37 results on '"brain–gut–microbiome axis"'
Search Results
2. The Role of Interactions along the Brain–Gut–Microbiome Axis in the Regulation of Circadian Rhythms, Sleep Mechanisms, and Their Disorders.
- Author
-
Shirolapov, I. V., Gribkova, O. V., Kovalev, A. M., Shafigullina, L. R., Ulivanova, V. A., Kozlov, A. V., Ereshchenko, A. A., Lyamin, A. V., and Zakharov, A. V.
- Subjects
DISEASE risk factors ,BACTERIAL cell walls ,SLEEP-wake cycle ,SHORT-chain fatty acids ,GUT microbiome - Abstract
The bidirectional communication between brain structures and the gastrointestinal tract involving the microbiota defines the scientific concept of the brain–gut–microbiome axis. The intestinal microbiome plays an important role in many physiological and biochemical processes in the body, in the immune response, in the maintenance of homeostasis, and in the regulation of circadian rhythms. There is a relationship between higher prevalences of a number of neurological disorders, sleep disorders, and changes in the intestinal microbiota, so it is important to study the complex mechanisms of such correlations for the development of new treatment and prevention strategies. Environmental factors associated with excessive exposure to light can aggravate dysbiosis of the intestinal microflora, producing sleep disorders as a result. This review discusses the integrative mechanisms of regulation of sleep involving the intestinal microbiota (the roles of neurotransmitters, short-chain fatty acids, unconjugated bile acids, bacterial cell wall components, and cytokines). Taking account of the influences of intestinal dysbiosis as a risk factor in the development of various diseases, this review systematizes the key aspects and modern scientific data on the importance of the balance of the microflora to ensure optimal interactions along the brain–gut–microbiome axis in the context of the regulatory role of the sleep-waking cycle and its disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
3. The Involvement of Immune Cells Between Ischemic Stroke and Gut Microbiota.
- Author
-
Pu, Bei, Zhu, Hua, Wei, Liang, Gu, Lijuan, Zhang, Shenqi, Jian, Zhihong, and Xiong, Xiaoxing
- Abstract
Ischemic stroke, a disease with high mortality and disability rate worldwide, currently has no effective treatment. The systemic inflammation response to the ischemic stroke, followed by immunosuppression in focal neurologic deficits and other inflammatory damage, reduces the circulating immune cell counts and multiorgan infectious complications such as intestinal and gut dysfunction dysbiosis. Evidence showed that microbiota dysbiosis plays a role in neuroinflammation and peripheral immune response after stroke, changing the lymphocyte populations. Multiple immune cells, including lymphocytes, engage in complex and dynamic immune responses in all stages of stroke and may be a pivotal moderator in the bidirectional immunomodulation between ischemic stroke and gut microbiota. This review discusses the role of lymphocytes and other immune cells, the immunological processes in the bidirectional immunomodulation between gut microbiota and ischemic stroke, and its potential as a therapeutic strategy for ischemic stroke. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
4. Мікробіом людини та ментальне здоров'я: новітні погляди
- Author
-
Артьоменко, В. В., Жовтенко, О. В., Стасій, Я. О., and Пірон-Думітраску, М.
- Abstract
Mental health is the emotional, psychological, and social well-being of a person who feels protected, needed, realizes his abilities, and can overcome life's stresses. Mental disorders (MD) are a serious health problem worldwide today, which lead to a significant medical burden and economic losses for both medical institutions and patients and their families. Today, MD are one of the priority directions in the daily practice of Ukrainian doctors. MD can include anxiety, depression, bipolar disorder, autism spectrum disorder, schizophrenia, eating disorders, etc. Recently, the attention of scientists and doctors has been paid to the interaction between the microbiome and mental health. The researches which are aimed at defining what is a healthy microbiome have revealed significant individual differences in its composition and diversity. It has been established that a certain composition of the gut microbiome (GM) is associated with the development of anxiety disorders. Research results show that patients and animal models (mice) with anxiety disorders have a dramatic decrease in microbial richness and diversity. Patients with anxiety disorder typically had reduced Firmicutes and increased Bacteroidetes and Fusobacteria. In addition, GM changes were associated with the onset and development of depressive disorder. Differences in the composition of fecal microbiota in patients with major depressive disorder were revealed. The main methods of MD treatment today are pharmacotherapy and psychotherapy, which have limited effectiveness. However, practitioners are looking for alternative ways to help such patients. The results of many studies indicate that prebiotics (e.g., dietary fiber and alpha-lactalbumin) as well as postbiotics show a protective effect on mental health, especially when used in combination. At the same time, prebiotics may be a potential agent for alleviating the side effects of antipsychotics used in the treatment of MD. More experimental researches and high-quality clinical trials are needed to study the effects of dietary components on MD through the brain-gut-microbiome axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
5. Bidirectional regulation of the brain–gut–microbiota axis following traumatic brain injury
- Author
-
Xinyu You, Lin Niu, Jiafeng Fu, Shining Ge, Jiangwei Shi, Yanjun Zhang, and Pengwei Zhuang
- Subjects
traumatic brain injury ,brain–gut–microbiome axis ,gut microbiota ,neuroimmune ,immunosuppression ,host defense ,vagal afferents ,bacterial infection ,dorsal root ganglia ,nociception neural circuitry ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Traumatic brain injury is a prevalent disorder of the central nervous system. In addition to primary brain parenchymal damage, the enduring biological consequences of traumatic brain injury pose long-term risks for patients with traumatic brain injury; however, the underlying pathogenesis remains unclear, and effective intervention methods are lacking. Intestinal dysfunction is a significant consequence of traumatic brain injury. Being the most densely innervated peripheral tissue in the body, the gut possesses multiple pathways for the establishment of a bidirectional “brain–gut axis” with the central nervous system. The gut harbors a vast microbial community, and alterations of the gut niche contribute to the progression of traumatic brain injury and its unfavorable prognosis through neuronal, hormonal, and immune pathways. A comprehensive understanding of microbiota-mediated peripheral neuroimmunomodulation mechanisms is needed to enhance treatment strategies for traumatic brain injury and its associated complications. We comprehensively reviewed alterations in the gut microecological environment following traumatic brain injury, with a specific focus on the complex biological processes of peripheral nerves, immunity, and microbes triggered by traumatic brain injury, encompassing autonomic dysfunction, neuroendocrine disturbances, peripheral immunosuppression, increased intestinal barrier permeability, compromised responses of sensory nerves to microorganisms, and potential effector nuclei in the central nervous system influenced by gut microbiota. Additionally, we reviewed the mechanisms underlying secondary biological injury and the dynamic pathological responses that occur following injury to enhance our current understanding of how peripheral pathways impact the outcome of patients with traumatic brain injury. This review aimed to propose a conceptual model for future risk assessment of central nervous system-related diseases while elucidating novel insights into the bidirectional effects of the “brain–gut–microbiota axis.”
- Published
- 2025
- Full Text
- View/download PDF
6. Cognitive behavioral therapy for irritable bowel syndrome induces bidirectional alterations in the brain-gut-microbiome axis associated with gastrointestinal symptom improvement
- Author
-
Jacobs, Jonathan P, Gupta, Arpana, Bhatt, Ravi R, Brawer, Jacob, Gao, Kan, Tillisch, Kirsten, Lagishetty, Venu, Firth, Rebecca, Gudleski, Gregory D, Ellingson, Benjamin M, Labus, Jennifer S, Naliboff, Bruce D, Lackner, Jeffrey M, and Mayer, Emeran A
- Subjects
Microbiology ,Biological Sciences ,Mind and Body ,Clinical Trials and Supportive Activities ,Clinical Research ,Digestive Diseases ,Neurosciences ,Mental Health ,Nutrition ,Mental health ,Oral and gastrointestinal ,Brain-Gut Axis ,Cognitive Behavioral Therapy ,Gastrointestinal Microbiome ,Humans ,Irritable Bowel Syndrome ,RNA ,Ribosomal ,16S ,Cognitive behavioral therapy ,Irritable bowel syndrome ,Brain-gut-microbiome axis ,Neuroimaging ,Biomarkers ,Outcome prediction ,Ecology ,Medical Microbiology ,Evolutionary biology - Abstract
BackgroundThere is growing recognition that bidirectional signaling between the digestive tract and the brain contributes to irritable bowel syndrome (IBS). We recently showed in a large randomized controlled trial that cognitive behavioral therapy (CBT) reduces IBS symptom severity. This study investigated whether baseline brain and gut microbiome parameters predict CBT response and whether response is associated with changes in the brain-gut-microbiome (BGM) axis.MethodsEighty-four Rome III-diagnosed IBS patients receiving CBT were drawn from the Irritable Bowel Syndrome Outcome Study (IBSOS; ClinicalTrials.gov NCT00738920) for multimodal brain imaging and psychological assessments at baseline and after study completion. Fecal samples were collected at baseline and post-treatment from 34 CBT recipients for 16S rRNA gene sequencing, untargeted metabolomics, and measurement of short-chain fatty acids. Clinical measures, brain functional connectivity and microstructure, and microbiome features associated with CBT response were identified by multivariate linear and negative binomial models.ResultsAt baseline, CBT responders had increased fecal serotonin levels, and increased Clostridiales and decreased Bacteroides compared to non-responders. A random forests classifier containing 11 microbial genera predicted CBT response with high accuracy (AUROC 0.96). Following treatment, CBT responders demonstrated reduced functional connectivity in regions of the sensorimotor, brainstem, salience, and default mode networks and changes in white matter in the basal ganglia and other structures. Brain changes correlated with microbiome shifts including Bacteroides expansion in responders.ConclusionsPre-treatment intestinal microbiota and serotonin levels were associated with CBT response, suggesting that peripheral signals from the microbiota can modulate central processes affected by CBT that generate abdominal symptoms in IBS. CBT response is characterized by co-correlated shifts in brain networks and gut microbiome that may reflect top-down effects of the brain on the microbiome during CBT. Video abstract.
- Published
- 2021
7. Association between selective serotonin reuptake inhibitor use and developing irritable bowel syndrome through retrospective analysis.
- Author
-
Kwak, Nayoung, Lee, Hankil, Kim, Beom Kyung, Yu, Yun Mi, and Kang, Hye‐Young
- Subjects
- *
SEROTONIN uptake inhibitors , *IRRITABLE colon , *PROPORTIONAL hazards models , *NATIONAL health insurance , *PSYCHIATRIC drugs - Abstract
Background and Aim: Serotonin affects the balance and integrity of the gut microbiome; however, studies have confirmed the influence of selective serotonin reuptake inhibitors (SSRIs) on irritable bowel syndrome (IBS). We evaluated the association between SSRI use and subsequent IBS occurrence in a real‐world setting. Methods: A multivariate Cox proportional hazard model was adopted, and the National Health Insurance Service cohort claims database between 2010 and 2019 was used. Non‐SSRI users were selected using the propensity score matching method. Subgroup analyses were performed using the point of use, cumulative dose, and duration of SSRI use. Additional analysis was performed using a control group without psychiatric medications. Results: We included 2901 SSRI users and 2727 non‐SSRI users. After adjusting covariates, the risk of developing IBS in SSRI users was 1.54 times that in non‐SSRI users (95% confidence interval [CI]: 1.01–2.33). The hazard ratio (HR) of the recent, heavy, and short‐term user groups were 3.19 (95% CI: 2.03–4.99), 2.22 (95% CI: 1.50–3.29), and 4.83 (95% CI: 3.02–7.73), respectively, compared with that of non‐users. In patients without a history of psychiatric medications, the risk of IBS incidence after SSRI use increased significantly (HR: 1.61, 95% CI: 1.06–2.42), whereas HR was insignificant in patients with a history of psychiatric medications (HR: 1.25, 95% CI: 0.98–1.60). Conclusions: The risk of subsequent IBS occurrence following SSRI use was high in patients who initially took a heavy SSRI dose and those who did not have a history of psychiatric drug use. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Dynamical alterations of brain function and gut microbiome in weight loss.
- Author
-
Jing Zhou, Xiaoling Wu, Tianyuan Xiang, Fei Liu, Hui Gao, Li Tong, Bin Yan, Zhonglin Li, Chi Zhang, Linyuan Wang, Lei Ou, Zhongxia Li, Wen Wang, Tingting Yang, Fengyun Li, Huimin Ma, Xiaojuan Zhao, Na Mi, Ziya Yu, and Canhui Lan
- Subjects
GUT microbiome ,CINGULATE cortex ,ESCHERICHIA coli ,FUNCTIONAL magnetic resonance imaging - Abstract
Objective: Intermittent energy restriction (IER) is an effective weight loss strategy. However, little is known about the dynamic effects of IER on the brain-gut-microbiome axis. Methods: In this study, a total of 25 obese individuals successfully lost weight after a 2-month IER intervention. FMRI was used to determine the activity of brain regions. Metagenomic sequencing was performed to identify differentially abundant gut microbes and pathways in from fecal samples. Results: Our results showed that IER longitudinally reduced the activity of obeserelated brain regions at different timepoints, including the inferior frontal orbital gyrus in the cognitive control circuit, the putamen in the emotion and learning circuit, and the anterior cingulate cortex in the sensory circuit. IER longitudinally reduced E. coli abundance across multiple timepoints while elevating the abundance of obesity-related Faecalibacterium prausnitzii, Parabacteroides distasonis, and Bacterokles uniformis. Correlation analysis revealed longitudinally correlations between gut bacteria abundance alterations and brain activity changes. Conclusions: There was dynamical alteration of BGM axis (the communication of E. coli with specific brain regions) during the weight loss under the IER. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Dynamical alterations of brain function and gut microbiome in weight loss
- Author
-
Jing Zhou, Xiaoling Wu, Tianyuan Xiang, Fei Liu, Hui Gao, Li Tong, Bin Yan, Zhonglin Li, Chi Zhang, Linyuan Wang, Lei Ou, Zhongxia Li, Wen Wang, Tingting Yang, Fengyun Li, Huimin Ma, Xiaojuan Zhao, Na Mi, Ziya Yu, Canhui Lan, Qi Wang, Hao Li, Liming Wang, Xiaoning Wang, Yongli Li, and Qiang Zeng
- Subjects
brain-gut-microbiome axis ,weight loss ,functional magnetic resonance imaging ,intermittent energy restriction ,metagenomics ,Microbiology ,QR1-502 - Abstract
ObjectiveIntermittent energy restriction (IER) is an effective weight loss strategy. However, little is known about the dynamic effects of IER on the brain-gut-microbiome axis.MethodsIn this study, a total of 25 obese individuals successfully lost weight after a 2-month IER intervention. FMRI was used to determine the activity of brain regions. Metagenomic sequencing was performed to identify differentially abundant gut microbes and pathways in from fecal samples.ResultsOur results showed that IER longitudinally reduced the activity of obese-related brain regions at different timepoints, including the inferior frontal orbital gyrus in the cognitive control circuit, the putamen in the emotion and learning circuit, and the anterior cingulate cortex in the sensory circuit. IER longitudinally reduced E. coli abundance across multiple timepoints while elevating the abundance of obesity-related Faecalibacterium prausnitzii, Parabacteroides distasonis, and Bacterokles uniformis. Correlation analysis revealed longitudinally correlations between gut bacteria abundance alterations and brain activity changes.ConclusionsThere was dynamical alteration of BGM axis (the communication of E. coli with specific brain regions) during the weight loss under the IER.
- Published
- 2023
- Full Text
- View/download PDF
10. Association of aberrant brain network dynamics with gut microbial composition uncovers disrupted brain–gut–microbiome interactions in irritable bowel syndrome: Preliminary findings.
- Author
-
Yang, Lin, Liu, Guangyao, Li, Shan, Yao, Chaofan, Zhao, Ziyang, Chen, Nan, Zhang, Pengfei, Shang, Yingying, Wang, Yin, Zhang, Dekui, Tian, Xiaozhu, Zhang, Jing, Yao, Zhijun, and Hu, Bin
- Subjects
- *
GUT microbiome , *IRRITABLE colon , *LARGE-scale brain networks , *FUNCTIONAL magnetic resonance imaging , *FUNCTIONAL connectivity - Abstract
Background and purpose: Growing evidence suggests that abnormalities in brain–gut–microbiome (BGM) interactions are involved in the pathogenesis of irritable bowel syndrome (IBS). Our study aimed to explore alterations in dynamic functional connectivity (DFC), the gut microbiome and the bidirectional interaction in the BGM. Methods: Resting‐state functional magnetic resonance imaging (rs‐fMRI), fecal samples and clinical chacteristics were collected from 33 IBS patients and 32 healthy controls. We performed a systematic DFC analysis on rs‐fMRI. The gut microbiome was analyzed by 16S rRNA gene sequencing. Associations between DFC characteristics and microbial alterations were explored. Results: In the DFC analysis, four dynamic functional states were identified. IBS patients exhibited increased mean dwell and fraction time in State 4, and reduced transitions from State 3 to State 1. Aberrant temporal properties in State 4 were only evident when choosing a short window (36 s or 44 s). Decreased functional connectivity (FC) variability was found in State 1 and State 3 in IBS patients, two of which (independent component [IC]51‐IC91, IC46‐IC11) showed significant correlations with clinical characteristics. Additionally, we identified nine significantly differential abundances in microbial composition. We also found that IBS‐related microbiota were associated with aberrant FC variability, although these exploratory results were obtained at an uncorrected threshold of significance. Conclusions: Although future studies are needed to confirm our results, the findings not only provide a new insight into the dysconnectivity hypothesis in IBS from a dynamic perspective, but also establish a possible link between DFC and the gut microbiome, which lays the foundation for future research on disrupted BGM interactions. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
11. Identification of "missing links" in C- and D-ring cleavage of steroids by Comamonas testosteroni TA441.
- Author
-
Masae Horinouchi and Toshiaki Hayashi
- Subjects
- *
KETONES , *HYDROXYL group , *AROMATIZATION , *CHOLIC acid , *BILE acids - Abstract
Comamonas testosteroni TA441 is capable of aerobically degrading steroids through the aromatization and cleavage of the A- and B-rings, followed by D- and C-ring cleavage via ß-oxidation. While most of the degradation steps have been previously characterized, a few intermediate compounds remained unidentified. In this study, we proposed that the cleavage of the D-ring at C13-17 required the ScdY hydratase, followed by C-ring cleavage via the ScdL1L2 transferase. The anticipated reaction was expected to yield 6-methyl-3,7-dioxo-decane-1,10-dioic acid-coenzyme A (CoA) ester. To confirm this hypothesis, we constructed a plasmid enabling the induction of targeted genes in TA441 mutant strains. Induction experiments of ScdL1L2 revealed that the major product was 3-hydroxy-6-methyl-7-oxo-decane-1,10-dioic acid-CoA ester. Similarly, induction experiments of ScdY demonstrated that the substrate of ScdY was a geminal diol, 17-dihydroxy-9-oxo-1,2,3,4,5,6,10,19-octanorandrost-8(14)-en-7-oic acid-CoA ester. These findings suggest that ScdY catalyzes the addition of a water molecule at C14 of 17-dihydroxy-9-oxo-1,2,3,4,5,6,10,19-octanorandrost-8(14)-en-7-oic acid-CoA ester, leading to D-ring cleavage at C13-17. Subsequently, the C9 ketone of the D-ring cleavage product is converted to a hydroxyl group, followed by C-ring cleavage, resulting in the production of 3-hydroxy-6-methyl-7-oxo-decane-1,10-dioic acid-CoA ester. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
12. Comprehensive summary of steroid metabolism in Comamonas testosteroni TA441: entire degradation process of basic four rings and removal of C12 hydroxyl group.
- Author
-
Masae Horinouchi and Toshiaki Hayashi
- Subjects
- *
HYDROXYL group , *DOUBLE bonds , *STEROIDS , *AEROBIC bacteria , *KETONES - Abstract
Comamonas testosteroni is one of the representative aerobic steroid-degrading bacteria. We previously revealed the mechanism of steroidal A,B,C,D-ring degradation by C. testosteroni TA441. The corresponding genes are located in two clusters at both ends of a mega-cluster of steroid degradation genes. ORF7 and ORF6 are the only two genes in these clusters, whose function has not been determined. Here, we characterized ORF7 as encoding the dehydrase responsible for converting the C12ß hydroxyl group to the C10(12) double bond on the C-ring (SteC), and ORF6 as encoding the hydrogenase responsible for converting the C10(12) double bond to a single bond (SteD). SteA and SteB, encoded just upstream of SteC and SteD, are in charge of oxidizing the C12a hydroxyl group to a ketone group and of reducing the latter to the C12ß hydroxyl group, respectively. Therefore, the C12a hydroxyl group in steroids is removed with SteABCD via the C12 ketone and C12ß hydroxyl groups. Given the functional characterization of ORF6 and ORF7, we disclose the entire pathway of steroidal A,B,C,D-ring breakdown by C. testosteroni TA441. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
13. The Role of the Microbiome in First Episode of Psychosis.
- Author
-
Nuncio-Mora, Lucero, Lanzagorta, Nuria, Nicolini, Humberto, Sarmiento, Emmanuel, Ortiz, Galo, Sosa, Fernanda, and Genis-Mendoza, Alma Delia
- Subjects
SHORT-chain fatty acids ,PSYCHOSES ,MENTAL illness - Abstract
The relationship between the gut-brain-microbiome axis has gained great importance in the study of psychiatric disorders, as it may represent a new target for their treatment. To date, the available literature suggests that the microbiota may influence the pathophysiology of several diseases, including psychosis. The aim of this review is to summarize the clinical and preclinical studies that have evaluated the differences in microbiota as well as the metabolic consequences related to psychosis. Current data suggest that the genera Lactobacillus and Megasphaera are increased in schizophrenia (SZ), as well as alterations in the glutamate-glutamine-GABA cycle, serum levels of tryptophan, kynurenic acid (KYNA), and short-chain fatty acids (SCFAs). There are still very few studies on early-onset psychosis, thus more studies are needed to be able to propose targeted therapies for a point when the disease has just started or has not yet progressed. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
14. Gut–Brain Axis: Role of Gut Microbiota in Neurodegenerative Disease
- Author
-
Husarik, Aarti Narang, Sandhir, Rajat, Deol, Parneet Kaur, editor, Sandhu, Simarjot Kaur, editor, and Kaur, Indu Pal, Editor-in-Chief
- Published
- 2022
- Full Text
- View/download PDF
15. Improvement in Uncontrolled Eating Behavior after Laparoscopic Sleeve Gastrectomy Is Associated with Alterations in the Brain–Gut–Microbiome Axis in Obese Women
- Author
-
Dong, Tien S, Gupta, Arpana, Jacobs, Jonathan P, Lagishetty, Venu, Gallagher, Elizabeth, Bhatt, Ravi R, Vora, Priten, Osadchiy, Vadim, Stains, Jean, Balioukova, Anna, Chen, Yijun, Dutson, Erik, Mayer, Emeran A, and Sanmiguel, Claudia
- Subjects
Biomedical and Clinical Sciences ,Nutrition and Dietetics ,Clinical Sciences ,Digestive Diseases ,Behavioral and Social Science ,Obesity ,Prevention ,Microbiome ,Neurosciences ,Nutrition ,Biomedical Imaging ,Brain Disorders ,Women's Health ,Clinical Research ,Basic Behavioral and Social Science ,Oral and gastrointestinal ,Adolescent ,Adult ,Bariatric Surgery ,Brain ,Diet ,Female ,Food Addiction ,Gastrectomy ,Gastrointestinal Microbiome ,Health Behavior ,Humans ,Laparoscopy ,Magnetic Resonance Imaging ,Middle Aged ,Surveys and Questionnaires ,Weight Loss ,Young Adult ,bariatric surgery ,brain– ,gut– ,microbiome axis ,metabolite ,obesity ,brain ,brain–gut–microbiome axis ,Food Sciences ,Clinical sciences ,Nutrition and dietetics ,Public health - Abstract
BackgroundBariatric surgery is proven to change eating behavior and cause sustained weight loss, yet the exact mechanisms underlying these changes are not clearly understood. We explore this in a novel way by examining how bariatric surgery affects the brain-gut-microbiome (BGM) axis.MethodsPatient demographics, serum, stool, eating behavior questionnaires, and brain magnetic resonance imaging (MRI) were collected before and 6 months after laparoscopic sleeve gastrectomy (LSG). Differences in eating behavior and brain morphology and resting-state functional connectivity in core reward regions were correlated with serum metabolite and 16S microbiome data.ResultsLSG resulted in significant weight loss and improvement in maladaptive eating behaviors as measured by the Yale Food Addiction Scale (YFAS). Brain imaging showed a significant increase in brain volume of the putamen (p.adj < 0.05) and amygdala (p.adj < 0.05) after surgery. Resting-state connectivity between the precuneus and the putamen was significantly reduced after LSG (p.adj = 0.046). This change was associated with YFAS symptom count. Bacteroides, Ruminococcus, and Holdemanella were associated with reduced connectivity between these areas. Metabolomic profiles showed a positive correlation between this brain connection and a phosphatidylcholine metabolite.ConclusionBariatric surgery modulates brain networks that affect eating behavior, potentially through effects on the gut microbiota and its metabolites.
- Published
- 2020
16. Wuzang Wenyang Huayu Tang promoting learning-memory ability in vascular dementia rats via brain-gut-microbiome axis
- Author
-
Yu-Cheng Lu, Meng-Qi Li, Ling Zhang, Yun-Li Tang, Yu-Fu Zeng, Yan-Chun Li, Jia-Jia Zhong, Yong-Fang Cheng, Wei-An Qiu, and Zi-Heng Huang
- Subjects
Vascular dementia ,Brain-gut-microbiome axis ,Autophag ,16S rRNA ,Fecal metabolomics ,Other systems of medicine ,RZ201-999 ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Introduction: Lots of research has revealed that vascular dementia (VD), which is closely related to the metabolic disorders of intestinal microbiota, can be treated through traditional Chinese drug by regulating the brain-gut-microbiome axis. However, the specific interaction between VD and intestinal microflora, as well as the mechanism of Wuzang Wenyang Huayu Tang (WWHT) in treating VD by regulating the brain-gut-microbiome axi remains unknown on the whole and needs to be further studied. Objectives: To explore the effect of WWHT on the learning and memory ability of rats with VD based on brain-gut-microbiome axis. Methods: In this study, taking the specific pathogen-free (SPF) SD rats as the subjects, a vascular dementia rat model was created by using the two-vessel occlusion model (2-VO). The subjects were divided into six groups, namely control group, sham group, VD model group, the piracetam group, the WWHT group, and the WWHT combined with piracetam (WCP) group. After 22 days of the gavage administration method, the Morris water maze experiment was adopted to determine the learning and memory abilities of each group through the water maze, observe the effects of hematoxylin and eosin (HE) staining and transmission electron microscopy in the hippocampal area of each group, and analyze the relevant proteins in the whole brain by Western Blot (WB). In addition, the feces of each group were collected and analyzed by fecal metabolomics combined with 16S rRNA gene sequencing to explore the interaction between VD and intestinal microbiota. Results: Through Morris water maze experiment, it was found that the learning and memory ability of VD rats in WWHT, piracetam and WCP groups was improved. The morphology of hippocampal neuron cells were improved in the WWHT group and the number of hippocampal neuron cells were increased in the VD rats by HE. By observing the ultrastructure of hippocampal cells, it can be found that the VD model group showed cell membrane edema and large organelle lysis. The ultrastructure of the hippocampus of all the administered groups was improved. In terms of the effect on the expression levels of related proteins in the whole brain of rats, compared with the VD model group, the expression levels of Caspase-3 protein decreased in all groups after drug administration. The protein expression levels of APG5L/ATG5, Beclin-1 and LC3A/B were upregulated, indicating that the drug administration group could regulate cellular autophagy in the whole brain to further protect brain tissue. Fecal metabolite profiles revealed 27 different metabolites related to VD. These metabolites are mainly involved in related metabolic pathways, such as vitamin B metabolism, purine metabolism, pyrimidine metabolism, and histidine metabolism. Further analysis of the gut microbiota by 16S rRNA gene sequencing data showed that WWHT-treated VD rats were different in gut microbiota composition. Discussion: Through the effect of brain-gut-microbiome axis, WWHT regulates the metabolism of vitamin B6 and tryptophan in VD rats, which can improve the intestinal microbiota of VD rats and regulate autophagy in the brain to improve cell morphology in the hippocampus, thus increasing the number of hippocampal cells to protect hippocampal neuron cells2510100, and finally improving the learning and memory ability of VD rats.
- Published
- 2023
- Full Text
- View/download PDF
17. Gut Microbiome and Gastrointestinal Disorders
- Author
-
Evangelia, Legaki, Karanasou, Eleni Anna, Gazouli, Maria, Gazouli, Maria, editor, and Theodoropoulos, George, editor
- Published
- 2021
- Full Text
- View/download PDF
18. Cognitive behavioral therapy for irritable bowel syndrome induces bidirectional alterations in the brain-gut-microbiome axis associated with gastrointestinal symptom improvement
- Author
-
Jonathan P. Jacobs, Arpana Gupta, Ravi R. Bhatt, Jacob Brawer, Kan Gao, Kirsten Tillisch, Venu Lagishetty, Rebecca Firth, Gregory D. Gudleski, Benjamin M. Ellingson, Jennifer S. Labus, Bruce D. Naliboff, Jeffrey M. Lackner, and Emeran A. Mayer
- Subjects
Cognitive behavioral therapy ,Irritable bowel syndrome ,Brain-gut-microbiome axis ,Neuroimaging ,Biomarkers ,Outcome prediction ,Microbial ecology ,QR100-130 - Abstract
Abstract Background There is growing recognition that bidirectional signaling between the digestive tract and the brain contributes to irritable bowel syndrome (IBS). We recently showed in a large randomized controlled trial that cognitive behavioral therapy (CBT) reduces IBS symptom severity. This study investigated whether baseline brain and gut microbiome parameters predict CBT response and whether response is associated with changes in the brain-gut-microbiome (BGM) axis. Methods Eighty-four Rome III-diagnosed IBS patients receiving CBT were drawn from the Irritable Bowel Syndrome Outcome Study (IBSOS; ClinicalTrials.gov NCT00738920) for multimodal brain imaging and psychological assessments at baseline and after study completion. Fecal samples were collected at baseline and post-treatment from 34 CBT recipients for 16S rRNA gene sequencing, untargeted metabolomics, and measurement of short-chain fatty acids. Clinical measures, brain functional connectivity and microstructure, and microbiome features associated with CBT response were identified by multivariate linear and negative binomial models. Results At baseline, CBT responders had increased fecal serotonin levels, and increased Clostridiales and decreased Bacteroides compared to non-responders. A random forests classifier containing 11 microbial genera predicted CBT response with high accuracy (AUROC 0.96). Following treatment, CBT responders demonstrated reduced functional connectivity in regions of the sensorimotor, brainstem, salience, and default mode networks and changes in white matter in the basal ganglia and other structures. Brain changes correlated with microbiome shifts including Bacteroides expansion in responders. Conclusions Pre-treatment intestinal microbiota and serotonin levels were associated with CBT response, suggesting that peripheral signals from the microbiota can modulate central processes affected by CBT that generate abdominal symptoms in IBS. CBT response is characterized by co-correlated shifts in brain networks and gut microbiome that may reflect top-down effects of the brain on the microbiome during CBT. Video abstract
- Published
- 2021
- Full Text
- View/download PDF
19. Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome.
- Author
-
Labus, Jennifer S, Hollister, Emily B, Jacobs, Jonathan, Kirbach, Kyleigh, Oezguen, Numan, Gupta, Arpana, Acosta, Jonathan, Luna, Ruth Ann, Aagaard, Kjersti, Versalovic, James, Savidge, Tor, Hsiao, Elaine, Tillisch, Kirsten, and Mayer, Emeran A
- Subjects
Brain ,Feces ,Humans ,Bacteria ,Irritable Bowel Syndrome ,DNA ,Bacterial ,DNA ,Ribosomal ,RNA ,Ribosomal ,16S ,Sequence Analysis ,DNA ,Phylogeny ,Adult ,Female ,Male ,Young Adult ,Gastrointestinal Microbiome ,Bacteroidetes ,Brain-gut-microbiome axis ,Firmicutes ,Irritable bowel syndrome ,Metagenome ,Ecology ,Microbiology ,Medical Microbiology - Abstract
BackgroundPreclinical and clinical evidence supports the concept of bidirectional brain-gut microbiome interactions. We aimed to determine if subgroups of irritable bowel syndrome (IBS) subjects can be identified based on differences in gut microbial composition, and if there are correlations between gut microbial measures and structural brain signatures in IBS.MethodsBehavioral measures, stool samples, and structural brain images were collected from 29 adult IBS and 23 healthy control subjects (HCs). 16S ribosomal RNA (rRNA) gene sequencing was used to profile stool microbial communities, and various multivariate analysis approaches were used to quantitate microbial composition, abundance, and diversity. The metagenomic content of samples was inferred from 16S rRNA gene sequence data using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). T1-weighted brain images were acquired on a Siemens Allegra 3T scanner, and morphological measures were computed for 165 brain regions.ResultsUsing unweighted Unifrac distances with hierarchical clustering on microbial data, samples were clustered into two IBS subgroups within the IBS population (IBS1 (n = 13) and HC-like IBS (n = 16)) and HCs (n = 23) (AUROC = 0.96, sensitivity 0.95, specificity 0.67). A Random Forest classifier provided further support for the differentiation of IBS1 and HC groups. Microbes belonging to the genera Faecalibacterium, Blautia, and Bacteroides contributed to this subclassification. Clinical features distinguishing the groups included a history of early life trauma and duration of symptoms (greater in IBS1), but not self-reported bowel habits, anxiety, depression, or medication use. Gut microbial composition correlated with structural measures of brain regions including sensory- and salience-related regions, and with a history of early life trauma.ConclusionsThe results confirm previous reports of gut microbiome-based IBS subgroups and identify for the first time brain structural alterations associated with these subgroups. They provide preliminary evidence for the involvement of specific microbes and their predicted metabolites in these correlations.
- Published
- 2017
20. The Role of the Microbiome in First Episode of Psychosis
- Author
-
Lucero Nuncio-Mora, Nuria Lanzagorta, Humberto Nicolini, Emmanuel Sarmiento, Galo Ortiz, Fernanda Sosa, and Alma Delia Genis-Mendoza
- Subjects
microbiome ,dysbiosis ,first episode of psychosis ,brain-gut-microbiome axis ,Biology (General) ,QH301-705.5 - Abstract
The relationship between the gut-brain-microbiome axis has gained great importance in the study of psychiatric disorders, as it may represent a new target for their treatment. To date, the available literature suggests that the microbiota may influence the pathophysiology of several diseases, including psychosis. The aim of this review is to summarize the clinical and preclinical studies that have evaluated the differences in microbiota as well as the metabolic consequences related to psychosis. Current data suggest that the genera Lactobacillus and Megasphaera are increased in schizophrenia (SZ), as well as alterations in the glutamate-glutamine-GABA cycle, serum levels of tryptophan, kynurenic acid (KYNA), and short-chain fatty acids (SCFAs). There are still very few studies on early-onset psychosis, thus more studies are needed to be able to propose targeted therapies for a point when the disease has just started or has not yet progressed.
- Published
- 2023
- Full Text
- View/download PDF
21. Continuous Ingestion of Lacticaseibacillus rhamnosus JB-1 during Chronic Stress Ensures Neurometabolic and Behavioural Stability in Rats.
- Author
-
Chudzik, Agata, Słowik, Tymoteusz, Kochalska, Katarzyna, Pankowska, Anna, Łazorczyk, Artur, Andres-Mach, Marta, Rola, Radosław, Stanisz, Greg J., and Orzyłowska, Anna
- Subjects
- *
PSYCHOLOGICAL stress , *NUCLEAR magnetic resonance spectroscopy , *GUT microbiome , *DIETARY supplements , *LABORATORY rats - Abstract
The intestinal microbiome composition and dietary supplementation with psychobiotics can result in neurochemical alterations in the brain, which are possible due to the presence of the brain–gut–microbiome axis. In the present study, magnetic resonance spectroscopy (MRS) and behavioural testing were used to evaluate whether treatment with Lacticaseibacillus rhamnosus JB-1 (JB-1) bacteria alters brain metabolites' levels and behaviour during continuous exposure to chronic stress. Twenty Wistar rats were subjected to eight weeks of a chronic unpredictable mild stress protocol. Simultaneously, half of them were fed with JB-1 bacteria, and the second half was given a daily placebo. Animals were examined at three-time points: before starting the stress protocol and after five and eight weeks of stress onset. In the elevated plus maze behavioural test the placebo group displayed increased anxiety expressed by almost complete avoidance of exploration, while the JB-1 dietary supplementation mitigated anxiety which resulted in a longer exploration time. Hippocampal MRS measurements demonstrated a significant decrease in glutamine + glutathione concentration in the placebo group compared to the JB-1 bacteria-supplemented group after five weeks of stress. With the progression of stress, the decrease of glutamate, glutathione, taurine, and macromolecular concentrations were observed in the placebo group as compared to baseline. The level of brain metabolites in the JB-1-supplemented rats were stable throughout the experiment, with only the taurine level decreasing between weeks five and eight of stress. These data indicated that the JB-1 bacteria diet might stabilize levels of stress-related neurometabolites in rat brain and could prevent the development of anxiety/depressive-like behaviour. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
22. Glycated milk protein fermented with Lactobacillus rhamnosus ameliorates the cognitive health of mice under mild-stress condition
- Author
-
Nam Su Oh, Jae Yeon Joung, Ji Young Lee, Jae Gwang Song, Sangnam Oh, Younghoon Kim, Hyung Wook Kim, and Sae Hun Kim
- Subjects
chronic mild stress ,brain-gut-microbiome axis ,casein ,glycation ,lactobacillus rhamnosus 4b15 ,Diseases of the digestive system. Gastroenterology ,RC799-869 - Abstract
This study aimed to investigate the effects of glycated milk casein (Gc) fermented with Lactobacillus rhamnosus 4B15 (FGc) on the intestinal microbiota and physiological and behavioral properties in mice under chronic stress. Mice were administered Gc or FGc for 10 weeks and then exposed to unpredictable chronic mild stress (UCMS) for 7 weeks. FGc administration restored alterations of gut microbiota induced by UCMS. Moreover, FGc significantly reduced the stress-induced increase in serum corticosterone and decrease in serotonin levels. Anxiety-like behaviors induced by UCMS were also significantly decreased in the FGc group. UCMS-induced dysregulation of gene and protein expression related to neuroendocrine function, neuronal development, and inflammation, and gut-blood-brain barrier function was controlled by FGc pre-treatment. These results strongly suggest the protective effects of FGc targeting of intestinal microbiota for abnormal brain activity, which is consistent with the view that FGc plays an important role in regulating stress-related gut-brain axis disorders.
- Published
- 2020
- Full Text
- View/download PDF
23. Cognitive function improvement after fecal microbiota transplantation in Alzheimer's dementia patient: a case report.
- Author
-
Park, Soo-Hyun, Lee, Jung Hwan, Shin, Jongbeom, Kim, Jun-Seob, Cha, Boram, Lee, Suhjoon, Kwon, Kye Sook, Shin, Yong Woon, and Choi, Seong Hye
- Subjects
- *
COGNITIVE ability , *FECAL microbiota transplantation , *ALZHEIMER'S disease , *ALZHEIMER'S patients , *COGNITIVE testing , *FECES - Abstract
After fecal microbiota transplantation (FMT) to treat Clostridioides difficile infection (CDI), cognitive improvement is noticeable, suggesting an essential association between the gut microbiome and neural function. Although it is known that the gut microbiome is linked with cognitive function, whether FMT may lead to cognitive improvement in patients with neurodegenerative disorders remains to be elucidated. We present the case of a 90-year-old woman with Alzheimer's dementia and severe CDI who underwent FMT. Cognitive function testing (Mini-Mental State Examination, Montreal Cognitive Assessment, and Clinical Dementia Rating assessment) was performed one month before FMT and one week and one month after FMT. We collected the patients' fecal samples before FMT and 3 weeks after FMT to compare the microbiota composition. The 16S rRNA gene amplicons were analyzed using the QIIME2 platform (version 2020.2) and the Phyloseq R package. The linear discriminant analysis effect size was performed to determine the taxonomic difference between pre- and post-FMT. Functional biomarker analysis using the Kruskal–Wallis H test was performed between the pre- and post-FMT. The cognitive function tests after FMT showed an improvement compared to the tests before the procedure. FMT changed the microbiota composition in recipient feces. We found that the genera were reported to be associated with cognitive function. In addition, short-chain fatty acids were found to be significantly different between before and after FMT. This finding suggests the presence of an association between the gut microbiome and cognitive function. Further, it emphasizes the need for clinical awareness regarding the effect of FMT on the brain-gut-microbiome axis and its potential as a therapy for patients with dementia. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
24. Repetitive transcranial direct current stimulation modulates the brain–gut–microbiome axis in obese rodents
- Author
-
Ziomber-Lisiak, Agata, Talaga-Ćwiertnia, Katarzyna, Sroka-Oleksiak, Agnieszka, Surówka, Artur D., Juszczak, Kajetan, and Szczerbowska-Boruchowska, Magdalena
- Published
- 2022
- Full Text
- View/download PDF
25. Differences in gut microbial composition correlate with regional brain volumes in irritable bowel syndrome
- Author
-
Jennifer S. Labus, Emily B. Hollister, Jonathan Jacobs, Kyleigh Kirbach, Numan Oezguen, Arpana Gupta, Jonathan Acosta, Ruth Ann Luna, Kjersti Aagaard, James Versalovic, Tor Savidge, Elaine Hsiao, Kirsten Tillisch, and Emeran A. Mayer
- Subjects
Brain-gut-microbiome axis ,Irritable bowel syndrome ,Metagenome ,Firmicutes ,Bacteroidetes ,Microbial ecology ,QR100-130 - Abstract
Abstract Background Preclinical and clinical evidence supports the concept of bidirectional brain-gut microbiome interactions. We aimed to determine if subgroups of irritable bowel syndrome (IBS) subjects can be identified based on differences in gut microbial composition, and if there are correlations between gut microbial measures and structural brain signatures in IBS. Methods Behavioral measures, stool samples, and structural brain images were collected from 29 adult IBS and 23 healthy control subjects (HCs). 16S ribosomal RNA (rRNA) gene sequencing was used to profile stool microbial communities, and various multivariate analysis approaches were used to quantitate microbial composition, abundance, and diversity. The metagenomic content of samples was inferred from 16S rRNA gene sequence data using Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt). T1-weighted brain images were acquired on a Siemens Allegra 3T scanner, and morphological measures were computed for 165 brain regions. Results Using unweighted Unifrac distances with hierarchical clustering on microbial data, samples were clustered into two IBS subgroups within the IBS population (IBS1 (n = 13) and HC-like IBS (n = 16)) and HCs (n = 23) (AUROC = 0.96, sensitivity 0.95, specificity 0.67). A Random Forest classifier provided further support for the differentiation of IBS1 and HC groups. Microbes belonging to the genera Faecalibacterium, Blautia, and Bacteroides contributed to this subclassification. Clinical features distinguishing the groups included a history of early life trauma and duration of symptoms (greater in IBS1), but not self-reported bowel habits, anxiety, depression, or medication use. Gut microbial composition correlated with structural measures of brain regions including sensory- and salience-related regions, and with a history of early life trauma. Conclusions The results confirm previous reports of gut microbiome-based IBS subgroups and identify for the first time brain structural alterations associated with these subgroups. They provide preliminary evidence for the involvement of specific microbes and their predicted metabolites in these correlations.
- Published
- 2017
- Full Text
- View/download PDF
26. Probiotics, Prebiotics and Postbiotics on Mitigation of Depression Symptoms: Modulation of the Brain–Gut–Microbiome Axis
- Author
-
Agata Chudzik, Anna Orzyłowska, Radosław Rola, and Greg J. Stanisz
- Subjects
microbiota ,brain–gut–microbiome axis ,depression ,probiotics ,prebiotics ,postbiotics ,Microbiology ,QR1-502 - Abstract
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.
- Published
- 2021
- Full Text
- View/download PDF
27. Cognitive behavioral therapy for irritable bowel syndrome induces bidirectional alterations in the brain-gut-microbiome axis associated with gastrointestinal symptom improvement
- Author
-
Kan Gao, Gregory D. Gudleski, Arpana Gupta, Bruce D. Naliboff, Ravi Bhatt, Jonathan P. Jacobs, Jennifer S. Labus, Jacob Brawer, Rebecca Firth, Kirsten Tillisch, Benjamin M. Ellingson, Venu Lagishetty, Emeran A. Mayer, and Jeffrey M. Lackner
- Subjects
Microbiology (medical) ,Oncology ,medicine.medical_specialty ,16S ,medicine.medical_treatment ,Clinical Trials and Supportive Activities ,Neuroimaging ,Biology ,Microbiology ,behavioral disciplines and activities ,Oral and gastrointestinal ,law.invention ,White matter ,Irritable Bowel Syndrome ,Microbial ecology ,Randomized controlled trial ,law ,Clinical Research ,Internal medicine ,RNA, Ribosomal, 16S ,Basal ganglia ,Brain-Gut Axis ,medicine ,Humans ,Microbiome ,Irritable bowel syndrome ,Default mode network ,Nutrition ,Ribosomal ,Cognitive Behavioral Therapy ,Ecology ,Research ,QR100-130 ,Neurosciences ,Outcome prediction ,medicine.disease ,Gastrointestinal Microbiome ,Cognitive behavioral therapy ,medicine.anatomical_structure ,Mental Health ,Medical Microbiology ,RNA ,Brain-gut-microbiome axis ,Digestive Diseases ,Mind and Body ,Biomarkers - Abstract
Background There is growing recognition that bidirectional signaling between the digestive tract and the brain contributes to irritable bowel syndrome (IBS). We recently showed in a large randomized controlled trial that cognitive behavioral therapy (CBT) reduces IBS symptom severity. This study investigated whether baseline brain and gut microbiome parameters predict CBT response and whether response is associated with changes in the brain-gut-microbiome (BGM) axis. Methods Eighty-four Rome III-diagnosed IBS patients receiving CBT were drawn from the Irritable Bowel Syndrome Outcome Study (IBSOS; ClinicalTrials.gov NCT00738920) for multimodal brain imaging and psychological assessments at baseline and after study completion. Fecal samples were collected at baseline and post-treatment from 34 CBT recipients for 16S rRNA gene sequencing, untargeted metabolomics, and measurement of short-chain fatty acids. Clinical measures, brain functional connectivity and microstructure, and microbiome features associated with CBT response were identified by multivariate linear and negative binomial models. Results At baseline, CBT responders had increased fecal serotonin levels, and increased Clostridiales and decreased Bacteroides compared to non-responders. A random forests classifier containing 11 microbial genera predicted CBT response with high accuracy (AUROC 0.96). Following treatment, CBT responders demonstrated reduced functional connectivity in regions of the sensorimotor, brainstem, salience, and default mode networks and changes in white matter in the basal ganglia and other structures. Brain changes correlated with microbiome shifts including Bacteroides expansion in responders. Conclusions Pre-treatment intestinal microbiota and serotonin levels were associated with CBT response, suggesting that peripheral signals from the microbiota can modulate central processes affected by CBT that generate abdominal symptoms in IBS. CBT response is characterized by co-correlated shifts in brain networks and gut microbiome that may reflect top-down effects of the brain on the microbiome during CBT.
- Published
- 2021
28. Shared Dysregulation of Homeostatic Brain-Body Pathways in Depression and Type 2 Diabetes.
- Author
-
Hoogendoorn, Claire, Roy, Juan, Gonzalez, Jeffrey, Hoogendoorn, Claire J, Roy, Juan F, and Gonzalez, Jeffrey S
- Subjects
BRAIN ,MENTAL depression ,ENDOCRINE glands ,HOMEOSTASIS ,HYPOTHALAMUS ,TYPE 2 diabetes ,RESEARCH funding - Abstract
Purpose Of Review: The purpose of this review is to provide an overview of shared dysregulation of the hypothalamic-pituitary-adrenal (HPA) and brain-gut-microbiome (BGM) axes associated with depression and type 2 diabetes (T2D). Clinical implications and future research are also discussed.Recent Findings: Both depression and T2D are associated with dysregulation of the HPA and BGM axes. These pathways regulate immune function, glucose metabolism, and sleep, which are altered in both illnesses. Dysregulation of homeostatic brain-body pathways may be positively influenced through different therapeutic actions, including psychotherapy, healthy eating, physical activity, sleep promotion, and certain anti-inflammatory or antidepressant medications. While the causal nature of the relationship between depression and T2D remains unclear, these conditions share dysregulation of homeostatic brain-body pathways that are central to mental and physical health. Better understanding of this dysregulation may provide opportunities for interventions that could benefit both conditions. Future research should examine the additive burden of depression and T2D on HPA and BGM dysregulation and better differentiate depression from emotional distress. [ABSTRACT FROM AUTHOR]- Published
- 2017
- Full Text
- View/download PDF
29. A psychology of the human brain-gut-microbiome axis.
- Author
-
Allen, Andrew P., Dinan, Timothy G., Clarke, Gerard, and Cryan, John F.
- Subjects
- *
BRAIN physiology , *PSYCHOBIOLOGY , *PSYCHOLOGICAL stress , *MENTAL depression , *COGNITION - Abstract
In recent years, we have seen increasing research within neuroscience and biopsychology on the interactions between the brain, the gastrointestinal tract, the bacteria within the gastrointestinal tract, and the bidirectional relationship between these systems: the brain-gut-microbiome axis. Although research has demonstrated that the gut microbiota can impact upon cognition and a variety of stress-related behaviours, including those relevant to anxiety and depression, we still do not know how this occurs. A deeper understanding of how psychological development as well as social and cultural factors impact upon the brain-gut-microbiome axis will contextualise the role of the axis in humans and inform psychological interventions that improve health within the brain-gut-microbiome axis. Interventions ostensibly aimed at ameliorating disorders in one part of the brain-gut-microbiome axis (e.g., psychotherapy for depression) may nonetheless impact upon other parts of the axis (e.g., microbiome composition and function), and functional gastrointestinal disorders such as irritable bowel syndrome represent a disorder of the axis, rather than an isolated problem either of psychology or of gastrointestinal function. The discipline of psychology needs to be cognisant of these interactions and can help to inform the future research agenda in this emerging field of research. In this review, we outline the role psychology has to play in understanding the brain-gut-microbiome axis, with a focus on human psychology and the use of research in laboratory animals to model human psychology. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
30. Glycated milk protein fermented with Lactobacillus rhamnosus ameliorates the cognitive health of mice under mild-stress condition
- Author
-
Hyung-Wook Kim, Ji-Young Lee, Sae Hun Kim, Sangnam Oh, Jae Gwang Song, Younghoon Kim, Jae Yeon Joung, and Nam Su Oh
- Subjects
0301 basic medicine ,Microbiology (medical) ,lactobacillus rhamnosus 4b15 ,Biology ,Microbiology ,casein ,Cognitive health ,03 medical and health sciences ,brain-gut-microbiome axis ,fluids and secretions ,0302 clinical medicine ,Lactobacillus rhamnosus ,Glycation ,Mild stress ,Casein ,Food science ,lcsh:RC799-869 ,Milk protein ,chronic mild stress ,Gastroenterology ,food and beverages ,biology.organism_classification ,030104 developmental biology ,Infectious Diseases ,glycation ,030211 gastroenterology & hepatology ,Fermentation ,lcsh:Diseases of the digestive system. Gastroenterology - Abstract
This study aimed to investigate the effects of glycated milk casein (Gc) fermented with Lactobacillus rhamnosus 4B15 (FGc) on the intestinal microbiota and physiological and behavioral properties in mice under chronic stress. Mice were administered Gc or FGc for 10 weeks and then exposed to unpredictable chronic mild stress (UCMS) for 7 weeks. FGc administration restored alterations of gut microbiota induced by UCMS. Moreover, FGc significantly reduced the stress-induced increase in serum corticosterone and decrease in serotonin levels. Anxiety-like behaviors induced by UCMS were also significantly decreased in the FGc group. UCMS-induced dysregulation of gene and protein expression related to neuroendocrine function, neuronal development, and inflammation, and gut-blood-brain barrier function was controlled by FGc pre-treatment. These results strongly suggest the protective effects of FGc targeting of intestinal microbiota for abnormal brain activity, which is consistent with the view that FGc plays an important role in regulating stress-related gut-brain axis disorders.
- Published
- 2020
31. Improvement in Uncontrolled Eating Behavior after Laparoscopic Sleeve Gastrectomy Is Associated with Alterations in the Brain-Gut-Microbiome Axis in Obese Women
- Author
-
Arpana Gupta, Anna Balioukova, Claudia P. Sanmiguel, Jean Stains, Priten Vora, Elizabeth Gallagher, Erik Dutson, Yijun Chen, Ravi Bhatt, Venu Lagishetty, Emeran A. Mayer, Jonathan P. Jacobs, Vadim Osadchiy, and Tien S. Dong
- Subjects
0301 basic medicine ,Adult ,medicine.medical_specialty ,obesity ,Adolescent ,bariatric surgery ,brain–gut–microbiome axis ,brain ,Health Behavior ,metabolite ,lcsh:TX341-641 ,Gastroenterology ,Article ,03 medical and health sciences ,Young Adult ,0302 clinical medicine ,Neuroimaging ,Weight loss ,Gastrectomy ,Internal medicine ,Surveys and Questionnaires ,Weight Loss ,Medicine ,Humans ,Microbiome ,skin and connective tissue diseases ,Yale Food Addiction Scale ,Nutrition and Dietetics ,business.industry ,Putamen ,Brain morphometry ,Middle Aged ,medicine.disease ,Obesity ,Magnetic Resonance Imaging ,Diet ,Gastrointestinal Microbiome ,030104 developmental biology ,Brain size ,Female ,Laparoscopy ,sense organs ,Food Addiction ,medicine.symptom ,business ,lcsh:Nutrition. Foods and food supply ,030217 neurology & neurosurgery ,Food Science - Abstract
Background: Bariatric surgery is proven to change eating behavior and cause sustained weight loss, yet the exact mechanisms underlying these changes are not clearly understood. We explore this in a novel way by examining how bariatric surgery affects the brain&ndash, gut&ndash, microbiome (BGM) axis. Methods: Patient demographics, serum, stool, eating behavior questionnaires, and brain magnetic resonance imaging (MRI) were collected before and 6 months after laparoscopic sleeve gastrectomy (LSG). Differences in eating behavior and brain morphology and resting-state functional connectivity in core reward regions were correlated with serum metabolite and 16S microbiome data. Results: LSG resulted in significant weight loss and improvement in maladaptive eating behaviors as measured by the Yale Food Addiction Scale (YFAS). Brain imaging showed a significant increase in brain volume of the putamen (p.adj <, 0.05) and amygdala (p.adj <, 0.05) after surgery. Resting-state connectivity between the precuneus and the putamen was significantly reduced after LSG (p.adj = 0.046). This change was associated with YFAS symptom count. Bacteroides, Ruminococcus, and Holdemanella were associated with reduced connectivity between these areas. Metabolomic profiles showed a positive correlation between this brain connection and a phosphatidylcholine metabolite. Conclusion: Bariatric surgery modulates brain networks that affect eating behavior, potentially through effects on the gut microbiota and its metabolites.
- Published
- 2020
32. Autism-related dietary preferences mediate autism-gut microbiome associations.
- Author
-
Yap, Chloe X., Henders, Anjali K., Alvares, Gail A., Wood, David L.A., Krause, Lutz, Tyson, Gene W., Restuadi, Restuadi, Wallace, Leanne, McLaren, Tiana, Hansell, Narelle K., Cleary, Dominique, Grove, Rachel, Hafekost, Claire, Harun, Alexis, Holdsworth, Helen, Jellett, Rachel, Khan, Feroza, Lawson, Lauren P., Leslie, Jodie, and Frenk, Mira Levis
- Subjects
- *
GUT microbiome , *AUTISM spectrum disorders , *AUTISTIC children , *FOOD consumption , *GENETIC models , *DIAGNOSIS , *PHENOTYPES - Abstract
There is increasing interest in the potential contribution of the gut microbiome to autism spectrum disorder (ASD). However, previous studies have been underpowered and have not been designed to address potential confounding factors in a comprehensive way. We performed a large autism stool metagenomics study (n = 247) based on participants from the Australian Autism Biobank and the Queensland Twin Adolescent Brain project. We found negligible direct associations between ASD diagnosis and the gut microbiome. Instead, our data support a model whereby ASD-related restricted interests are associated with less-diverse diet, and in turn reduced microbial taxonomic diversity and looser stool consistency. In contrast to ASD diagnosis, our dataset was well powered to detect microbiome associations with traits such as age, dietary intake, and stool consistency. Overall, microbiome differences in ASD may reflect dietary preferences that relate to diagnostic features, and we caution against claims that the microbiome has a driving role in ASD. [Display omitted] • Limited autism-microbiome associations from stool metagenomics of n = 247 children • Romboutsia timonensis was the only taxa associated with autism diagnosis • Autistic traits such as restricted interests are associated with less-diverse diet • Less-diverse diet, in turn, is associated with lower microbiome alpha-diversity Large autism stool metagenomics study finds limited direct autism associations, in contrast to strong relationships with dietary traits, stool consistency, and age, suggestive of a model whereby genetic and phenotypic measures of the autism spectrum promote a less-diverse diet that reduces microbiome diversity. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
33. Probiotics, Prebiotics and Postbiotics on Mitigation of Depression Symptoms: Modulation of the Brain–Gut–Microbiome Axis.
- Author
-
Chudzik, Agata, Orzyłowska, Anna, Rola, Radosław, and Stanisz, Greg J.
- Subjects
PREBIOTICS ,PROBIOTICS ,GUT microbiome ,MENTAL depression ,CENTRAL nervous system ,SYMPTOMS - Abstract
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. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
34. Psychological comorbidity in gastrointestinal diseases: Update on the brain-gut-microbiome axis.
- Author
-
Person, Hannibal and Keefer, Laurie
- Subjects
- *
COMORBIDITY , *GASTROINTESTINAL diseases , *ENTERIC nervous system , *INFLAMMATORY bowel diseases , *IRRITABLE colon , *GUT microbiome - Abstract
The high comorbidity of psychological disorders in both functional and organic gastrointestinal diseases suggests the intimate and complex link between the brain and the gut. Termed the brain-gut axis, this bidirectional communication between the central nervous system and enteric nervous system relies on immune, endocrine, neural, and metabolic pathways. There is increasing evidence that the gut microbiome is a key part of this system, and dysregulation of the brain-gut-microbiome axis (BGMA) has been implicated in disorders of brain-gut interaction, including irritable bowel syndrome, and in neuropsychiatric disorders, including depression, Alzheimer's disease, and autism spectrum disorder. Further, alterations in the gut microbiome have been implicated in the pathogenesis of organic gastrointestinal diseases, including inflammatory bowel disease. The BGMA is an attractive therapeutic target, as using prebiotics, probiotics, or postbiotics to modify the gut microbiome or mimic gut microbial signals could provide novel treatment options to address these debilitating diseases. However, despite significant advancements in our understanding of the BGMA, clinical data is lacking. In this article, we will review current understanding of the comorbidity of gastrointestinal diseases and psychological disorders. We will also review the current evidence supporting the key role of the BGMA in this pathology. Finally, we will discuss the clinical implications of the BGMA in the evaluation and management of psychological and gastrointestinal disorders. • Psychological concerns and disorders are highly comorbid with gastrointestinal diseases. • Disorders of gut-brain interaction are maintained by environmental, psychological, and biological factors. • The brain-gut-microbiome axis allows bidirectional communication between the central nervous system and the gut. • Dysregulation of the brain-gut-microbiome axis is implicated in a variety of psychological and gastrointestinal diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
35. A psychology of the human brain–gut–microbiome axis
- Author
-
Gerard Clarke, Timothy G. Dinan, Andrew P. Allen, and John F. Cryan
- Subjects
cognition ,0301 basic medicine ,Gerontology ,Psychotherapist ,Social Psychology ,mood ,Psychological intervention ,Gut flora ,Stress ,Article ,stress ,03 medical and health sciences ,Cognition ,Mood ,medicine ,Microbiome ,Irritable bowel syndrome ,biology ,brain‐gut‐microbiome axis ,medicine.disease ,biology.organism_classification ,030104 developmental biology ,Health ,Anxiety ,Brain-gut-microbiome axis ,medicine.symptom ,Gastrointestinal function ,Psychology - Abstract
In recent years, we have seen increasing research within neuroscience and biopsychology on the interactions between the brain, the gastrointestinal tract, the bacteria within the gastrointestinal tract, and the bidirectional relationship between these systems: the brain–gut–microbiome axis. Although research has demonstrated that the gut microbiota can impact upon cognition and a variety of stress‐related behaviours, including those relevant to anxiety and depression, we still do not know how this occurs. A deeper understanding of how psychological development as well as social and cultural factors impact upon the brain–gut–microbiome axis will contextualise the role of the axis in humans and inform psychological interventions that improve health within the brain–gut–microbiome axis. Interventions ostensibly aimed at ameliorating disorders in one part of the brain–gut–microbiome axis (e.g., psychotherapy for depression) may nonetheless impact upon other parts of the axis (e.g., microbiome composition and function), and functional gastrointestinal disorders such as irritable bowel syndrome represent a disorder of the axis, rather than an isolated problem either of psychology or of gastrointestinal function. The discipline of psychology needs to be cognisant of these interactions and can help to inform the future research agenda in this emerging field of research. In this review, we outline the role psychology has to play in understanding the brain–gut–microbiome axis, with a focus on human psychology and the use of research in laboratory animals to model human psychology.
- Published
- 2017
36. Natural products-based polypharmacological modulation of the peripheral immune system for the treatment of neuropsychiatric disorders.
- Author
-
Mok, Simon Wing-Fai, Wong, Vincent Kam-Wai, Lo, Hang-Hong, de Seabra Rodrigues Dias, Ivo Ricardo, Leung, Elaine Lai-Han, Law, Betty Yuen-Kwan, and Liu, Liang
- Subjects
- *
NEUROBEHAVIORAL disorders , *IMMUNE system , *IMMUNOREGULATION , *PATHOLOGY , *IMMUNOLOGIC diseases , *CENTRAL nervous system physiology - Abstract
Chronic inflammation of the central nervous system (CNS) is critical to the pathogenesis of neuropsychiatric disorders (NPDs) that affect the global population. Current therapeutics for NPDs are limited to relieving symptoms and induce many adverse effects. Therefore, the discovery of novel therapeutic agents from natural sources is urgently needed. Intriguingly, the immune responses of peripheral organs are closely linked through the molecular communication between resident and blood-borne cellular components, which shape the neuroinflammatory phenotypes of NPDs. Since the gut and spleen are the two largest immunological organs of the body, the brain–gut–microbiome and brain–spleen axes have been implicated in the connection between the CNS and the peripheral immune system. Accordingly, it has been proposed that the local CNS inflammation observed in NPDs is regulated via the manipulation of the systemic immune system by targeting the gut and spleen. Additionally, the complexity of the signalling network underlying the communication between the CNS and the systemic immune system suggests a strong potential for treating NPDs through a polypharmacological approach. The close association between systemic immunity and the homeostasis of the CNS points to the concept of repurposing interventions for systemic immune disorders to treat NPDs. Notably, natural products represent a promising source of such effective compounds due to both their pharmacological potency and safety. This review discusses the complex implications of dysregulated systemic immunity mediated by the brain–spleen and brain–gut–microbiome axes in NPDs, such as Alzheimer's disease, Parkinson's disease, schizophrenia and major depressive disorder. In addition, the potential of repurposing natural product-based bioactive compounds for treating NPDs via modulating systemic immune disorders is intensively discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
37. Brain-Gut Axis: Clinical Implications.
- Author
-
Khlevner J, Park Y, and Margolis KG
- Subjects
- Humans, Brain physiopathology, Gastrointestinal Microbiome physiology, Gastrointestinal Tract pathology, Gastrointestinal Tract physiopathology, Irritable Bowel Syndrome etiology
- Abstract
This article provides an overarching view of what is currently known about the physiology of the brain-gut axis in both health and disease and how these concepts apply to irritable bowel syndrome, the most common functional gastrointestinal disorder in pediatrics., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.