Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Publication year range
1.
Brain Behav Immun ; 122: 510-526, 2024 Aug 25.
Article in English | MEDLINE | ID: mdl-39191350

ABSTRACT

The intricacy and multifaceted nature of Alzheimer's disease (AD) necessitate therapies that target multiple aspects of the disease. Mesenchymal stromal cells (MSCs) emerge as potential agents to mitigate AD symptoms; however, whether their therapeutic efficacy involves modulation of gut microbiota and the microbiome-gut-brain axis (MGBA) remains unexplored. In this study, we evaluated the effects of three distinct MSCs types-derived from the umbilical cord (UCMSC), dental pulp (SHED), and adipose tissue (ADSC)-in an APP/PS1 mouse model of AD. In comparison to saline control, MSCs administration resulted in a significant reduction of behavioral disturbances, amyloid plaques, and phosphorylated tau in the hippocampus and frontal cortex, accompanied by an increase in neuronal count and Nissl body density across AD-afflicted brain regions. Through 16S rRNA gene sequencing, we identified partial restoration of gut microbial balance in AD mice post-MSCs treatment, evidenced by the elevation of neuroprotective Akkermansia and reduction of the AD-associated Sphingomonas. To examine whether gut microbiota involved in MSCs efficacy in treating AD, SHED with better anti-inflammatory and gut microbiota recovery effects among three MSCs, and another AD model 5 × FAD mice with earlier and more pathological proteins in brain than APP/PS1, were selected for further studies. Antibiotic-mediated gut microbial inactivation attenuated MSCs efficacy in 5 × FAD mice, implicating the involvement of gut microbiota in the therapeutic mechanism. Functional analysis of altered gut microbiota and targeted bile acid metabolism profiling revealed a significant enhancement in bile acid variety following MSCs therapy. A chief bile acid constituent, taurocholic acid (TCA), was orally administered to AD mice and similarly abated AD symptoms. Nonetheless, the disruption of intestinal neuronal integrity with enterotoxin abrogated the ameliorative impact of both MSCs and TCA treatments. Collectively, our findings substantiate that MSCs confer therapeutic benefits in AD within a paradigm that primarily involves regulation of gut microbiota and their metabolites through the MGBA.

2.
Biol Res ; 57(1): 23, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38705984

ABSTRACT

Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.


Subject(s)
Anxiety , Brain-Gut Axis , Diet, High-Fat , Gastrointestinal Microbiome , Animals , Male , Diet, High-Fat/adverse effects , Gastrointestinal Microbiome/physiology , Anxiety/microbiology , Brain-Gut Axis/physiology , Rats , Rats, Sprague-Dawley , Obesity/microbiology , Obesity/psychology , Obesity/metabolism , Signal Transduction/physiology , Behavior, Animal/physiology
3.
Expert Rev Proteomics ; 20(12): 397-418, 2023.
Article in English | MEDLINE | ID: mdl-37934939

ABSTRACT

INTRODUCTION: An increasing number of studies indicate that the microbiota-gut-brain axis is an important pathway involved in the onset and progression of depression. The responses of the organism (or its microorganisms) to external cues cannot be separated from a key intermediate element: their metabolites. AREAS COVERED: In recent years, with the rapid development of metabolomics, an increasing amount of metabolites has been detected and studied, especially the gut metabolites. Nevertheless, the increasing amount of metabolites described has not been reflected in a better understanding of their functions and metabolic pathways. Moreover, our knowledge of the biological interactions among metabolites is also incomplete, which limits further studies on the connections between the microbial-entero-brain axis and depression. EXPERT OPINION: This paper summarizes the current knowledge on depression-related metabolites and their involvement in the onset and progression of this disease. More importantly, this paper summarized metabolites from the intestine, and defined them as enterogenic metabolites, to further clarify the function of intestinal metabolites and their biochemical cross-talk, providing theoretical support and new research directions for the prevention and treatment of depression.


Subject(s)
Gastrointestinal Microbiome , Humans , Depression , Metabolomics , Metabolome , Brain
4.
Paediatr Perinat Epidemiol ; 37(6): 516-526, 2023 08.
Article in English | MEDLINE | ID: mdl-36978215

ABSTRACT

BACKGROUND: Prenatal antibiotic exposure induces changes in the maternal microbiome, which could influence the development of the infant's microbiome-gut-brain axis. OBJECTIVES: We assessed whether prenatal antibiotic exposure is associated with an increased risk of autism spectrum disorder (ASD) in offspring born at term. METHODS: This population-based retrospective cohort study included everyone who delivered a live singleton-term infant in British Columbia, Canada between April 2000 and December 2014. Exposure was defined as filling antibiotic prescriptions during pregnancy. The outcome was an ASD diagnosis from the British Columbia Autism Assessment Network, with a follow-up to December 2016. To examine the association among pregnant individuals treated for the same indication, we studied a sub-cohort diagnosed with urinary tract infections. Cox proportional hazards models were used to estimate unadjusted and adjusted hazard ratios (HR). The analysis was stratified by sex, trimester, cumulative duration of exposure, class of antibiotic, and mode of delivery. We ran a conditional logistic regression of discordant sibling pairs to control for unmeasured environmental and genetic confounding. RESULTS: Of the 569,953 children included in the cohort, 8729 were diagnosed with ASD (1.5%) and 169,922 were exposed to prenatal antibiotics (29.8%). Prenatal antibiotic exposure was associated with an increased risk of ASD (HR 1.10, 95% confidence interval [CI] 1.05, 1.15), particularly for exposure during the first and second trimesters (HR 1.11, 95% CI 1.04, 1.18 and HR 1.09, 95% CI 1.03, 1.16, respectively), and exposure lasting ≥15 days (HR 1.13, 95% CI 1.04, 1.23). No sex differences were observed. The association was attenuated in the sibling analysis (adjusted odds ratio 1.04, 95% CI 0.92, 1.17). CONCLUSIONS: Prenatal antibiotic exposure was associated with a small increase in the risk of ASD in offspring. Given the possibility of residual confounding, these results should not influence clinical decisions regarding antibiotic use during pregnancy.


Subject(s)
Autism Spectrum Disorder , Child , Female , Humans , Infant , Pregnancy , Anti-Bacterial Agents/adverse effects , Autism Spectrum Disorder/chemically induced , Autism Spectrum Disorder/epidemiology , Cohort Studies , Retrospective Studies , Term Birth , Prenatal Exposure Delayed Effects
5.
BMC Vet Res ; 19(1): 57, 2023 Mar 03.
Article in English | MEDLINE | ID: mdl-36864510

ABSTRACT

BACKGROUND: Epilepsy is the most common chronic neurological disease in dogs. More than two-thirds of these patients suffer from associated behavioural comorbidities. The latter could have their origin in partially overlapping pathomechanisms, with the intestinal microbiome as a potential key link between them. The current arsenal of drugs for epilepsy management remains limited. Most canine patients continue to have seizures despite treatment and the occurrence of comorbidities is not sufficiently addressed, limiting quality of life of affected dogs and owners. Therefore, novel additional epilepsy management options are urgently needed. The microbiome-gut-brain axis may serve as a new target for the development of innovative multimodal therapeutic approaches to overcome current shortcomings in epilepsy management. METHODS: A six-month prospective, randomised, double-blinded, placebo-controlled, crossover, dietary trial was designed to investigate the potential of the psychobiotic Bifidobacterium longum on behavioural comorbidities in canine epilepsy. Seizure semiology will be evaluated as a secondary outcome measure. Thirty-four privately owned dogs are planned to be included in the ongoing study meeting the following inclusion criteria: Dogs displaying increased anxiety/fear behaviour since the start of the idiopathic epilepsy. Tier II confidence level of the International Veterinary Epilepsy Task Force for the diagnosis of idiopathic epilepsy, with a maximum seizure interval of 3 month and a minimum of three generalised seizures within that period and chronically treated with at least one antiseizure drug without improvement in seizure frequency Each dog will receive the allocated supplement (probiotic vs. placebo) alongside its normal diet for a 3-month period. After a three-week wash out period, the second phase starts by administering the respective other supplement for another 3 months. DISCUSSION: The current study considers modern high-quality standards for epilepsy medication trials. Common biasing effects should be limited to a possible minimum (regression-to-the mean effect, placebo effect, observer effect), ensuring a high validity and accuracy of the acquired results, thus enabling a representative nature of the efficacy of Bifidobacterium longum as add-on supplement for dogs suffering from epilepsy and its comorbidities. This publication should provide a description of the study procedure and data acquisition methods, including prognosed statistical analysis.


Subject(s)
Dog Diseases , Epilepsy , Dogs , Animals , Prospective Studies , Quality of Life , Epilepsy/drug therapy , Epilepsy/veterinary , Seizures/drug therapy , Seizures/veterinary , Diet , Dog Diseases/drug therapy , Clinical Trials, Veterinary as Topic
6.
Ecotoxicol Environ Saf ; 259: 115035, 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37224779

ABSTRACT

Approximately 400 million people work and live in high-altitude areas and suffer from memory dysfunction worldwide. Until now, the role of the intestinal flora in plateau-induced brain damage has rarely been reported. To address this, we investigated the effect of intestinal flora on spatial memory impairment induced by high altitudes based on the microbiome-gut-brain axis theory. C57BL/6 mice were divided into three groups: control, high-altitude (HA), and high-altitude antibiotic treatment (HAA) group. The HA and HAA groups were exposed to a low-pressure oxygen chamber that simulated an altitude of 4000 m above sea level (m. a. s.l.) for 14 days, with the air pressure in the chamber set at 60-65 kPa. The results showed that spatial memory dysfunction induced by the high-altitude environment was aggravated by antibiotic treatment, manifesting as lowered escape latency and hippocampal memory-related proteins (BDNF and PSD-95). 16 S rRNA sequencing showed a remarkable separation of the ileal microbiota among the three groups. Antibiotic treatment exacerbated the reduced richness and diversity of the ileal microbiota in mice in the HA group. Lactobacillaceae were the main target bacteria and were significantly reduced in the HA group, which was exacerbated by antibiotic treatment. Meanwhile, reduced intestinal permeability and ileal immune function in mice exposed high-altitude environment was also aggravated by antibiotic treatment, as indicated by the lowered tight junction proteins and IL-1ß and IFN-γ levels. Furthermore, indicator species analysis and Netshift co-analysis revealed that Lactobacillaceae (ASV11) and Corynebacteriaceae (ASV78, ASV25, and ASV47) play important roles in high-altitude exposure-induced memory dysfunction. Interestingly, ASV78 was negatively correlated with IL-1ß and IFN-γ levels, indicating that ASV78 may be induced by reduced ileal immune function, which mediates high-altitude environment exposure-induced memory dysfunction. This study provides evidence that the intestinal flora is effective in preventing brain dysfunction caused by exposure to high-altitude environments, suggesting a relationship between the microbiome-gut-brain axis and altitude exposure.


Subject(s)
Gastrointestinal Microbiome , Animals , Mice , Brain-Gut Axis , Altitude , Spatial Memory , Mice, Inbred C57BL , Anti-Bacterial Agents/pharmacology
7.
Pharm Biol ; 61(1): 598-609, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37013944

ABSTRACT

CONTEXT: Sinisan (SNS) has been used to treat psychosomatic diseases of the digestive system. But little is known about how SNS affects water immersion restraint stress (WIRS). OBJECTIVE: To study the effects of SNS on colonic tissue injury in the WIRS model. MATERIALS AND METHODS: Forty-eight Kunming (KM) mice were randomized into 6 groups (n = 8): The control and WIRS groups receiving deionized water; the SNS low-dose (SL, 3.12 g/kg/d), SNS middle-dose (SM, 6.24 g/kg/d), SNS high-dose (SH, 12.48 g/kg/d), and diazepam (DZ, 5 mg/kg/d) groups; each with two daily administrations for 5 consecutive days. The 5 treatment groups were subjected to WIRS for 24 h on day 6. The effects of SNS on colon tissue injury caused by WIRS were assessed by changes in colon histology, inflammatory cytokines, brain-gut peptides, and tight junction (TJ) proteins levels. 16S rRNA gene sequencing was used to detect the regulation of the gut microbiota. RESULTS: SNS pretreatment significantly reduced TNF-α (0.75- to 0.81-fold), IL-6 (0.77-fold), and IFN-γ (0.69-fold) levels; and increased TJ proteins levels, such as ZO-1 (4.06- to 5.27-fold), claudin-1 (3.33- to 5.14-fold), and occludin (6.46- to 11.82-fold). However, there was no significant difference between the levels of substance P (SP) and vasoactive intestinal peptide (VIP) in the control and WIRS groups. SNS regulated the composition of gut microbiota in WIRS mice. CONCLUSION: The positive effects of SNS on WIRS could provide a theoretical basis to treat stress-related gastrointestinal disorders.


Subject(s)
Gastrointestinal Microbiome , Mice , Animals , Intestinal Mucosa , Immersion , RNA, Ribosomal, 16S , Colon/pathology , Tight Junction Proteins/metabolism , Water/pharmacology
8.
J Neuroinflammation ; 18(1): 9, 2021 Jan 06.
Article in English | MEDLINE | ID: mdl-33407614

ABSTRACT

BACKGROUND: Both genetic factors and environmental hazards, including environmental noise stress, have been associated with gut microbiome that exacerbates Alzheimer's disease (AD) pathology. However, the role and mechanism of environmental risk factors in early-onset AD (EOAD) pathogenesis remain unclear. METHODS: The molecular pathways underlying EOAD pathophysiology following environmental noise exposure were evaluated using C57BL/6 wild-type (WT) and APP/PS1 Tg mouse models. The composition differences in intestinal microbiota were analyzed by 16S rRNA sequencing and Tax4Fun to predict the metagenome content from sequencing results. An assessment of the flora dysbiosis-triggered dyshomeostasis of oxi-inflamm-barrier and the effects of the CNS end of the gut-brain axis was conducted to explore the underlying pathological mechanisms. RESULTS: Both WT and APP/PS1 mice showed a statistically significant relationship between environmental noise and the taxonomic composition of the corresponding gut microbiome. Bacterial-encoded functional categories in noise-exposed WT and APP/PS1 mice included phospholipid and galactose metabolism, oxidative stress, and cell senescence. These alterations corresponded with imbalanced intestinal oxidation and anti-oxidation systems and low-grade systemic inflammation following noise exposure. Mechanistically, axis-series experiments demonstrated that following noise exposure, intestinal and hippocampal tight junction protein levels reduced, whereas serum levels of inflammatory mediator were elevated. Regarding APP/PS1 overexpression, noise-induced abnormalities in the gut-brain axis may contribute to aggravation of neuropathology in the presymptomatic stage of EOAD mice model. CONCLUSION: Our results demonstrate that noise exposure has deleterious effects on the homeostasis of oxi-inflamm-barrier in the microbiome-gut-brain axis. Therefore, at least in a genetic context, chronic noise may aggravate the progression of EOAD.


Subject(s)
Alzheimer Disease/metabolism , Environmental Exposure/adverse effects , Gastrointestinal Microbiome/physiology , Homeostasis/physiology , Inflammation Mediators/metabolism , Noise/adverse effects , Acoustic Stimulation/adverse effects , Alzheimer Disease/etiology , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Animals , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic
9.
Brain Behav Immun ; 98: 245-250, 2021 11.
Article in English | MEDLINE | ID: mdl-34403735

ABSTRACT

Patients with inflammatory bowel disease (IBD) are particularly susceptible to behavioral diagnoses, and the microbiome has been repeatedly implicated in the pathogenesis of IBD. The intestinal microbiome's ability to affect behavior has become increasingly recognized and studied. The so-called 'psychobiome' has been linked to a plethora of neurological and psychological diagnoses, including autism and Parkinson's disease. Despite the ability of many bacterial species within the human intestinal microbiome to synthesize neurotransmitters, it has never been previously reported that a single bacterial species is sufficient to induce depression. Here, we demonstrate that our mouse model of Crohn's disease (CD)-like ileitis, the SAMP1/YitFc (SAMP1), does not exhibit baseline behavioral abnormalities. By comparison, SAMP6 mice develop depressive-like behavior that is associated with a rise in the GABA-producing bacterial genus Parabacteroides. We finally demonstrate that administration of Parabacteroides distasonis into our SAMP1 mice induces depressive-like behavior. Colonization with P. distasonis was not associated with increased intestinal inflammation or alterations in other measures of behavior. The intestinal environment of CD may be particularly conducive to colonization with P. distasonis and subsequent induction of depressive-like behavior. To our knowledge, this is the first report of a bacterial species specifically inducing depressive-like behavior.


Subject(s)
Crohn Disease , Ileitis , Animals , Bacteroidetes , Disease Models, Animal , Humans , Mice
10.
Pharmacol Res ; 173: 105909, 2021 11.
Article in English | MEDLINE | ID: mdl-34543739

ABSTRACT

Recently, increasing evidence has shown gut microbiota dysbiosis might be implicated in the physiological mechanisms of neuropsychiatric disorders. Altered microbial community composition, diversity and distribution traits have been reported in neuropsychiatric disorders. However, the exact pathways by which the intestinal microbiota contribute to neuropsychiatric disorders remain largely unknown. Given that the onset and progression of neuropsychiatric disorders are characterized with complicated alterations of neuroendocrine and immunology, both of which can be continually affected by gut microbiota via "microbiome-gut-brain axis". Thus, we assess the complicated crosstalk between neuroendocrine and immunological regulation might underlie the mechanisms of gut microbiota associated with neuropsychiatric disorders. In this review, we summarized clinical and preclinical evidence on the role of the gut microbiota in neuropsychiatry disorders, especially in mood disorders and neurodevelopmental disorders. This review may elaborate the potential mechanisms of gut microbiota implicating in neuroendocrine-immune regulation and provide a comprehensive understanding of physiological mechanisms for neuropsychiatric disorders.


Subject(s)
Gastrointestinal Microbiome , Mental Disorders/immunology , Mental Disorders/microbiology , Animals , Brain-Gut Axis , Humans
11.
Support Care Cancer ; 29(2): 605-617, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32918608

ABSTRACT

PURPOSE: The microbiome-gut-brain (MGB) axis provides a dynamic model to understand associations between the gut microbiota and psychoneurological comorbidities. The role of the MGB axis in cancer treatment-related psychoneurological symptoms (PNS) remains unknown. The purpose of this study was to conduct a systematic review of the existing literature to identify the influence of the gut microbiota on cancer and cancer treatment-related PNS and toxicities mediated by the MGB axis. METHODS: We searched the databases of PubMed, Embase, and Web of Science from their earliest records to October 2019. All studies identified in the database searches were screened by title and abstract, followed by a review of the full texts. The Johns Hopkins Nursing Evidence-Based Practice Model was adopted to assess the evidence levels and qualities; the Joanna Briggs Institute critical appraisal tools were used to assess the methodological quality and the possibility of bias for each included study. All the study findings were combined, synthesized, and presented through narrative format. RESULTS: Six studies were included in this systematic review. These studies primarily focused on cancer survivorship while receiving chemotherapy, and they were conducted between 2016 and 2019. The gut microbiome was assessed via fecal samples, which were analyzed using 16S rRNA sequencing approaches. With small-scale studies, the gut microbiota was associated with cancer treatment-related PNS, including fatigue, anxiety, depression, sleep disturbance, cognitive impairment, and chemotherapy-induced peripheral neuropathy. A higher relative abundance of Bacteroides was associated with a higher level of fear of cancer recurrence but a higher relative abundance of Lachnospiraceae.g and Ruminococcus was associated with a lower level in fear of cancer recurrence. Changes in fatigue interference were associated with the frequency of genera Faecalibacterium and Prevotella, and changes in anxiety were associated with the frequency of genera Coprococcus and Bacteroides. CONCLUSIONS: The gut microbiota showed significant associations with cancer treatment-related PNS. Recent work regarding the MGB axis in cancer psychoneurological toxicities focused primarily on individual toxicity and symptoms in cancer survivors with chemotherapy exposure. Associations between the gut microbiota and PNS should be further studied in cancer populations across different ages, cancer types, and treatment modalities.


Subject(s)
Brain/microbiology , Gastrointestinal Microbiome/physiology , Neoplasms/microbiology , Neoplasms/psychology , Animals , Anxiety/etiology , Anxiety/microbiology , Cancer Survivors/psychology , Feces/microbiology , Humans , Neoplasms/pathology , Neoplasms, Second Primary
12.
Biomed Chromatogr ; 35(5): e5058, 2021 May.
Article in English | MEDLINE | ID: mdl-33373060

ABSTRACT

Rhubarb anthraquinone glycosides (RAGs) have been proven to have significant therapeutic effects on ischemic stroke, and this effect may be related to the microbiome-gut-brain axis. In this study, an HPLC-FLD method was established to measure brain-gut neurotransmitters of rats with cerebral ischemia-reperfusion injury (CIRI), to explore whether the mechanism of RAGs against CIRI is related to the microbiome-gut-brain axis. A Shimadzu ODS-3 C18 column was used for chromatographic separation, and 5-hydroxytryptamine (5-HT), 5-hydroxy indole acetic acid (5-HIAA), glutamic acid (Glu), aspartic acid (Asp), and γ-aminobutyric acid (GABA) were determined simultaneously. The results showed that there is an excellent linear relationship (R2 ≥ 0.9990) and a high separation degree in the HPLC-FLD method. Whereas the contents of Asp and Glu in the brain and colon increased (p < 0.05), the contents of 5-HT, 5-HIAA, and GABA in the brain and colon decreased (p < 0.05) after CIRI. RAGs could effectively reduce the contents of Asp and Glu (p < 0.05), and increase the contents of 5-HT, 5-HIAA, and GABA in the brain and colon (p < 0.05). Combined with the previous experimental results, we can speculate that RAGs can regulate intestinal flora disorder caused by CIRI, and then regulate the imbalance between the release and decomposition of neurotransmitters caused by intestinal flora disorder.


Subject(s)
Anthraquinones , Brain Ischemia/drug therapy , Neurotransmitter Agents , Protective Agents , Reperfusion Injury/drug therapy , Animals , Anthraquinones/pharmacology , Brain Chemistry , Colon/chemistry , Male , Neurotransmitter Agents/metabolism , Protective Agents/pharmacology , Rats , Rats, Sprague-Dawley , Rheum/chemistry
13.
Neurobiol Dis ; 135: 104578, 2020 02.
Article in English | MEDLINE | ID: mdl-31454550

ABSTRACT

Depression affects at least 322 million people globally, or approximately 4.4% of the world's population. While the earnestness of researchers and clinicians to understand and treat depression is not waning, the number of individuals suffering from depression continues to increase over and above the rate of global population growth. There is a sincere need for a paradigm shift. Research in the past decade is beginning to take a more holistic approach to understanding depression etiology and treatment, integrating multiple body systems into whole-body conceptualizations of this mental health affliction. Evidence supports the hypothesis that the gut microbiome, or the collective trillions of microbes inhabiting the gastrointestinal tract, is an important factor determining both the risk of development of depression and persistence of depressive symptoms. This review discusses recent advances in both rodent and human research that explore bidirectional communication between the gut microbiome and the immune, endocrine, and central nervous systems implicated in the etiology and pathophysiology of depression. Through interactions with circulating inflammatory markers and hormones, afferent and efferent neural systems, and other, more niche, pathways, the gut microbiome can affect behavior to facilitate the development of depression, exacerbate current symptoms, or contribute to treatment and resilience. While the challenge of depression may be the direst mental health crisis of our age, new discoveries in the gut microbiome, when integrated into a holistic perspective, hold great promise for the future of positive mental health.


Subject(s)
Central Nervous System/microbiology , Depression/microbiology , Gastrointestinal Microbiome/physiology , Gastrointestinal Tract/metabolism , Microbiota/physiology , Central Nervous System/metabolism , Depression/physiopathology , Gastrointestinal Tract/physiopathology , Humans , Mental Disorders/metabolism , Mental Disorders/microbiology
14.
Brain Behav Immun ; 89: 641-655, 2020 10.
Article in English | MEDLINE | ID: mdl-32827699

ABSTRACT

Growing evidences show that gut microbiota is associated with the pathogenesis of Parkinson's disease (PD) and the gut-brain axis can be promising target for the development of the therapeutic strategies for PD. Acupuncture has been used to improve brain functions and inflammation in neurological disorders such as PD, and to recover the gastrointestinal dysfunctions in various gastrointestinal disorders. Thus, we investigated whether acupuncture could improve Parkinsonism and gut microbial dysbiosis induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine. First, we observed that acupuncture treatment at acupoints GB34 and ST36 could improve motor functions and comorbid anxiety in PD mice. Next, we found that acupuncture increased the levels of dopaminergic fibers and neurons in the striatum and the substantia nigra, respectively. Acupuncture also restored the overexpression of microglia and astrocyte as well as conversion of Bax and Bcl-2 expression in both the striatum and the substantia nigra, indicating that inflammatory responses and apoptosis were blocked by acupuncture. Additionally, via 16S rRNA sequence analysis, we observed that the relative abundance of 18 genera were changed in acupuncture-treated mice compared to the PD mice. Of them, Butyricimonas, Holdemania, Frisingicoccus, Gracilibacter, Phocea, and Aestuariispira showed significant correlations with anxiety as well as motor functions. Furthermore, the predicted functional analyses showed that acupuncture restored the physiology functions such as glutathione metabolism, methane metabolism, and PD pathway. In conclusion, we suggest that the effects of acupuncture on the enhanced motor function and the protection of the dopaminergic neurons may be associated with the regulation of the gut microbial dysbiosis and thus the inhibition of the neuroinflammation in the PD mice.


Subject(s)
Acupuncture Therapy , Gastrointestinal Microbiome , Parkinson Disease , Animals , Disease Models, Animal , Dopaminergic Neurons , Dysbiosis/complications , Mice , Mice, Inbred C57BL , Parkinson Disease/therapy , RNA, Ribosomal, 16S , Substantia Nigra
15.
Br J Anaesth ; 123(5): 637-654, 2019 11.
Article in English | MEDLINE | ID: mdl-31551115

ABSTRACT

The relationship between gut microbiota and neurological diseases, including chronic pain, has received increasing attention. The gut microbiome is a crucial modulator of visceral pain, whereas recent evidence suggests that gut microbiota may also play a critical role in many other types of chronic pain, including inflammatory pain, headache, neuropathic pain, and opioid tolerance. We present a narrative review of the current understanding on the role of gut microbiota in pain regulation and discuss the possibility of targeting gut microbiota for the management of chronic pain. Numerous signalling molecules derived from gut microbiota, such as by-products of microbiota, metabolites, neurotransmitters, and neuromodulators, act on their receptors and remarkably regulate the peripheral and central sensitisation, which in turn mediate the development of chronic pain. Gut microbiota-derived mediators serve as critical modulators for the induction of peripheral sensitisation, directly or indirectly regulating the excitability of primary nociceptive neurones. In the central nervous system, gut microbiota-derived mediators may regulate neuroinflammation, which involves the activation of cells in the blood-brain barrier, microglia, and infiltrating immune cells, to modulate induction and maintenance of central sensitisation. Thus, we propose that gut microbiota regulates pain in the peripheral and central nervous system, and targeting gut microbiota by diet and pharmabiotic intervention may represent a new therapeutic strategy for the management of chronic pain.


Subject(s)
Chronic Pain/microbiology , Gastrointestinal Microbiome/physiology , Pain Management/methods , Blood-Brain Barrier/physiopathology , Chronic Pain/physiopathology , Chronic Pain/therapy , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Humans , Inflammation/microbiology , Neuroimmunomodulation/physiology , Visceral Pain/microbiology , Visceral Pain/therapy
16.
Immunology ; 154(2): 230-238, 2018 06.
Article in English | MEDLINE | ID: mdl-29637999

ABSTRACT

A vast number of studies have demonstrated a remarkable role for the gut microbiota and their metabolites in the pathogenesis of inflammatory diseases, including multiple sclerosis (MS). Recent studies in experimental autoimmune encephalomyelitis, an animal model of MS, have revealed that modifying certain intestinal bacterial populations may influence immune cell priming in the periphery, resulting in dysregulation of immune responses and neuroinflammatory processes in the central nervous system (CNS). Conversely, some commensal bacteria and their antigenic products can protect against inflammation within the CNS. Specific components of the gut microbiome have been implicated in the production of pro-inflammatory cytokines and subsequent generation of Th17 cells. Similarly, commensal bacteria and their metabolites can also promote the generation of regulatory T-cells (Treg), contributing to immune suppression. Short-chain fatty acids may induce Treg either by G-protein-coupled receptors or inhibition of histone deacetylases. Tryptophan metabolites may suppress inflammatory responses by acting on the aryl hydrocarbon receptor in T-cells or astrocytes. Interestingly, secretion of these metabolites can be impaired by excess consumption of dietary components, such as long-chain fatty acids or salt, indicating that the diet represents an environmental factor affecting the complex crosstalk between the gut microbiota and the immune system. This review discusses new aspects of host-microbiota interaction and the immune system with a special focus on MS as a prototype T-cell-mediated autoimmune disease of the CNS.


Subject(s)
Autoimmunity , Energy Metabolism , Immunomodulation , Microbiota , Animals , Autoimmune Diseases of the Nervous System/etiology , Autoimmune Diseases of the Nervous System/metabolism , Autoimmune Diseases of the Nervous System/pathology , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/pathology , Disease Susceptibility , Fatty Acids, Volatile/metabolism , Gastrointestinal Microbiome/immunology , Humans , Microbiota/immunology , Neuroimmunomodulation , Probiotics , Tryptophan/metabolism
17.
J Neuroinflammation ; 15(1): 190, 2018 Jun 22.
Article in English | MEDLINE | ID: mdl-29933742

ABSTRACT

BACKGROUND: Chronic noise exposure is associated with neuroinflammation and gut microbiota dysregulation and increases the risk of Alzheimer's disease (AD). Environmental hazards are also thought to be associated with genetic susceptibility factors that increase AD pathogenesis. However, there is limited experimental evidence regarding the link between chronic noise stress and microbiome-gut-brain axis alterations, which may be closely related to AD development. METHODS: The aim of the present study was to systematically investigate the effects of chronic noise exposure on the microbiome-gut-brain axis in the senescence-accelerated mouse prone 8 (SAMP8) strain. We established SAMP8 mouse models to examine the consequences of noise exposure on the microbiome-gut-brain axis. Hippocampal amyloid-ß (Aß) assessment and the Morris water maze were used to evaluate AD-like changes, 16S ribosomal RNA sequencing analyses were used for intestinal flora measurements, and assessment of endothelial tight junctions and serum neurotransmitter and inflammatory mediator levels, as well as fecal microbiota transplant, was conducted to explore the underlying pathological mechanisms. RESULTS: Chronic noise exposure led to cognitive impairment and Aß accumulation in young SAMP8 mice, similar to that observed in aging SAMP8 mice. Noise exposure was also associated with decreased gut microbiota diversity and compositional alterations. Axis-series studies showed that endothelial tight junction proteins were decreased in both the intestine and brain, whereas serum neurotransmitter and inflammatory mediator levels were elevated in young SAMP8 mice exposed to chronic noise, similar to the observations made in the aging group. The importance of intestinal bacteria in noise exposure-induced epithelial integrity impairment and Aß accumulation was further confirmed through microbiota transplantation experiments. Moreover, the effects of chronic noise were generally intensity-dependent. CONCLUSION: Chronic noise exposure altered the gut microbiota, accelerated age-related neurochemical and inflammatory dysregulation, and facilitated AD-like changes in the brain of SAMP8 mice.


Subject(s)
Aging/genetics , Brain/metabolism , Cytokines/metabolism , Gastrointestinal Microbiome/physiology , Noise/adverse effects , Aging/blood , Animals , Avoidance Learning/physiology , Claudins/genetics , Claudins/metabolism , Cognition Disorders/etiology , Corticosterone/blood , Cytokines/genetics , Disease Models, Animal , Endotoxins/blood , Inflammation/etiology , Inflammation/metabolism , Mice , Microbiota/physiology , Occludin/genetics , Occludin/metabolism , RNA, Ribosomal, 16S/metabolism , Serotonin/blood , Tight Junctions/pathology , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-2 Protein/genetics , Zonula Occludens-2 Protein/metabolism , gamma-Aminobutyric Acid/blood
18.
Digestion ; 97(2): 137-145, 2018.
Article in English | MEDLINE | ID: mdl-29310114

ABSTRACT

BACKGROUND: The gut microbiome has been developing and making adaption all the time, which is consistent with their host from the initial colonization at birth or earlier. Emerging evidence is showing that dysbiosis is involved in various diseases associated with immune, metabolism, infection, nervous system, social behaviors, and psychopathology, etc., maybe via modulating gut barrier, microbiome-gut-brain axis, or some metabolites like short-chain fatty acids (SCFAs). SUMMARY: In the review, we will conclude the recent researches related to the influence of microbiome on local structure, function, regulation, metabolism of gut, and systematic modulation to the host, as well as some affective factors such as diet or antibiotics. Key Messages: It is a reasonable hypothesis that the balance of bioactive factors or cells and the opposites such as the regulatory T/helper T17 balance and interleukin (IL)-10/IL-17 balance plays a vital role in homeostasis of immunity system. Meanwhile, the link between gut microbiome and immune system via microbiota-derived metabolite SCFAs involved in multi-function of the host locally and systematically has been revealed. We hope to contribute to the microbiome-targeted treatment and prevention of some diseases.


Subject(s)
Dysbiosis/immunology , Dysbiosis/physiopathology , Gastrointestinal Diseases/immunology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/metabolism , Anti-Bacterial Agents/adverse effects , Diet , Dysbiosis/chemically induced , Fatty Acids, Volatile/metabolism , Gastrointestinal Diseases/microbiology , Gastrointestinal Microbiome/drug effects , Humans , Immunity/physiology , Intestinal Mucosa/innervation , Intestinal Mucosa/microbiology , Myenteric Plexus , Permeability , T-Lymphocytes/immunology
19.
Microb Ecol Health Dis ; 29(2): 1548249, 2018.
Article in English | MEDLINE | ID: mdl-30510497

ABSTRACT

This article focuses on autointoxication, a discredited medical theory from the late nineteenth century that provides important points of reflection for today's research on the role of microbes in the human gut for mental health. It considers how the theory of autointoxication, which came into great prominence amongst physicians and the general public worldwide, fell from grace by the middle of the twentieth century, and briefly asks why studies of the human microbiome are now back in vogue. It departs from earlier articles on the topic firstly by arguing that autointoxication theory was especially prevalent in France, and secondly by focusing on the application of this theory to mental health. Bringing to light medical treatises and theses from this period which have so far remained unexamined, it shows that examining the development and reception of medical theories form the past can help us today in understanding both the pitfalls and promise of research in this area.

20.
Microb Ecol Health Dis ; 29(2): 1548250, 2018.
Article in English | MEDLINE | ID: mdl-30532687

ABSTRACT

Background: In recent decades, dominant models of mental illness have become increasingly focused on the head, with mental disorders being figured as brain disorders. However, research into the active role that the microbiome-gut-brain axis plays in affecting mood and behaviour may lead to the conclusion that mental health is more than an internalised problem of individual brains. Objective: This article explores the implications of shifting understandings about mental health that have come about through research into links between the gut microbiome and mental health problems such as depression and anxiety. It aims to analyse the different ways that the lines between mind and body and mental and physical health are re-shaped by this research, which is starting to inform clinical and public understanding. Design: As mental health has become a pressing issue of political and public concern it has become increasingly constructed in socio-cultural and personal terms beyond clinical spaces, requiring a conceptual response that exceeds biomedical inquiry. This article argues that an interdisciplinary critical medical humanities approach is well positioned to analyse the impact of microbiome-gut-brain research on conceptions of mind. Results: The entanglement of mind and matter evinced by microbiome-gut-brain axis research potentially provides a different way to conceptualise the physical and social concomitants of mental distress. Conclusion: Mental health is not narrowly located in the head but is assimilated by the physical body and intermingled with the natural world, requiring different methods of research to unfold the meanings and implications of gut thinking for conceptions of human selfhood.

SELECTION OF CITATIONS
SEARCH DETAIL