSTRACT

Male middle age is a transitional period where many physiological and psychological changes occur leading to cognitive and behavioural alterations, and a deterioration of brain function. However, the mechanisms underpinning such changes are unclear. The gut microbiome has been implicated as a key mediator in the communication between the gut and the brain, and in the regulation of brain homeostasis, including brain immune cell function. Thus, we tested whether targeting the gut microbiome by prebiotic supplementation may alter microglia activation and brain function in ageing. Male young adult (8 weeks) and middle-aged (10 months) C57BL/6 mice received diet enriched with a prebiotic (10% oligofructose-enriched inulin) or control chow for 14 weeks. Prebiotic supplementation differentially altered the gut microbiota profile in young and middle-aged mice with changes correlating with faecal metabolites. Functionally, this translated into a reversal of stress-induced immune priming in middle-aged mice. In addition, a reduction in ageing-induced infiltration of Ly-6Chi monocytes into the brain coupled with a reversal in ageing-related increases in a subset of activated microglia (Ly-6C+) was observed. Taken together, these data highlight a potential pathway by which targeting the gut microbiome with prebiotics can modulate the peripheral immune response and alter neuroinflammation in middle age. Our data highlight a novel strategy for the amelioration of age-related neuroinflammatory pathologies and brain function.


Subject(s)
Aging/immunology , Brain/immunology , Gastrointestinal Microbiome/physiology , Prebiotics , Animals , Feces/chemistry , Feces/microbiology , Male , Mice , Mice, Inbred C57BL , Microglia/immunology
4.
Nutr Neurosci ; 24(12): 951-962, 2021 Dec.
Article in English | MEDLINE | ID: mdl-31814540

ABSTRACT

Introduction: Phytoestrogens are non-steroidal estrogen analogues and are found primarily in soy products. They have received increasing attention as dietary supplements for estrogen deficiency and as modulators of endogenous estrogen functions, including cognition and emotion. In addition to modifying the levels of circulating sex hormones, phytoestrogens also exert direct effects on estrogen and androgen receptors in the brain and thus effectively modulate the neural circuit functions.Objective: The aim of this study was to investigate the long-term effects of low phytoestrogen intake (∼6 weeks) on the hippocampal plasticity and hippocampus-dependent memory formation in the adult C57BL/6 male mice.Methods and Results: In comparison to mice on a diet with normal phytoestrogen content, mice on low phytoestrogen diet showed a significant reduction in the phosphorylation of NR2B subunit, a molecular correlate of plasticity in the Schaffer collateral-CA1 synapse. We observed a profound decrease in long-term potentiation (LTP) in the ventral hippocampus, whereas no effect on plasticity was evident in its dorsal portion. Furthermore, we demonstrated that acute perfusion of slices with an estrogen analogue equol, an isoflovane metabolized from daidzein produced by the bacterial flora in the gut, was able to rescue the observed LTP deficit. Examining potential behavioral correlates of the plasticity attenuation, we found that mice on phytoestrogen-free diet display decreased contextual fear memory at remote but not at recent time points after training.Conclusions: Our data suggests that nutritional phytoestrogens have profound effects on the plasticity in the ventral hippocampus and ventral hippocampus-dependent memory.


Subject(s)
Diet , Hippocampus/physiology , Memory/physiology , Neuronal Plasticity/physiology , Phytoestrogens/administration & dosage , Animals , Behavior, Animal , Equol/pharmacology , Fear/physiology , Hippocampus/drug effects , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , Male , Memory/drug effects , Mice , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Phosphorylation/physiology , Receptors, N-Methyl-D-Aspartate/physiology , Synapses/physiology
5.
Front Neuroendocrinol ; 51: 80-101, 2018 10.
Article in English | MEDLINE | ID: mdl-29753796

ABSTRACT

The human gut harbours trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. These intestinal microbes are also key components of the gut-brain axis, the bidirectional communication pathway between the gut and the central nervous system (CNS). In addition, the CNS is closely interconnected with the endocrine system to regulate many physiological processes. An expanding body of evidence is supporting the notion that gut microbiota modifications and/or manipulations may also play a crucial role in the manifestation of specific behavioural responses regulated by neuroendocrine pathways. In this review, we will focus on how the intestinal microorganisms interact with elements of the host neuroendocrine system to modify behaviours relevant to stress, eating behaviour, sexual behaviour, social behaviour, cognition and addiction.


Subject(s)
Behavior, Addictive , Brain/physiology , Feeding Behavior/physiology , Gastrointestinal Microbiome/physiology , Learning/physiology , Neurosecretory Systems/physiology , Sexual Behavior/physiology , Social Behavior , Stress, Psychological , Animals , Behavior, Addictive/metabolism , Behavior, Addictive/physiopathology , Brain/metabolism , Humans , Neurosecretory Systems/metabolism , Stress, Psychological/metabolism , Stress, Psychological/physiopathology
6.
Can J Psychiatry ; 64(11): 747-760, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31530002

ABSTRACT

The gut microbiome as a potential therapeutic target for mental illness is a hot topic in psychiatry. Trillions of bacteria reside in the human gut and have been shown to play a crucial role in gut-brain communication through an influence on neural, immune, and endocrine pathways. Patients with various psychiatric disorders including depression, bipolar disorder, schizophrenia, and autism spectrum disorder have been shown to have significant differences in the composition of their gut microbiome. Enhancing beneficial bacteria in the gut, for example, through the use of probiotics, prebiotics, or dietary change, has the potential to improve mood and reduce anxiety in both healthy people and patient groups. Much attention is being given to this subject in the general media, and patients are becoming increasingly interested in the potential to treat mental illness with microbiome-based therapies. It is imperative that those working with people with mental illness are aware of the rationale and current evidence base for such treatment strategies. In this review, we provide an overview of the gut microbiome, what it is, and what it does in relation to gut-brain communication and psychological function. We describe the fundamental principles and basic techniques used in microbiome-gut-brain axis research in an accessible way for a clinician audience. We summarize the current evidence in relation to microbiome-based strategies for various psychiatric disorders and provide some practical advice that can be given to patients seeking to try a probiotic for mental health benefit.


Subject(s)
Gastrointestinal Microbiome , Mental Disorders/microbiology , Mental Disorders/therapy , Probiotics/therapeutic use , Humans
7.
Arch Microbiol ; 192(1): 69-77, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20020102

ABSTRACT

A Tn5-induced mutant of Mesorhizobium ciceri, TL28, requiring the amino acid lysine for growth on minimal medium was isolated and characterized. The Tn5 insertion in the mutant strain TL28 was located on a 6.8-kb EcoRI fragment of the chromosomal DNA. Complementation analysis with cloned DNA indicated that 1.269 kb of DNA of the 6.8-kb EcoRI fragment restored the wild-type phenotype of the lysine-requiring mutant. This region was further characterized by DNA sequence analysis and was shown to contain a coding sequence homologous to lysA gene of different bacteria. The lys (-) mutant TL28 was unable to elicit development of effective nodules on the roots of Cicer arietinum L. There was no detectable level of lysine in the root exudates of chickpea. However, addition of lysine to the plant growth medium restored the ability of the mutant to produce effective nodules with nitrogen fixation ability on the roots of C. arietinum.


Subject(s)
Alphaproteobacteria/genetics , Carboxy-Lyases/genetics , Genes, Bacterial , Symbiosis/genetics , Alphaproteobacteria/enzymology , Alphaproteobacteria/physiology , Amino Acid Sequence , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cicer/microbiology , Cloning, Molecular , DNA Transposable Elements , DNA, Bacterial/genetics , DNA, Bacterial/metabolism , Dicarboxylic Acid Transporters/genetics , Dicarboxylic Acid Transporters/metabolism , Gene Expression Regulation, Bacterial , Genome, Bacterial , Molecular Sequence Data , Multigene Family , Mutation , Nitrogen Fixation/genetics , Phenotype , Plant Roots/genetics , Plant Roots/metabolism , Plant Roots/microbiology , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Root Nodules, Plant/genetics , Root Nodules, Plant/metabolism
8.
Transl Res ; 179: 223-244, 2017 01.
Article in English | MEDLINE | ID: mdl-27832936

ABSTRACT

The microbial population residing within the human gut represents one of the most densely populated microbial niche in the human body with growing evidence showing it playing a key role in the regulation of behavior and brain function. The bidirectional communication between the gut microbiota and the brain, the microbiota-gut-brain axis, occurs through various pathways including the vagus nerve, the immune system, neuroendocrine pathways, and bacteria-derived metabolites. This axis has been shown to influence neurotransmission and the behavior that are often associated with neuropsychiatric conditions. Therefore, research targeting the modulation of this gut microbiota as a novel therapy for the treatment of various neuropsychiatric conditions is gaining interest. Numerous factors have been highlighted to influence gut microbiota composition, including genetics, health status, mode of birth, and environment. However, it is diet composition and nutritional status that has repeatedly been shown to be one of the most critical modifiable factors regulating the gut microbiota at different time points across the lifespan and under various health conditions. Thus the microbiota is poised to play a key role in nutritional interventions for maintaining brain health.


Subject(s)
Brain/metabolism , Diet , Gastrointestinal Tract/metabolism , Microbiota , Neuropsychiatry , Age Factors , Gastrointestinal Microbiome , Gastrointestinal Tract/microbiology , Humans
9.
CNS Drugs ; 30(11): 1019-1041, 2016 11.
Article in English | MEDLINE | ID: mdl-27417321

ABSTRACT

The role of the gut microbiota in health and disease is becoming increasingly recognized. The microbiota-gut-brain axis is a bi-directional pathway between the brain and the gastrointestinal system. The bacterial commensals in our gut can signal to the brain through a variety of mechanisms, which are slowly being resolved. These include the vagus nerve, immune mediators and microbial metabolites, which influence central processes such as neurotransmission and behaviour. Dysregulation in the composition of the gut microbiota has been identified in several neuropsychiatric disorders, such as autism, schizophrenia and depression. Moreover, preclinical studies suggest that they may be the driving force behind the behavioural abnormalities observed in these conditions. Understanding how bacterial commensals are involved in regulating brain function may lead to novel strategies for development of microbiota-based therapies for these neuropsychiatric disorders.


Subject(s)
Brain/microbiology , Brain/physiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/microbiology , Microbiota/physiology , Animals , Humans , Mental Disorders/microbiology , Mental Disorders/psychology , Neuropsychiatry/methods
SELECTION OF CITATIONS
SEARCH DETAIL