1
|
Lee SH, Han C, Shin C. IUPHAR Review: Microbiota-Gut-Brain Axis and its role in Neuropsychiatric Disorders. Pharmacol Res 2025; 216:107749. [PMID: 40306604 DOI: 10.1016/j.phrs.2025.107749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2025] [Revised: 04/20/2025] [Accepted: 04/25/2025] [Indexed: 05/02/2025]
Abstract
The human gut microbiome, composed of a vast array of microorganisms that have co-evolved with humans, is crucial for the development and function of brain systems. Research has consistently shown bidirectional communication between the gut and the brain through neuronal, endocrine, and immunological, and chemical pathways. Recent neuroscience studies have linked changes in the microbiome and microbial metabolites to various neuropsychiatric disorders such as autism, depression, anxiety, schizophrenia, eating disorders, and neurocognitive disorders. Novel metagenome-wide association studies have confirmed these microbiome variations in large samples and expanded our understanding of the interactions between human genes and the gut microbiome. The causal relationship between gut microbiota and neuropsychiatric disorders is being elucidated through the establishment of large cohort studies incorporating microbiome data and advanced statistical techniques. Ongoing animal and human studies focused on the microbiota-gut-brain axis are promising for developing new prevention and treatment strategies for neuropsychiatric conditions. The scope of these studies has broadened from microbiome-modulating therapies including prebiotics, probiotics, synbiotics and postbiotics to more extensive approaches such as fecal microbiota transplantation. Recent systematic reviews and meta-analyses have strengthened the evidence base for these innovative treatments. Despite extensive research over the past decade, many intriguing aspects still need to be elucidated regarding the role and therapeutic interventions of the microbiota-gut-brain axis in neuropsychiatric disorders.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Changsu Han
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Cheolmin Shin
- Department of Psychiatry, Korea University Ansan Hospital, Korea University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
2
|
Lee J, Menon NV, Truong HD, Lim CT. Dynamics of Spatial Organization of Bacterial Communities in a Tunable Flow Gut Microbiome-on-a-Chip. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2410258. [PMID: 40201941 DOI: 10.1002/smll.202410258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/10/2025] [Indexed: 04/10/2025]
Abstract
The human intestine, a biomechanically active organ, generates cyclic mechanical forces crucial for maintaining its health and functions. Yet, the physiological impact of these forces on gut microbiota dynamics remains largely unexplored. In this study, we investigate how cyclic intestinal motility influences the dynamics of gut microbial communities within a 3D gut-like structure (µGut). To enable the study, a tunable flow Gut Microbiome-on-a-Chip (tfGMoC) is developed that recapitulates the cyclic expansion and compression of intestinal motility while allowing high-magnification imaging of microbial communities within a 3D stratified, biomimetic gut epithelium. Using deep learning-based microbial analysis, it is found that hydrodynamic forces organize microbial communities by promoting distinct spatial exploration behaviors in microorganisms with varying motility characteristics. Empirical evidence demonstrates the impact of gut motility forces in maintaining a balanced gut microbial composition, enhancing both the diversity and stability of the community - key factors for a healthy microbiome. This study, leveraging the new tfGMoC platform, uncovers previously unknown effects of intestinal motility on modulating gut microbial behaviors and community organizations. This will be critical for a deeper understanding of host-microbiome interactions in the emerging field of microbiome therapeutics.
Collapse
Affiliation(s)
- Jeeyeon Lee
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, Singapore
| | | | - Hung Dong Truong
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chwee Teck Lim
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 117599, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| |
Collapse
|
3
|
Chen Y, Chen X, Lin S, Huang S, Li L, Hong M, Li J, Ma L, Ma J. Effects of psychological stress on inflammatory bowel disease via affecting the microbiota-gut-brain axis. Chin Med J (Engl) 2025; 138:664-677. [PMID: 39965932 PMCID: PMC11925421 DOI: 10.1097/cm9.0000000000003389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Indexed: 02/20/2025] Open
Abstract
ABSTRACT Inflammatory bowel disease (IBD) is an idiopathic intestinal inflammatory condition with chronic and relapsing manifestations and is characterized by a disturbance in the interplay between the intestinal microbiota, the gut, and the brain. The microbiota-gut-brain axis involves interactions among the nervous system, the neuroendocrine system, the gut microbiota, and the host immune system. Increasing published data indicate that psychological stress exacerbates the severity of IBD due to its negative effects on the microbiota-gut-brain axis, including alterations in the stress response of the hypothalamic-pituitary-adrenal (HPA) axis, the balance between the sympathetic nervous system and vagus nerves, the homeostasis of the intestinal flora and metabolites, and normal intestinal immunity and permeability. Although the current evidence is insufficient, psychotropic agents, psychotherapies, and interventions targeting the microbiota-gut-brain axis show the potential to improve symptoms and quality of life in IBD patients. Therefore, further studies that translate recent findings into therapeutic approaches that improve both physical and psychological well-being are needed.
Collapse
Affiliation(s)
- Yuhan Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Xiaofen Chen
- Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Suqin Lin
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shengjun Huang
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Lijuan Li
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Mingzhi Hong
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Medical College, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jianzhou Li
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People's Hospital of Xining, Xining, Qinghai 810003, China
| | - Lili Ma
- Department of Gastroenterology and Hepatology, Qinghai Provincial People's Hospital, Xining, Qinghai 810007, China
| | - Juan Ma
- Department of Gastroenterology and Hepatology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Department of Diagnosis and Treatment Center of High Altitude Digestive Disease, The Second People's Hospital of Xining, Xining, Qinghai 810003, China
| |
Collapse
|
4
|
Liu M, Fan G, Meng L, Yang K, Liu H. New perspectives on microbiome-dependent gut-brain pathways for the treatment of depression with gastrointestinal symptoms: from bench to bedside. J Zhejiang Univ Sci B 2025; 26:1-25. [PMID: 39428337 PMCID: PMC11735910 DOI: 10.1631/jzus.b2300343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 08/29/2023] [Indexed: 10/22/2024]
Abstract
Patients with depression are more likely to have chronic gastrointestinal (GI) symptoms than the general population, but such symptoms are considered only somatic symptoms of depression and lack special attention. There is a chronic lack of appropriate diagnosis and effective treatment for patients with depression accompanied by GI symptoms, and studying the association between depression and GI disorders (GIDs) is extremely important for clinical management. There is growing evidence that depression is closely related to the microbiota present in the GI tract, and the microbiota-gut-brain axis (MGBA) is creating a new perspective on the association between depression and GIDs. Identifying and treating GIDs would provide a key opportunity to prevent episodes of depression and may also improve the outcome of refractory depression. Current studies on depression and the microbially related gut-brain axis (GBA) lack a focus on GI function. In this review, we combine preclinical and clinical evidence to summarize the roles of the microbially regulated GBA in emotions and GI function, and summarize potential therapeutic strategies to provide a reference for the study of the pathomechanism and treatment of depression in combination with GI symptoms.
Collapse
Affiliation(s)
- Menglin Liu
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Brain Disease Regional Diagnosis and Treatment Center, Zhengzhou 450000, China
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
| | - Genhao Fan
- Tianjin University of Traditional Chinese Medicine, Tianjin 301600, China
- The First Affiliated Hospital of Zhengzhou University, Department of Geriatrics, Zhengzhou 450052, China
| | - Lingkai Meng
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Kuo Yang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China
| | - Huayi Liu
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin 300131, China.
| |
Collapse
|
5
|
Saurabh NK, Khan MM, Kirabo A. A Future Avenue of Treatment Ulcerative Colitis Targeting Macrophage Polarization: A Phytochemical Application. CROHN'S & COLITIS 360 2024; 6:otae070. [PMID: 39668979 PMCID: PMC11635166 DOI: 10.1093/crocol/otae070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Indexed: 12/14/2024] Open
Abstract
Background Ulcerative colitis (UC) is a prevalent inflammatory bowel disease primarily impacting the mucosa of the colon. It is characterized by recurring and incurable symptoms and causes immense suffering and significant economic burden due to limited treatment options. Typical symptoms of UC include diarrhea, alterations in bowel patterns, bleeding from the rectum, rectal pain or urgency, anemia, and tiredness. Therefore, developing novel and effective treatment strategies for UC is imperative. Purpose This review aimed to explain how macrophage polarization contributes to UC development and compiled information on natural compounds with promising therapeutic potential that can target the macrophage phenotype and shed light on its potential mode of action. Results The phenotypic alteration of macrophages profoundly affects the development of UC, and these cells are essential for preserving intestinal immunological homeostasis. Evidence from research suggests that one effective method for UC prevention and therapy is to guide macrophage polarization toward the M2 phenotype. Phytochemicals, which are compounds extracted from plants, possess a wide array of biological activities. For example: Ginsenoside Rg1 emerges as a crucial regulator of macrophage polarization, promoting the M2 phenotype while inhibiting the M1 phenotype. Notably, their low toxicity and high effectiveness render them promising candidates for therapeutic interventions. These compounds have demonstrated encouraging protective effects against inflammation in the colon. Conclusions Exploring phytochemicals as a therapeutic avenue targeting macrophage polarization presents an innovative approach to treating UC.
Collapse
Affiliation(s)
- Nishant Kumar Saurabh
- Division of Molecular Biology, National Institute of Cancer Prevention & Research (ICMR-NICPR), I-7, Sector-39, Noida 201301, India
| | - Mohd Mabood Khan
- Department of Medicine, Robinson Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232-6602, USA
| | - Annet Kirabo
- Department of Medicine, Robinson Research Building, Vanderbilt University Medical Centre, Nashville, TN 37232-6602, USA
| |
Collapse
|
6
|
Beurel E, Nemeroff CB. Early Life Adversity, Microbiome, and Inflammatory Responses. Biomolecules 2024; 14:802. [PMID: 39062516 PMCID: PMC11275239 DOI: 10.3390/biom14070802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
Early life adversity has a profound impact on physical and mental health. Because the central nervous and immune systems are not fully mature at birth and continue to mature during the postnatal period, a bidirectional interaction between the central nervous system and the immune system has been hypothesized, with traumatic stressors during childhood being pivotal in priming individuals for later adult psychopathology. Similarly, the microbiome, which regulates both neurodevelopment and immune function, also matures during childhood, rendering this interaction between the brain and the immune system even more complex. In this review, we provide evidence for the role of the immune response and the microbiome in the deleterious effects of early life adversity, both in humans and rodent models.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Charles B. Nemeroff
- Department of Psychiatry and Behavioral Sciences, Mulva Clinic for Neurosciences, University of Texas (UT) Dell Medical School, Austin, TX 78712, USA
- Mulva Clinic for Neurosciences, UT Austin Dell Medical School, Austin, TX 78712, USA
| |
Collapse
|
7
|
Watai K, Suda W, Kurokawa R, Sekiya K, Hayashi H, Iwata M, Nagayama K, Nakamura Y, Hamada Y, Kamide Y, Fukutomi Y, Nakabayashi T, Tanaka K, Kamita M, Taniguchi M, Hattori M. Metagenomic gut microbiome analysis of Japanese patients with multiple chemical sensitivity/idiopathic environmental intolerance. BMC Microbiol 2024; 24:84. [PMID: 38468206 PMCID: PMC10926566 DOI: 10.1186/s12866-024-03239-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 02/26/2024] [Indexed: 03/13/2024] Open
Abstract
BACKGROUND Although the pathology of multiple chemical sensitivity (MCS) is unknown, the central nervous system is reportedly involved. The gut microbiota is important in modifying central nervous system diseases. However, the relationship between the gut microbiota and MCS remains unclear. This study aimed to identify gut microbiota variations associated with MCS using shotgun metagenomic sequencing of fecal samples. METHODS We prospectively recruited 30 consecutive Japanese female patients with MCS and analyzed their gut microbiomes using shotgun metagenomic sequencing. The data were compared with metagenomic data obtained from 24 age- and sex-matched Japanese healthy controls (HC). RESULTS We observed no significant difference in alpha and beta diversity of the gut microbiota between the MCS patients and HC. Focusing on the important changes in the literatures, at the genus level, Streptococcus, Veillonella, and Akkermansia were significantly more abundant in MCS patients than in HC (p < 0.01, p < 0.01, p = 0.01, respectively, fold change = 4.03, 1.53, 2.86, respectively). At the species level, Akkermansia muciniphila was significantly more abundant (p = 0.02, fold change = 3.3) and Faecalibacterium prausnitzii significantly less abundant in MCS patients than in HC (p = 0.03, fold change = 0.53). Functional analysis revealed that xylene and dioxin degradation pathways were significantly enriched (p < 0.01, p = 0.01, respectively, fold change = 1.54, 1.46, respectively), whereas pathways involved in amino acid metabolism and synthesis were significantly depleted in MCS (p < 0.01, fold change = 0.96). Pathways related to antimicrobial resistance, including the two-component system and cationic antimicrobial peptide resistance, were also significantly enriched in MCS (p < 0.01, p < 0.01, respectively, fold change = 1.1, 1.2, respectively). CONCLUSIONS The gut microbiota of patients with MCS shows dysbiosis and alterations in bacterial functions related to exogenous chemicals and amino acid metabolism and synthesis. These findings may contribute to the further development of treatment for MCS. TRIAL REGISTRATION This study was registered with the University Hospital Medical Information Clinical Trials Registry as UMIN000031031. The date of first trial registration: 28/01/2018.
Collapse
Affiliation(s)
- Kentaro Watai
- Center for Immunology and Allergy, Shonan Kamakura General Hospital, 1370-1 Okamoto, Kamakura, Kanagawa, 247-8533, Japan.
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan.
| | - Wataru Suda
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Rina Kurokawa
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Kiyoshi Sekiya
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Hiroaki Hayashi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Maki Iwata
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Kisako Nagayama
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuto Nakamura
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuto Hamada
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yosuke Kamide
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Yuma Fukutomi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | | | - Kosei Tanaka
- H.U. Group Research Institute G.K., Akiruno, Tokyo, Japan
| | | | - Masami Taniguchi
- Clinical Research Center for Allergy and Rheumatology, NHO Sagamihara National Hospital, Sagamihara, Kanagawa, Japan
| | - Masahira Hattori
- Laboratory for Microbiome Sciences, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
- Graduate School of Advanced Science and Engineering, Waseda University, Tokyo, Japan
| |
Collapse
|
8
|
Shen D, Chang L, Su F, Huang S, Xu H, Si Y, Wang F, Xue Y. The gut microbiome modulates the susceptibility to traumatic stress in a sex-dependent manner. J Neurosci Res 2024; 102:e25315. [PMID: 38439584 DOI: 10.1002/jnr.25315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 01/30/2024] [Accepted: 02/19/2024] [Indexed: 03/06/2024]
Abstract
Post-traumatic stress disorder (PTSD), a psychological condition triggered by exposure to extreme or chronic stressful events, exhibits a sex bias in incidence and clinical manifestations. Emerging research implicates the gut microbiome in the pathogenesis of PTSD and its roles in stress susceptibility. However, it is unclear whether differential gut microbiota contribute to PTSD susceptibility in male and female rats. Here, we utilized the single prolonged stress animal model and employed unsupervised machine learning to classify stressed animals into stress-susceptible subgroups and stress-resilient subgroups. Subsequently, using 16S V3-V4 rDNA sequencing, we investigated the differential gut microbiota alterations between susceptible and resilient individuals in male and female rats. Our findings revealed distinct changes in gut microbiota composition between the sexes at different taxonomic levels. Furthermore, the abundance of Parabacteroides was lower in rats that underwent SPS modeling compared to the control group. In addition, the abundance of Tenericutes in the stress-susceptible subgroup was higher than that in the control group and stress-resilient subgroup, suggesting that Tenericutes may be able to characterize stress susceptibility. What is particularly interesting here is that Cyanobacteria may be particularly associated with anti-anxiety effects in male rats. This study underscores sex-specific variations in gut microbiota composition in response to stress and sex differences should be taken into account when using macrobiotics for neuropsychiatric treatment, highlighting potential targets for PTSD therapeutic interventions.
Collapse
Affiliation(s)
- Dan Shen
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Liang Chang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Feng Su
- College of Future Technology, Peking University, Beijing, China
| | - Shihao Huang
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Hubo Xu
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Yue Si
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| | - Feng Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China
| | - Yanxue Xue
- National Institute on Drug Dependence and Beijing Key Laboratory of Drug Dependence, Peking University, Beijing, China
| |
Collapse
|
9
|
Joung JY, Song JG, Lee B, Kim HW, Oh NS. Preventive effect of peptides derived from fermented milk on chronic stress-induced brain damage and intestinal dysfunction in mice. J Dairy Sci 2023; 106:8287-8298. [PMID: 37690713 DOI: 10.3168/jds.2023-23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 07/24/2023] [Indexed: 09/12/2023]
Abstract
This study investigated the preventive effects of peptides derived from milk fermented with the probiotic strain Lactobacillus gasseri 505 (505) against stress-related brain damage and anxiety-like behavior. The peptides MKPWIQPKTKVIPYVRYL (Pep14) and VYQHQKAMKPWIQPKTKVIPYVRYL (Pep21), which exhibit high antioxidant and anti-inflammatory activities, were administered to stressed mice. The results showed that the stress mechanism in the gut-brain axis was regulated by pretreatment with both peptides, leading to inhibition of neurodevelopment and neuroinflammation through the hypothalamic-pituitary-adrenal (HPA) axis, based on the expression of related mRNA and proteins. The expression of colonic inflammation-related mRNA and proteins was also reduced. Moreover, anxiety-like behavior was significantly reduced in mice treated with Pep14 and Pep21. These results indicate that the bioactive peptides Pep14 and Pep21, derived from milk fermented with 505, may prevent stress-induced brain damage and anxiety-like behavior via regulation of the HPA axis.
Collapse
Affiliation(s)
- Jae Yeon Joung
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Korea
| | - Jae Gwang Song
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, 05006, Republic of Korea
| | - Bomi Lee
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, 05006, Republic of Korea
| | - Hyung Wook Kim
- Department of Bio-integrated Science and Technology, College of Life Sciences, Sejong University, Seoul, 05006, Republic of Korea.
| | - Nam Su Oh
- Department of Food and Biotechnology, Korea University, Sejong, 30019, Korea.
| |
Collapse
|
10
|
Nass BYS, Dibbets P, Markus CR. The Impact of Psychotrauma and Emotional Stress Vulnerability on Physical and Mental Functioning of Patients with Inflammatory Bowel Disease. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2023; 20:6976. [PMID: 37947534 PMCID: PMC10648781 DOI: 10.3390/ijerph20216976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 10/06/2023] [Accepted: 10/22/2023] [Indexed: 11/12/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic health condition thought to be influenced by personal life experiences and emotional stress sensitivity (neuroticism). In the present study, we examined the impact of cumulative trauma experiences and trait neuroticism (as a measure for emotional stress vulnerability) on physical and mental functioning of n = 211 patients diagnosed with IBD (112 Crohn's disease, 99 ulcerative colitis). All patients were assessed for self-reported trauma histories, emotional stress vulnerability, clinical disease activity, functional gastrointestinal (GI) symptoms, and quality of life. Results showed that patients with severe IBD activity have endured significantly more interpersonal trauma and victimization than those with quiescent IBD. Moreover, cumulative trauma was found to exert an indirect (neuroticism-mediated) effect on patients' symptom complexity, with trauma and neuroticism conjointly explaining 16-21% of the variance in gastrointestinal and 35% of the variance in mental symptoms. Upon correction for condition (using a small group of available controls, n = 51), the predictive capacity of trauma and neuroticism increased further, with both predictors now explaining 31% of the somatic-and almost 50% of the mental symptom heterogeneity. In terms of trauma type, victimization (domestic violence and intimate abuse) proved the best predictor of cross-sample symptom variability and the only trauma profile with a consistent direct and indirect (neuroticism-mediated) effect on patients' mental (QoL) and physical fitness. Results are consistent with the growing body of evidence linking experiential vulnerability factors (trauma and neuroticism) and associated feelings of personal ineffectiveness, helplessness, and uncontrollability to interindividual differences in (GI) disease activity and quality of life.
Collapse
Affiliation(s)
- Boukje Yentl Sundari Nass
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
- Dr. Rath Health Foundation, 6422 RG Heerlen, The Netherlands
| | - Pauline Dibbets
- Clinical Psychological Science, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - C. Rob Markus
- Department of Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
11
|
Dargenio VN, Dargenio C, Castellaneta S, De Giacomo A, Laguardia M, Schettini F, Francavilla R, Cristofori F. Intestinal Barrier Dysfunction and Microbiota-Gut-Brain Axis: Possible Implications in the Pathogenesis and Treatment of Autism Spectrum Disorder. Nutrients 2023; 15:1620. [PMID: 37049461 PMCID: PMC10096948 DOI: 10.3390/nu15071620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/21/2023] [Accepted: 03/22/2023] [Indexed: 03/29/2023] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental disorder with multifactorial etiology, characterized by impairment in two main functional areas: (1) communication and social interactions, and (2) skills, interests and activities. ASD patients often suffer from gastrointestinal symptoms associated with dysbiotic states and a "leaky gut." A key role in the pathogenesis of ASD has been attributed to the gut microbiota, as it influences central nervous system development and neuropsychological and gastrointestinal homeostasis through the microbiota-gut-brain axis. A state of dysbiosis with a reduction in the Bacteroidetes/Firmicutes ratio and Bacteroidetes level and other imbalances is common in ASD. In recent decades, many authors have tried to study and identify the microbial signature of ASD through in vivo and ex vivo studies. In this regard, the advent of metabolomics has also been of great help. Based on these data, several therapeutic strategies, primarily the use of probiotics, are investigated to improve the symptoms of ASD through the modulation of the microbiota. However, although the results are promising, the heterogeneity of the studies precludes concrete evidence. The aim of this review is to explore the role of intestinal barrier dysfunction, the gut-brain axis and microbiota alterations in ASD and the possible role of probiotic supplementation in these patients.
Collapse
Affiliation(s)
- Vanessa Nadia Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Costantino Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Stefania Castellaneta
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Andrea De Giacomo
- Child Neuropsychiatry Unit, Department of Translational Biomedicine and Neuroscience, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Marianna Laguardia
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Federico Schettini
- Neonatology and Neonatal Intensive Care Unit (NICU), University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy
| |
Collapse
|
12
|
Zhou W, Zhan L, Xu H, Zhang L. Structural Alteration of Gut Microbiota During the Amelioration of Chronic Psychological Stress-Aggravated Diabetes-Associated Cognitive Decline by a Traditional Chinese Herbal Formula, ZiBu PiYin Recipe. J Alzheimers Dis 2022; 90:1465-1483. [PMID: 36278351 DOI: 10.3233/jad-220692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Chronic psychological stress (PS) hinders the treatment of diabetes-associated cognitive decline (DACD). However, the impact of chronic PS on the risk of developing DACD remains unclear. There is growing evidence that gut flora interventions are promising targets for treating stress-related diseases. OBJECTIVE We examined whether chronic PS triggers or exacerbates the onset of DACD in rats and aimed to elucidate whether ZiBuPiYin recipe (ZBPYR) prevents and treats chronic PS-aggravated DACD by dynamically maintaining the components of the gut microbiota. METHODS We performed chronic PS (restraint, rotation, and congestion) on ZDF rats to establish a model. Cognitive function was evaluated by behavioral experiments, and activation of the hypothalamic-pituitary-adrenal axis was detected by ELISA. Weekly feces from rats were collected for 16 S RNA sequencing. RESULTS We found that chronic PS promoted cognitive abnormalities and exacerbated DACD phenotypes. Additionally, chronic PS altered intestinal flora diversity, dynamically elevating the abundance of Alistipes and Coprococcus; enriching Module 1 (Dorea, Blautia, Ruminococcus) and Module 48 (Blautia); and inhibiting Module 20 (Lactobacillus, SMB53), and Module 42 (Akkermansia). ZBPYR significantly alleviated hyperglycemia and cognitive impairment in chronic PS-aggravated DACD rats and dynamically reduced the abundance of Alistipes and Coprococcus; significantly enriched Module 3 (Ruminococcus) and Module 45 (Lactobacillus, Coprococcus, SMB53); and suppressed Module 2 (Lactobacillus), Module 16 (Turicibacter, Trichococcus, Lactobacillus, 02d06, Clostridium), Module 23 (Bifidobacterium), and Module 43 (Clostridium). CONCLUSION ZBPYR might prevent and treat chronic PS-aggravated DACD by dynamically regulating Lactobacillus, Alistipes, and Coprococcus.
Collapse
Affiliation(s)
- Wen Zhou
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Libin Zhan
- Centre for Innovative Engineering Technology in Traditional Chinese Medicine, Liaoning University of Traditional Chinese Medicine, Shenyang, China
| | - Huiying Xu
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lijing Zhang
- School of Traditional Chinese Medicine & School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Li Y, Duan Z. Updates in interaction of gastroesophageal reflux disease and extragastroesophageal digestive diseases. Expert Rev Gastroenterol Hepatol 2022; 16:1053-1063. [PMID: 35860994 DOI: 10.1080/17474124.2022.2056018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Gastroesophageal reflux disease (GERD) is one of the common chronic diseases with prevalence increasing in the last decades. Because of its prevalence and chronicity, GERD affects the quality of life and increases health-care costs. Gastroesophageal diseases leading to GERD have been thoroughly studied, while extragastroesophageal digestive diseases (EGEDDs) may coexist with GERD and affect the occurrence and persistence of GERD symptoms and therapeutic effect. AREAS COVERED In this review, we aim to summarize the EGEDDs correlated with GERD and explore the potential mechanisms of this interaction. EXPERT OPINION Individuals with troublesome GERD symptoms may have some common gastroesophageal etiologies, but EGEDDs may also overlap and impact on the progression of GERD, which are often ignored in clinic. The lesions in the small intestine, colon, and hepatobiliary tract as well as functional bowel disorders had positive or negative associations with GERD through potential mechanisms. These diseases aggravate GERD symptoms, increase the esophageal acid burden, cause esophageal hypersensitivity, and finally affect the response to therapy in GERD patients. Therefore, it is necessary to clear the interaction between GERD and EGEDDs and their mechanisms.
Collapse
Affiliation(s)
- Yanqiu Li
- Second Gastroenterology Department, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhijun Duan
- Second Gastroenterology Department, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
14
|
Bixby M, Gennings C, Malecki KMC, Sethi AK, Safdar N, Peppard PE, Eggers S. Individual Nutrition Is Associated with Altered Gut Microbiome Composition for Adults with Food Insecurity. Nutrients 2022; 14:3407. [PMID: 36014913 PMCID: PMC9416073 DOI: 10.3390/nu14163407] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/13/2022] [Accepted: 08/14/2022] [Indexed: 11/17/2022] Open
Abstract
Diet is widely recognized as a key contributor to human gut microbiome composition and function. However, overall nutrition can be difficult to compare across a population with varying diets. Moreover, the role of food security in the relationship with overall nutrition and the gut microbiome is unclear. This study aims to investigate the association between personalized nutrition scores, variation in the adult gut microbiome, and modification by food insecurity. The data originate from the Survey of the Health of Wisconsin and the Wisconsin Microbiome Study. Individual nutrition scores were assessed using My Nutrition Index (MNI), calculated using data from food frequency questionnaires, and additional health history and demographic surveys. Food security and covariate data were measured through self-reported questionnaires. The gut microbiome was assessed using 16S amplicon sequencing of DNA extracted from stool samples. Associations, adjusted for confounding and interaction by food security, were estimated using Weighted Quantile Sum (WQS) regression models with Random Subset and Repeated Holdout extensions (WQSRSRH), with bacterial taxa used as components in the weighted index. Of 643 participants, the average MNI was 66.5 (SD = 31.9), and 22.8% of participants were food insecure. Increased MNI was significantly associated with altered gut microbial composition (β = 2.56, 95% CI = 0.52−4.61), with Ruminococcus, Oscillospira, and Blautia among the most heavily weighted of the 21 genera associated with the MNI score. In the stratified interaction WQSRSRH models, the bacterial taxa most heavily weighted in the association with MNI differed by food security, but the level of association between MNI and the gut microbiome was not significantly different. More bacterial genera are important in the association with higher nutrition scores for people with food insecurity versus food security, including Streptococcus, Parabacteroides Faecalibacterium, and Desulfovibrio. Individual nutrition scores are associated with differences in adult gut microbiome composition. The bacterial taxa most associated with nutrition vary by level of food security. While further investigation is needed, results showed a higher nutrition score was associated with a wider range of bacterial taxa for food insecure vs. secure, suggesting nutritional quality in food insecure individuals is important in maintaining health and reducing disparities.
Collapse
Affiliation(s)
- Moira Bixby
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Chris Gennings
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Kristen M. C. Malecki
- Population Health Sciences, University of Wisconsin School of Medicine and Public Health, 610 Walnut St., WARF 707, Madison, WI 53726, USA
| | - Ajay K. Sethi
- Population Health Sciences, University of Wisconsin School of Medicine and Public Health, 610 Walnut St., WARF 707, Madison, WI 53726, USA
| | - Nasia Safdar
- Department of Medicine, Division of Infectious Disease, School of Medicine and Public Health, University of Wisconsin-Madison, UW Med. Fndtn. Centennial Bldg., 1685 Highland Ave, Madison, WI 53705, USA
- William S. Middleton Veterans Affairs Medical Center, 2500 Overlook Terrace, Madison, WI 53705, USA
| | - Paul E. Peppard
- Population Health Sciences, University of Wisconsin School of Medicine and Public Health, 610 Walnut St., WARF 707, Madison, WI 53726, USA
| | - Shoshannah Eggers
- Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| |
Collapse
|
15
|
Mallaret G, Lashermes A, Meleine M, Boudieu L, Barbier J, Aissouni Y, Gelot A, Chassaing B, Gewirtz AT, Ardid D, Carvalho FA. Involvement of toll-like receptor 5 in mouse model of colonic hypersensitivity induced by neonatal maternal separation. World J Gastroenterol 2022; 28:3903-3916. [PMID: 36157543 PMCID: PMC9367235 DOI: 10.3748/wjg.v28.i29.3903] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/09/2022] [Accepted: 07/06/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Chronic abdominal pain is the most common cause for gastroenterology consultation and is frequently associated with functional gastrointestinal disorders including irritable bowel syndrome and inflammatory bowel disease. These disorders present similar brain/gut/microbiota trialogue alterations, associated with abnormal intestinal permeability, intestinal dysbiosis and colonic hypersensitivity (CHS). Intestinal dysbiosis can alter colon homeostasis leading to abnormal activation of the innate immunity that promotes CHS, perhaps involving the toll-like receptors (TLRs), which play a central role in innate immunity.
AIM To understand the mechanisms between early life event paradigm on intestinal permeability, fecal microbiota composition and CHS development in mice with TLRs expression in colonocytes.
METHODS Maternal separation model (NMS) CHS model, which mimics deleterious events in childhood that can induce a wide range of chronic disorders during adulthood were used. Colonic sensitivity of NMS mice was evaluated by colorectal distension (CRD) coupled with intracolonic pressure variation (IPV) measurement. Fecal microbiota composition was analyzed by 16S rRNA sequencing from weaning to CRD periods. TLR mRNA expression was evaluated in colonocytes. Additionally, the effect of acute intrarectal instillation of the TLR5 agonist flagellin (FliC) on CHS in adult naive wildtype mice was analyzed.
RESULTS Around 50% of NMS mice exhibited increased intestinal permeability and CHS associated with intestinal dysbiosis, characterized by a significant decrease of species richness, an alteration of the core fecal microbiota and a specific increased relative abundance of flagellated bacteria. Only TLR5 mRNA expression was increased in colonocytes of NMS mice with CHS. Acute intrarectal instillation of FliC induced transient increase of IPV, reflecting transient CHS appearance.
CONCLUSION Altogether, these data suggest a pathophysiological continuum between intestinal dysbiosis and CHS, with a role for TLR5.
Collapse
Affiliation(s)
- Geoffroy Mallaret
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Amandine Lashermes
- Department of Microbiology, Université Paris-Saclay, National Research Institute for Agriculture, Food and the Environment, AgroParisTech, Micalis Institute, Jouy-en-Josas 78350, France
| | - Mathieu Meleine
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Ludivine Boudieu
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Julie Barbier
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Youssef Aissouni
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Agathe Gelot
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Benoit Chassaing
- Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, INSERM U1016, CNRS UMR 8104, Université Paris Cité, Paris 75014, France
| | - Andrew T Gewirtz
- Center for Inflammation, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA30033, United States
| | - Denis Ardid
- Department of Pharmacology, UMR 1107 NeuroDol, University of Clermont Auvergne, Clermont-Ferrand 63000, France
| | - Frederic Antonio Carvalho
- Department of Pharmacology, INSERM 1107 NeuroDOL/University of Clermont Auvergne, Clermont-Ferrand 63000, France
| |
Collapse
|
16
|
Jiang FR, Hang L, Zhou Y, Feng Y, Yuan JY. Estrogen-gut microbiota interactions and irritable bowel syndrome. Shijie Huaren Xiaohua Zazhi 2022; 30:511-520. [DOI: 10.11569/wcjd.v30.i12.511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a common functional gastrointestinal disorder with a complex pathogenesis that has a serious impact on the quality of life of patients. Abnormal visceral sensation, disordered gut motility, dysregulated immunity, and damaged intestinal barrier are thought to be involved in the pathogenesis of IBS. Female predisposition to IBS strongly suggests that sex hormones such as estrogen are involved in the development of IBS. In addition, dysbiosis of the intestinal flora is closely related to IBS. The interaction between estrogen and gut microbiota in IBS has not been fully elucidated. This review summarizes and evaluates the progress of related studies. Based on the new findings and shortcomings of current studies, we discuss the directions and issues that need to be resolved in future research.
Collapse
Affiliation(s)
- Feng-Ru Jiang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Lu Hang
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Yan Zhou
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Ya Feng
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| | - Jian-Ye Yuan
- Institute of Digestive Diseases, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China
| |
Collapse
|
17
|
SATO MASAMICHI, KUDO TAKAHIRO, ARAI NOBUYASU, KYODO REIKO, HOSOI KENJI, SAKAGUCHI KEITA, IKUSE TAMAKI, JIMBO KEISUKE, OHTSUKA YOSHIKAZU, SHIMIZU TOSHIAKI. Evaluating Small Intestinal Motility in a Rat Model of Adolescent Irritable Bowel Syndrome. JUNTENDO IJI ZASSHI = JUNTENDO MEDICAL JOURNAL 2022; 68:271-281. [PMID: 39021725 PMCID: PMC11250020 DOI: 10.14789/jmj.jmj21-0050-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 02/10/2022] [Indexed: 07/20/2024]
Abstract
Objectives The correlation between altered small intestinal motility and irritable bowel syndrome is not well evaluated. This study aimed to assess the small intestinal and colonic transits in an adolescent irritable bowel syndrome rat model with restraint stress and determine the role of small intestinal motility in the irritable bowel syndrome pathophysiology. Materials Restraint stress was utilized to prepare adolescent irritable bowel syndrome rat models that were evaluated for clinical signs, including stool frequency and diarrhea. The small intestinal motility and transit rate were also evaluated. Methods The amounts of mRNA encoding corticotropin-releasing hormone, mast cell, and serotonin (5-Hydroxytryptamine) receptor 3a were quantified using real-time polymerase chain reaction; the 5-Hydroxytryptamine expression was evaluated using immunostaining. Results Restraint stress significantly increased the number of fecal pellet outputs, stool water content, and small intestinal motility in the adolescent irritable bowel syndrome rat models. There was no difference in real-time polymerase chain reaction results; however, immunostaining analysis revealed that 5-Hydroxytryptamine expression in the small intestine was significantly increased in the adolescent irritable bowel syndrome rat models. Conclusions In the rat model of adolescent irritable bowel syndrome with restraint stress, we observed an increase in small intestinal and colonic motility. In the small intestine, enhanced 5-Hydroxytryptamine secretion in the distal portion may be involved in increasing the small intestinal motility. Although the present study focused on 5-Hydroxytryptamine, further investigation of other factors that regulate intestinal peristalsis may lead to the establishment of more effective treatment methods for adolescent irritable bowel syndrome.
Collapse
Affiliation(s)
| | - TAKAHIRO KUDO
- Corresponding author: Takahiro Kudo (ORCID: 0000-0003-4708-8049), Department of Pediatrics, Juntendo University Faculty of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan, TEL: +81-3-3813-3111 FAX: +81-3-5800-0216 E-mail:
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Shobeiri P, Kalantari A, Teixeira AL, Rezaei N. Shedding light on biological sex differences and microbiota-gut-brain axis: a comprehensive review of its roles in neuropsychiatric disorders. Biol Sex Differ 2022; 13:12. [PMID: 35337376 PMCID: PMC8949832 DOI: 10.1186/s13293-022-00422-6] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 03/14/2022] [Indexed: 12/15/2022] Open
Abstract
Women and men are suggested to have differences in vulnerability to neuropsychiatric disorders, including major depressive disorder (MDD), generalized anxiety disorder (GAD), schizophrenia, eating disorders, including anorexia nervosa, and bulimia nervosa, neurodevelopmental disorders, such as autism spectrum disorder (ASD), and neurodegenerative disorders including Alzheimer's disease, Parkinson's disease. Genetic factors and sex hormones are apparently the main mediators of these differences. Recent evidence uncovers that reciprocal interactions between sex-related features (e.g., sex hormones and sex differences in the brain) and gut microbiota could play a role in the development of neuropsychiatric disorders via influencing the gut-brain axis. It is increasingly evident that sex-microbiota-brain interactions take part in the occurrence of neurologic and psychiatric disorders. Accordingly, integrating the existing evidence might help to enlighten the fundamental roles of these interactions in the pathogenesis of neuropsychiatric disorders. In addition, an increased understanding of the biological sex differences on the microbiota-brain may lead to advances in the treatment of neuropsychiatric disorders and increase the potential for precision medicine. This review discusses the effects of sex differences on the brain and gut microbiota and the putative underlying mechanisms of action. Additionally, we discuss the consequences of interactions between sex differences and gut microbiota on the emergence of particular neuropsychiatric disorders.
Collapse
Affiliation(s)
- Parnian Shobeiri
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran
| | - Amirali Kalantari
- School of Medicine, Tehran University of Medical Sciences (TUMS), Children's Medical Center Hospital, Dr. Qarib St., Keshavarz Blvd, 14194, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Antônio L Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Nima Rezaei
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Dr. Gharib St, Keshavarz Blvd, Tehran, Iran.
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Yue Q, Cai M, Xiao B, Zhan Q, Zeng C. The Microbiota-Gut-Brain Axis and Epilepsy. Cell Mol Neurobiol 2022; 42:439-453. [PMID: 34279746 PMCID: PMC11441249 DOI: 10.1007/s10571-021-01130-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022]
Abstract
Honoured as the second genome in humans, the gut microbiota is involved in a constellation of physiological and pathological processes, including those related to the central nervous system. The communication between the gut microbiota and the brain is realized by a complex bidirectional connection, known as the "microbiota-gut-brain axis", via neuroendocrine, immunological, and direct neural mechanisms. Recent studies indicate that gut dysfunction/dysbiosis is presumably involved in the pathogenesis of and susceptibility to epilepsy. In addition, the reconstruction of the intestinal microbiome through, for example, faecal microbiota transplantation, probiotic intervention, and a ketogenic diet, has exhibited beneficial effects on drug-resistant epilepsy. The purposes of this review are to provide a brief overview of the microbiota-gut-brain axis and to synthesize what is known about the involvement of the gut microbiota in the pathogenesis and treatment of epilepsy, to bring new insight into the pathophysiology of epilepsy and to present a preliminary discussion of novel therapeutic options for epilepsy based on the gut microbiota.
Collapse
Affiliation(s)
- Qiang Yue
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Mingfei Cai
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, China
| | - Bo Xiao
- Department of Neurology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China
| | - Qiong Zhan
- Department of Neurology, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011, China.
| | - Chang Zeng
- Health Management Center, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, China.
| |
Collapse
|
20
|
MEJÍA-GRANADOS DM, VILLASANA-SALAZAR B, COAN AC, RIZZI L, BALTHAZAR MLF, GODOI ABD, CANTO AMD, ROSA DCD, SILVA LS, TACLA RDR, DAMASCENO A, DONATTI A, AVELAR WM, SOUSA A, LOPES-CENDES I. Gut microbiome in neuropsychiatric disorders. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:192-207. [PMID: 35352757 DOI: 10.1590/0004-282x-anp-2021-0052] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/10/2021] [Indexed: 12/20/2022]
Abstract
ABSTRACT Background: Neuropsychiatric disorders are a significant cause of death and disability worldwide. The mechanisms underlying these disorders include a constellation of structural, infectious, immunological, metabolic, and genetic etiologies. Advances in next-generation sequencing techniques have demonstrated that the composition of the enteric microbiome is dynamic and plays a pivotal role in host homeostasis and several diseases. The enteric microbiome acts as a key mediator in neuronal signaling via metabolic, neuroimmune, and neuroendocrine pathways. Objective: In this review, we aim to present and discuss the most current knowledge regarding the putative influence of the gut microbiome in neuropsychiatric disorders. Methods: We examined some of the preclinical and clinical evidence and therapeutic strategies associated with the manipulation of the gut microbiome. Results: targeted taxa were described and grouped from major studies to each disease. Conclusions: Understanding the complexity of these ecological interactions and their association with susceptibility and progression of acute and chronic disorders could lead to novel diagnostic biomarkers based on molecular targets. Moreover, research on the microbiome can also improve some emerging treatment choices, such as fecal transplantation, personalized probiotics, and dietary interventions, which could be used to reduce the impact of specific neuropsychiatric disorders. We expect that this knowledge will help physicians caring for patients with neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Ana Carolina COAN
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | - Liara RIZZI
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | | | | | - Amanda Morato do CANTO
- Universidade de Campinas, Brazil; Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil
| | - Douglas Cescon da ROSA
- Universidade de Campinas, Brazil; Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil
| | - Lucas Scárdua SILVA
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | | | - Alfredo DAMASCENO
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | - Amanda DONATTI
- Universidade de Campinas, Brazil; Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil
| | - Wagner Mauad AVELAR
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | - Alessandro SOUSA
- Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil; Universidade de Campinas, Brazil
| | - Iscia LOPES-CENDES
- Universidade de Campinas, Brazil; Instituto Brasileiro de Neurociências e Neurotecnologia, Brazil
| |
Collapse
|
21
|
Diaz J, Reese AT. Possibilities and limits for using the gut microbiome to improve captive animal health. Anim Microbiome 2021; 3:89. [PMID: 34965885 PMCID: PMC8715647 DOI: 10.1186/s42523-021-00155-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 12/18/2021] [Indexed: 12/13/2022] Open
Abstract
Because of its potential to modulate host health, the gut microbiome of captive animals has become an increasingly important area of research. In this paper, we review the current literature comparing the gut microbiomes of wild and captive animals, as well as experiments tracking the microbiome when animals are moved between wild and captive environments. As a whole, these studies report highly idiosyncratic results with significant differences in the effect of captivity on the gut microbiome between host species. While a few studies have analyzed the functional capacity of captive microbiomes, there has been little research directly addressing the health consequences of captive microbiomes. Therefore, the current body of literature cannot broadly answer what costs, if any, arise from having a captive microbiome in captivity. Addressing this outstanding question will be critical to determining whether it is worth pursuing microbial manipulations as a conservation tool. To stimulate the next wave of research which can tie the captive microbiome to functional and health impacts, we outline a wide range of tools that can be used to manipulate the microbiome in captivity and suggest a variety of methods for measuring the impact of such manipulation preceding therapeutic use. Altogether, we caution researchers against generalizing results between host species given the variability in gut community responses to captivity and highlight the need to understand what role the gut microbiome plays in captive animal health before putting microbiome manipulations broadly into practice.
Collapse
Affiliation(s)
- Jessica Diaz
- Section of Ecology, Behavior, and Evolution, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Aspen T Reese
- Section of Ecology, Behavior, and Evolution, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Katasonov AB. [Gut microbiome as a therapeutic target in the treatment of depression and anxiety]. Zh Nevrol Psikhiatr Im S S Korsakova 2021; 121:129-135. [PMID: 34932298 DOI: 10.17116/jnevro2021121111129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
There is a bi-directional connection between the gut microbiome and the brain. Changes in the composition of the microbiome affect emotions, behavior, and the stress response involved in the pathogenesis of depression. Depression and anxiety are often associated with dysbiosis and inflammatory bowel disease. Dysbiosis enhances stress response and low-grade systemic inflammation, and vice versa. This vicious circle may be responsible for the formation of depression. Antidepressants therapy should be accompanied by the elimination of dysbiosis. For these purposes diet, prebiotics, probiotics and faecal microbiota transplantation can be used. The advantages and disadvantages of each method are considered. The manipulation of microbiome composition has been shown to have great therapeutic potential in the treatment of depression and anxiety.
Collapse
|
23
|
Park ES, Freeborn J, Venna VR, Roos S, Rhoads JM, Liu Y. Lactobacillus reuteri effects on maternal separation stress in newborn mice. Pediatr Res 2021; 90:980-988. [PMID: 33531679 DOI: 10.1038/s41390-021-01374-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/23/2020] [Accepted: 12/30/2020] [Indexed: 01/30/2023]
Abstract
BACKGROUND Probiotic Lactobacillus reuteri DSM 17938 (LR 17938) is beneficial to infants with colic. To understand its mechanism of action, we assessed ultrasonic vocalizations (USV) and brain pain/stress genes in newborn mice exposed to maternal separation stress. METHODS Pups were exposed to unpredictable maternal separation (MSU or SEP) or MSU combined with unpredictable maternal stress (MSU + MSUS or S + S), from postnatal days 5 to 14. USV calls and pain/stress/neuroinflammation-related genes in the brain were analyzed. RESULTS We defined 10 different neonatal call patterns, none of which increased after MSU. Stress reduced overall USV calls. Orally feeding LR 17938 also did not change USV calls after MSU. However, LR 17938 markedly increased vocalizations in mice allowed to stay with their dams. Even though LR 17938 did not change MSU-related calls, LR 17938 modulated brain genes related to stress and pain. Up-regulated genes following LR 17938 treatment were opioid peptides, kappa-opioid receptor 1 genes, and CD200, important in anti-inflammatory signaling. LR 17938 down-regulated CCR2 transcripts, a chemokine receptor, in the stressed neonatal brain. CONCLUSIONS USV calls in newborn mice are interpreted as "physiological calls" instead of "cries." Feeding LR 17938 after MSU did not change USV calls but modulated cerebral genes favoring pain and stress reduction and anti-inflammatory signaling. IMPACT We defined mouse ultrasonic vocalization (USV) call patterns in this study, which will be important in guiding future studies in other mouse strains. Newborn mice with maternal separation stress have reduced USVs, compared to newborn mice without stress, indicating USV calls may represent "physiological calling" instead of "crying." Oral feeding of probiotic Lactobacillus reuteri DSM 17938 raised the number of calls when newborn mice continued to suckle on their dams, but not when mice were under stress. The probiotic bacteria had a dampening effect on monocyte activation and on epinephrine and glutamate-related stress gene expression in the mouse brain.
Collapse
Affiliation(s)
- Evelyn S Park
- Departments of Pediatrics at McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jasmin Freeborn
- Departments of Pediatrics at McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Venugopal Reddy Venna
- Departments of Neurology at McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia AB, Stockholm, Sweden
| | - J Marc Rhoads
- Departments of Pediatrics at McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yuying Liu
- Departments of Pediatrics at McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
24
|
Lopizzo N, Marizzoni M, Begni V, Mazzelli M, Provasi S, Borruso L, Riva MA, Cattaneo A. Social isolation in adolescence and long-term changes in the gut microbiota composition and in the hippocampal inflammation: Implications for psychiatric disorders - Dirk Hellhammer Award Paper 2021. Psychoneuroendocrinology 2021; 133:105416. [PMID: 34593267 DOI: 10.1016/j.psyneuen.2021.105416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 09/08/2021] [Indexed: 01/03/2023]
Abstract
Exposure to early adverse experiences induces persistent changes in physiological, emotional and behavioural functions predisposing the individual to an enhanced vulnerability to develop different disorders during lifespan. The adverse outcomes depend upon the timing of the stressful experiences, and in this contest, adolescence represents a key sensitive period for brain development. Among the biological systems involved, gut microbiota has recently been proposed to act on the interplay between the stress response, brain functions and immune system, through the gut-brain axis communication. In the current study we aimed to evaluate, in a preclinical model, changes over time in the microbiota community structure in physiological condition and in response to stress during adolescence. We also aimed to correlate the microbiota composition to the inflammatory status in brain. We used the preclinical model of social deprivation in rats during adolescence, based on the lack of all social contacts, for four weeks after weaning, followed by re-socialization until adulthood. We collected fecal samples at different post-natal days to investigate the short- and long-lasting effects of social isolation on gut microbiota composition and we collected brain areas (dorsal and ventral hippocampus) samples at killing to measure a panel of inflammatory and microglia activation markers. 16 S metataxonomic sequencing analysis revealed that microbial changes were influenced by age in both isolated and controls rats, regardless of sex, whereas social isolation impacted the microbial composition in a sex-dependent manner. A multivariate analysis showed that social isolation induced short-term gut microbiota alterations in females but not in males. We also identified several stress-related genera associated with social isolation condition. In brain areas we found a specific inflammatory pattern, in dorsal and ventral hippocampus, that significantly correlated with gut microbiota composition. Overall, in this study we reported a novel sex-specific association between gut microbiota composition and inflammatory response related to social isolation paradigm during adolescence, suggesting that stressful experiences during this sensitive period could have a long-lasting impact on the development of different biological systems that could in turn influence the vulnerability to develop mental disorders later in life.
Collapse
Affiliation(s)
- Nicola Lopizzo
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Moira Marizzoni
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Laboratory of Neuroimaging and Alzheimer's Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Veronica Begni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Monica Mazzelli
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Stefania Provasi
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Luigimaria Borruso
- Faculty of Science and Technology, Free University of Bozen/Bolzano, piazza Università 5, 39100 Bolzano, Italy
| | - Marco Andrea Riva
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Annamaria Cattaneo
- Laboratory of Biological Psychiatry, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy.
| |
Collapse
|
25
|
Chidambaram SB, Essa MM, Rathipriya AG, Bishir M, Ray B, Mahalakshmi AM, Tousif AH, Sakharkar MK, Kashyap RS, Friedland RP, Monaghan TM. Gut dysbiosis, defective autophagy and altered immune responses in neurodegenerative diseases: Tales of a vicious cycle. Pharmacol Ther 2021; 231:107988. [PMID: 34536490 DOI: 10.1016/j.pharmthera.2021.107988] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/16/2021] [Accepted: 08/25/2021] [Indexed: 02/08/2023]
Abstract
The human microbiota comprises trillions of symbiotic microorganisms and is involved in regulating gastrointestinal (GI), immune, nervous system and metabolic homeostasis. Recent observations suggest a bidirectional communication between the gut microbiota and the brain via immune, circulatory and neural pathways, termed the Gut-Brain Axis (GBA). Alterations in gut microbiota composition, such as seen with an increased number of pathobionts and a decreased number of symbionts, termed gut dysbiosis or microbial intestinal dysbiosis, plays a prominent role in the pathogenesis of central nervous system (CNS)-related disorders. Clinical reports confirm that GI symptoms often precede neurological symptoms several years before the development of neurodegenerative diseases (NDDs). Pathologically, gut dysbiosis disrupts the integrity of the intestinal barrier leading to ingress of pathobionts and toxic metabolites into the systemic circulation causing GBA dysregulation. Subsequently, chronic neuroinflammation via dysregulated immune activation triggers the accumulation of neurotoxic misfolded proteins in and around CNS cells resulting in neuronal death. Emerging evidence links gut dysbiosis to the aggravation and/or spread of proteinopathies from the peripheral nervous system to the CNS and defective autophagy-mediated proteinopathies. This review summarizes the current understanding of the role of gut microbiota in NDDs, and highlights a vicious cycle of gut dysbiosis, immune-mediated chronic neuroinflammation, impaired autophagy and proteinopathies, which contributes to the development of neurodegeneration in Alzheimer's disease, Parkinson's disease, Huntington's disease, multiple sclerosis, amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We also discuss novel therapeutic strategies targeting the modulation of gut dysbiosis through prebiotics, probiotics, synbiotics or dietary interventions, and faecal microbial transplantation (FMT) in the management of NDDs.
Collapse
Affiliation(s)
- Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India.
| | - Musthafa Mohamed Essa
- Department of Food Science and Nutrition, CAMS, Sultan Qaboos University, Muscat 123, Oman; Ageing and Dementia Research Group, Sultan Qaboos University, Muscat 123, Oman; Biomedical Sciences Department, University of Pacific, Sacramento, CA, USA.
| | - A G Rathipriya
- Food and Brain Research Foundation, Chennai 600 094, Tamil Nadu, India
| | - Muhammed Bishir
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - A H Tousif
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru 570015, KA, India; Centre for Experimental Pharmacology and Toxicology (CPT), JSS Academy of Higher Education & Research, Mysuru 570015, KA, India
| | - Meena K Sakharkar
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Rajpal Singh Kashyap
- Research Centre, Dr G. M. Taori Central India Institute of Medical Sciences (CIIMS), Nagpur, Maharashtra, India
| | - Robert P Friedland
- Department of Neurology, University of Louisville, Louisville, KY 40292, USA
| | - Tanya M Monaghan
- NIHR Nottingham Biomedical Research Centre, University of Nottingham, Nottingham NG7 2UH, UK; Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK.
| |
Collapse
|
26
|
Tang HY, Jiang AJ, Wang XY, Wang H, Guan YY, Li F, Shen GM. Uncovering the pathophysiology of irritable bowel syndrome by exploring the gut-brain axis: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1187. [PMID: 34430628 PMCID: PMC8350700 DOI: 10.21037/atm-21-2779] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022]
Abstract
Objective To improve the pathophysiological understanding of irritable bowel syndrome (IBS) by exploring the gut-brain axis. Background Disorders of gut-brain interaction (DGBIs) are gastrointestinal (GI) disorders in which alterations in bowel functions occur. IBS, which is one of the most studied DGBIs, is linked with abdominal distress or pain without obvious structural or biochemical anomalies. Methods The etiology of IBS has not been clearly described but is known to be multifactorial, involving GI motility changes, post-infectious reactivity, visceral hypersensitivity, gut-brain interactions, microbiota dysbiosis, small intestinal bacterial overgrowth, food sensitivity, carbohydrate malabsorption, and intestinal inflammation. Conclusions One of the main features of IBS is the occurrence of structural and functional disruptions in the gut-brain axis, which alter reflective and perceptual nervous system reactions. Herein, we provide a brief summary of this topic. Furthermore, we discuss animal models, which are important in the study of IBS, especially as it is linked with stressors. These animal models cannot fully represent the human disease but serve as important tools for understanding this complicated disorder. In the future, technologies, such as organ-on-a-chip models and metabolomics, will provide novel information regarding the pathophysiology of IBS, which will play an important role in treatment development. Finally, we take a brief glance at how acupuncture treatments may hold potential for patients with IBS.
Collapse
Affiliation(s)
- He-Yong Tang
- Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Ai-Juan Jiang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Xi-Yang Wang
- Graduate School of Anhui University of Chinese Medicine, Hefei, China
| | - Hao Wang
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Yuan-Yuan Guan
- Department of Acupuncture and Moxibustion, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Fei Li
- Department of Rehabilitation, Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, China
| | - Guo-Ming Shen
- School of Integrated Traditional Chinese and Western Medicine, Anhui University of Traditional Chinese Medicine, Hefei, China
| |
Collapse
|
27
|
Bell RL, Withers GS, Kuypers FA, Stehr W, Bhargava A. Stress and corticotropin releasing factor (CRF) promote necrotizing enterocolitis in a formula-fed neonatal rat model. PLoS One 2021; 16:e0246412. [PMID: 34111125 PMCID: PMC8191945 DOI: 10.1371/journal.pone.0246412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/25/2021] [Indexed: 11/18/2022] Open
Abstract
The etiology of necrotizing enterocolitis (NEC) is not known. Alterations in gut microbiome, mucosal barrier function, immune cell activation, and blood flow are characterized events in its development, with stress as a contributing factor. The hormone corticotropin-releasing factor (CRF) is a key mediator of stress responses and influences these aforementioned processes. CRF signaling is modulated by NEC's main risk factors of prematurity and formula feeding. Using an established neonatal rat model of NEC, we tested hypotheses that: (i) increased CRF levels-as seen during stress-promote NEC in formula-fed (FF) newborn rats, and (ii) antagonism of CRF action ameliorates NEC. Newborn pups were formula-fed to initiate gut inflammation and randomized to: no stress, no stress with subcutaneous CRF administration, stress (acute hypoxia followed by cold exposure-NEC model), or stress after pretreatment with the CRF peptide antagonist Astressin. Dam-fed unstressed and stressed littermates served as controls. NEC incidence and severity in the terminal ileum were determined using a histologic scoring system. Changes in CRF, CRF receptor (CRFRs), and toll-like receptor 4 (TLR4) expression levels were determined by immunofluorescence and immunoblotting, respectively. Stress exposure in FF neonates resulted in 40.0% NEC incidence, whereas exogenous CRF administration resulted in 51.7% NEC incidence compared to 8.7% in FF non-stressed neonates (p<0.001). Astressin prevented development of NEC in FF-stressed neonates (7.7% vs. 40.0%; p = 0.003). CRF and CRFR immunoreactivity increased in the ileum of neonates with NEC compared to dam-fed controls or FF unstressed pups. Immunoblotting confirmed increased TLR4 protein levels in FF stressed (NEC model) animals vs. controls, and Astressin treatment restored TLR4 to control levels. Peripheral CRF may serve as specific pharmacologic target for the prevention and treatment of NEC.
Collapse
MESH Headings
- Animals
- Female
- Rats
- Animals, Newborn
- Corticotropin-Releasing Hormone/metabolism
- Disease Models, Animal
- Enterocolitis, Necrotizing/metabolism
- Enterocolitis, Necrotizing/pathology
- Enterocolitis, Necrotizing/prevention & control
- Enterocolitis, Necrotizing/etiology
- Ileum/metabolism
- Ileum/pathology
- Peptide Fragments/metabolism
- Rats, Sprague-Dawley
- Receptors, Corticotropin-Releasing Hormone/metabolism
- Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
- Stress, Physiological
- Toll-Like Receptor 4/metabolism
Collapse
Affiliation(s)
- Robert L. Bell
- East Bay Surgery Program, Department of Surgery, University of California San Francisco (UCSF) Benioff Children’s Hospital, Oakland, California, United States of America
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- The Permanente Medical Group, Department of Surgery, Walnut Creek, California, United States of America
| | - Ginger S. Withers
- Department of Biology, Whitman College, Walla Walla, Washington, United States of America
| | - Frans A. Kuypers
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- UCSF Benioff Children’s Hospital Oakland, Oakland, California, United States of America
| | - Wolfgang Stehr
- Children’s Hospital Oakland Research Institute, Oakland, California, United States of America
- UCSF Benioff Children’s Hospital Oakland, Oakland, California, United States of America
| | - Aditi Bhargava
- Department of Obstetrics and Gynecology, Center for Reproductive Sciences, University of California San Francisco, San Francisco, California, United States of America
| |
Collapse
|
28
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 165] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
29
|
Ting CH, Chen CY. Gut symptoms in the depressed elderly: The interactions between emotion and gastrointestinal neuroendocrinology. J Chin Med Assoc 2021; 84:455-456. [PMID: 33742997 DOI: 10.1097/jcma.0000000000000521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Ching-Heng Ting
- Department of Pathology, MacKay Memorial Hospital, Taipei, Taiwan, ROC
- Department of Medicine, MacKay Medical College, New Taipei City, Taiwan, ROC
| | - Chih-Yen Chen
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Faculty of Medicine and Institute of Emergency and Critical Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei, Taiwan, ROC
| |
Collapse
|
30
|
Scassellati C, Marizzoni M, Cattane N, Lopizzo N, Mombelli E, Riva MA, Cattaneo A. The Complex Molecular Picture of Gut and Oral Microbiota-Brain-Depression System: What We Know and What We Need to Know. Front Psychiatry 2021; 12:722335. [PMID: 34819883 PMCID: PMC8607517 DOI: 10.3389/fpsyt.2021.722335] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/30/2021] [Indexed: 12/12/2022] Open
Abstract
Major depressive disorder (MDD) is a complex mental disorder where the neurochemical, neuroendocrine, immune, and metabolic systems are impaired. The microbiota-gut-brain axis is a bidirectional network where the central and enteric nervous systems are linked through the same endocrine, immune, neural, and metabolic routes dysregulated in MDD. Thus, gut-brain axis abnormalities in MDD patients may, at least in part, account for the symptomatic features associated with MDD. Recent investigations have suggested that the oral microbiome also plays a key role in this complex molecular picture of relationships. As on one hand there is a lot of what we know and on the other hand little of what we still need to know, we structured this review focusing, in the first place, on putting all pieces of this complex puzzle together, underlying the endocrine, immune, oxidative stress, neural, microbial neurotransmitters, and metabolites molecular interactions and systems lying at the base of gut microbiota (GM)-brain-depression interphase. Then, we focused on promising but still under-explored areas of research strictly linked to the GM and potentially involved in MDD development: (i) the interconnection of GM with oral microbiome that can influence the neuroinflammation-related processes and (ii) gut phageome (bacteria-infecting viruses). As conclusions and future directions, we discussed potentiality but also pitfalls, roadblocks, and the gaps to be bridged in this exciting field of research. By the development of a broader knowledge of the biology associated with MDD, with the inclusion of the gut/oral microbiome, we can accelerate the growth toward a better global health based on precision medicine.
Collapse
Affiliation(s)
- Catia Scassellati
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Moira Marizzoni
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Laboratory of Alzheimer's Neuroimaging and Epidemiology, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Nadia Cattane
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Nicola Lopizzo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Elisa Mombelli
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Marco Andrea Riva
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Annamaria Cattaneo
- Biological Psychiatry Unit, Istituto di Recupero e Cura a Carattere Scientifico (IRCCS) Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| |
Collapse
|
31
|
Daugé V, Philippe C, Mariadassou M, Rué O, Martin JC, Rossignol MN, Dourmap N, Svilar L, Tourniaire F, Monnoye M, Jardet D, Bangratz M, Holowacz S, Rabot S, Naudon L. A Probiotic Mixture Induces Anxiolytic- and Antidepressive-Like Effects in Fischer and Maternally Deprived Long Evans Rats. Front Behav Neurosci 2020; 14:581296. [PMID: 33312120 PMCID: PMC7708897 DOI: 10.3389/fnbeh.2020.581296] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/20/2020] [Indexed: 12/22/2022] Open
Abstract
A role of the gut microbiota in psychiatric disorders is supported by a growing body of literature. The effects of a probiotic mixture of four bacterial strains were studied in two models of anxiety and depression, naturally stress-sensitive Fischer rats and Long Evans rats subjected to maternal deprivation. Rats chronically received either the probiotic mixture (1.109 CFU/day) or the vehicle. Anxiety- and depressive-like behaviors were evaluated in several tests. Brain monoamine levels and gut RNA expression of tight junction proteins (Tjp) and inflammatory markers were quantified. The gut microbiota was analyzed in feces by 16S rRNA gene sequencing. Untargeted metabolite analysis reflecting primary metabolism was performed in the cecal content and in serum. Fischer rats treated with the probiotic mixture manifested a decrease in anxiety-like behaviors, in the immobility time in the forced swimming test, as well as in levels of dopamine and its major metabolites, and those of serotonin metabolites in the hippocampus and striatum. In maternally deprived Long Evans rats treated with the probiotic mixture, the number of entries into the central area in the open-field test was increased, reflecting an anxiolytic effect. The probiotic mixture increased Tjp1 and decreased Ifnγ mRNA levels in the ileum of maternally deprived rats. In both models, probiotic supplementation changed the proportions of several Operational Taxonomic Units (OTU) in the gut microbiota, and the levels of certain cecal and serum metabolites were correlated with behavioral changes. Chronic administration of the tested probiotic mixture can therefore beneficially affect anxiety- and depressive-like behaviors in rats, possibly owing to changes in the levels of certain metabolites, such as 21-deoxycortisol, and changes in brain monoamines.
Collapse
Affiliation(s)
- Valérie Daugé
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| | - Catherine Philippe
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Mahendra Mariadassou
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France
| | - Olivier Rué
- Université Paris-Saclay, INRAE, BioinfOmics, MIGALE Bioinformatics Facility, Jouy-en-Josas, France
| | | | | | - Nathalie Dourmap
- UNIROUEN, UFR Médecine-Pharmacie, Inserm U 1245 Team 4, Rouen, France
| | | | | | - Magali Monnoye
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Deborah Jardet
- Université Paris-Saclay, INRAE, MaIAGE, Jouy-en-Josas, France
| | | | | | - Sylvie Rabot
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France
| | - Laurent Naudon
- Université Paris-Saclay, INRAE, AgroParisTech, CNRS, Micalis Institute, Jouy-en-Josas, France
| |
Collapse
|
32
|
Yuan F, Tan W, Ren H, Yan L, Wang Y, Luo H. The Effects of Short-Chain Fatty Acids on Rat Colonic Hypermotility Induced by Water Avoidance Stress. Drug Des Devel Ther 2020; 14:4671-4684. [PMID: 33173277 PMCID: PMC7646441 DOI: 10.2147/dddt.s246619] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Short-chain fatty acids (SCFAs) have been reported to play an important role in regulating gastrointestinal motility. The aim of this study is to investigate the possible role of SCFAs in water avoidance stress-induced colonic hypermotility. METHODS A rat IBS model was established by water avoidance stress (WAS). Intestinal motility was assessed by fecal pellets expulsion. The fecal SCFA level was detected using gas chromatography-mass spectrometry (GC-MS). Western blotting was performed to assess the expression of SCFAs receptors. To determine the role of SCFAs in gut dysmotility, the rats of the WAS+SCFAS and SCFAs group were administrated with oral SCFAs. The colonic contractile activity was recorded with a RM6240 multichannel physiological signal system. KEY RESULTS WAS induced gastrointestinal hypermotility and increased defecation in rats. After repeated stress, the fecal SCFAs decreased significantly and the proportion of acetic acid, propionic acid, and butyric acid had changed from Control 2.6:1:1.5 to WAS 2:1:2.3. Protein levels of SCFAs receptors in the colon were promoted by WAS. In addition, oral SCFAs partly inhibited the colonic spontaneous motility both for SCFAs and WAS+SCFAs group in vivo. Meanwhile, we observed acetate had no effect on the contractile amplitudes of muscle strips, but it could slow down contractile frequency in a dose-dependent manner (1-100 mM). Propionate significantly inhibited the motor activity of colonic strips (1-30 mM). Butyrate inhibited the contractile amplitude of CM strips in a dose-dependent manner (1-30 mM), but for LM, it exhibited a stimulating effect at low concentrations of butyrate 1 mM-10 mM and was suppressed at high concentrations of 30 mM butyrate. Total SCFAs increased the contractile amplitude at low concentration (5-50 mM) and inhibited it at high concentration (50-150 mM). All SCFAs slowed down the frequency of colonic activity. CONCLUSION The stress-induced colonic hypermotility by WAS could be ameliorated through oral SCFA supplementation. SCFAs may have potential clinical therapeutic use in modulating gut motility.
Collapse
Affiliation(s)
- FangTing Yuan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Wei Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - HaiXia Ren
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Lin Yan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Ying Wang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
- Key Laboratory of Hubei Province for Digestive System Diseases, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - HeSheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
33
|
Lian W, Chen W. Cyanidin-3-O-Glucoside Improves Colonic Motility During Severe Acute Pancreatitis by Inhibiting the H 2S-Regulated AMPK/mTOR Pathway. Drug Des Devel Ther 2020; 14:3385-3391. [PMID: 32943841 PMCID: PMC7468407 DOI: 10.2147/dddt.s256450] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 07/22/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Cyanidin-3-O-glucoside (C3G) is an important anthocyanin that can modulate digestive system functioning. Inflammation associated with severe acute pancreatitis (SAP) induces H2S production, which impairs the gastrointestinal (GI) system. We investigated the effects of C3G in attenuating SAP-associated colonic motility loss by examining the H2S level and activity of AMP-activated protein kinase (AMPK)/mammalian target of rapamycin (mTOR) pathway. METHODS A rat model of SAP was induced using sodium taurocholate, and the effect of C3G on colonic mobility, H2S production, and the inflammatory response was investigated. AMPK/mTOR pathway changes were detected to assess the pathways by which H2S influences colonic mobility in SAP-model rats. The mechanism underlying H2S function was further examined by subjecting colonic muscle cells (CMCs) to C3G, SAP plasma and an AMPK activator. RESULTS Administering C3G improved colonic motility but suppressed the inflammatory response and H2S production in the SAP-model rats, which was associated with inhibiting the AMPK/mTOR pathway. Furthermore, activating the AMPK/mTOR pathway in CMCs promoted inflammation but suppressed Ca2+ levels, even after administering C3G. CONCLUSION Administering C3G may improve SAP-associated colonic mobility by inhibiting the H2S-mediated AMPK/mTOR pathway.
Collapse
Affiliation(s)
- Wei Lian
- Department of Gastroenterology, Southwest Hospital of Army Medical University, Chongqing, People’s Republic of China
| | - Wensheng Chen
- Department of Gastroenterology, Southwest Hospital of Army Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
34
|
Ma S, Yeom J, Lim YH. Dairy Propionibacterium freudenreichii ameliorates acute colitis by stimulating MUC2 expression in intestinal goblet cell in a DSS-induced colitis rat model. Sci Rep 2020; 10:5523. [PMID: 32218552 PMCID: PMC7099060 DOI: 10.1038/s41598-020-62497-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 03/09/2020] [Indexed: 12/21/2022] Open
Abstract
An intact mucus layer is important in managing inflammatory bowel disease (IBD). Dairy Propionibacterium freudenreichii has probiotic potential, produces propionic acid and is known to promote health. The aim of this study was to evaluate the effects of P. freudenreichii on the improvement of colitis. LS 174T goblet cells and a dextran sodium sulfate (DSS)-induced colitis rat model were used to investigate the P. freudenreichii-induced stimulation of mucin production in vitro and in vivo, respectively. The mRNA and protein expression levels of MUC2, a main component of intestinal mucus, increased in the supernatant of P. freudenreichii culture (SPFC)-treated LS 174 cells. The SPFC and live P. freudenreichii (LPF) reduced the disease activity index (DAI) in the rats with DSS-induced colitis. After treatment with SPFC or LPF, the mRNA levels of typical pro-inflammatory cytokines decreased and the inflammatory state was histologically improved in the rats with DSS-induced colitis. The SPFC and LPF treatments increased the gene and protein expression levels of MUC2 in the rats with DSS-induced colitis compared with the expression levels in the negative control rats, and immunohistochemistry (IHC) showed an increase of the intestinal MUC2 level. In addition, SPFC and LPF augmented the level of propionate in the faeces of the rats with DSS-induced colitis. In conclusion, P. freudenreichii might improve acute colitis by restoring goblet cell number and stimulating the expression of MUC2 in intestinal goblet cells.
Collapse
Affiliation(s)
- Seongho Ma
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Jiah Yeom
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Young-Hee Lim
- Department of Integrated Biomedical and Life Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Public Health Science (Brain Korea 21 PLUS program), Graduate School, Korea University, Seoul, 02841, Republic of Korea. .,Department of Laboratory Medicine, Korea University Guro Hospital, Seoul, 08308, Republic of Korea.
| |
Collapse
|
35
|
Wei P, Keller C, Li L. Neuropeptides in gut-brain axis and their influence on host immunity and stress. Comput Struct Biotechnol J 2020; 18:843-851. [PMID: 32322366 PMCID: PMC7160382 DOI: 10.1016/j.csbj.2020.02.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 02/04/2020] [Accepted: 02/25/2020] [Indexed: 02/08/2023] Open
Abstract
In recent decades, neuropeptides have been found to play a major role in communication along the gut-brain axis. Various neuropeptides are expressed in the central and peripheral nervous systems, where they facilitate the crosstalk between the nervous systems and other major body systems. In addition to being critical to communication from the brain in the nervous systems, neuropeptides actively regulate immune functions in the gut in both direct and indirect ways, allowing for communication between the immune and nervous systems. In this mini review, we discuss the role of several neuropeptides, including calcitonin gene-related peptide (CGRP), pituitary adenylate cyclase-activating polypeptide (PACAP), corticotropin-releasing hormone (CRH) and phoenixin (PNX), in the gut-brain axis and summarize their functions in immunity and stress. We choose these neuropeptides to highlight the diversity of peptide communication in the gut-brain axis.
Collapse
Key Words
- ACTH, adrenocorticotrophic hormone
- Antimicrobial peptides
- CGRP, calcitonin gene-related peptide
- CNS, central nervous system
- CRH, corticotropin-releasing hormone
- CRLR, calcitonin receptor like receptor
- Gut-brain axis
- HPA axis, hypothalamic–pituitary–adrenal axis
- Hypothalamic–pituitary–adrenal axis
- Immunity
- LPS, lipopolysaccharides
- NPY, neuropeptide Y
- Neuropeptide
- PACAP, pituitary adenylate cyclase-activating polypeptide
- PNX, phoenixin
- RAMP1, receptor activity-modifying protein1
- SP, substance P
- Stress
- TRPV1, transient receptor potential vanilloid receptor-1
- VIP, vasoactive intestinal peptide
- α-MSH, α-melanocyte-stimulating hormone
Collapse
Affiliation(s)
- Pingli Wei
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Caitlin Keller
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Corresponding author at: School of Pharmacy & Department of Chemistry, University of Wisconsin-Madison, 777 Highland Ave, Madison, WI 53705, USA.
| |
Collapse
|
36
|
Abstract
Irritable bowel syndrome (IBS) is an extremely common and often very debilitating chronic functional gastrointestinal disorder. Despite its prevalence, significant associated healthcare costs, and quality-of-life issues for affected individuals, our understanding of its etiology remained limited. However, it is now evident that microbial factors play key roles in IBS pathophysiology. Acute gastroenteritis following exposure to pathogens can precipitate the development of IBS, and studies have demonstrated changes in the gut microbiome in IBS patients. These changes may explain some of the symptoms of IBS, including visceral hypersensitivity, as gut microbes exert effects on the host immune system and gut barrier function, as well as the brain-gut axis. Microbial differences also appear to underlie the two main functional categories of IBS: diarrhea-predominant IBS (IBS-D) is associated with small intestinal bacterial overgrowth, which can be diagnosed by a positive hydrogen breath test, and constipation-predominant IBS (IBS-C) is associated with increased levels of methanogenic archaea, which can be diagnosed by a positive methane breath test. Mechanistically, the pathogens that cause gastroenteritis and trigger subsequent IBS development produce a common toxin, cytolethal distending toxin B (CdtB), and antibodies raised against CdtB cross-react with the cytoskeletal protein vinculin and impair gut motility, facilitating bacterial overgrowth. In contrast, methane gas slows intestinal contractility, which may facilitate the development of constipation. While antibiotics and dietary manipulations have been used to relieve IBS symptoms, with varying success, elucidating the specific mechanisms by which gut microbes exert their effects on the host may allow the development of targeted treatments that may successfully treat the underlying causes of IBS.
Collapse
Affiliation(s)
- Mark Pimentel
- Medically Associated Science and Technology (MAST) Program, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Anthony Lembo
- Division of Gastroenterology, Beth Israel Deaconess Hospital, Boston, MA, USA
| |
Collapse
|
37
|
Yong SJ, Tong T, Chew J, Lim WL. Antidepressive Mechanisms of Probiotics and Their Therapeutic Potential. Front Neurosci 2020; 13:1361. [PMID: 32009871 PMCID: PMC6971226 DOI: 10.3389/fnins.2019.01361] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022] Open
Abstract
The accumulating knowledge of the host-microbiota interplay gives rise to the microbiota-gut-brain (MGB) axis. The MGB axis depicts the interkingdom communication between the gut microbiota and the brain. This communication process involves the endocrine, immune and neurotransmitters systems. Dysfunction of these systems, along with the presence of gut dysbiosis, have been detected among clinically depressed patients. This implicates the involvement of a maladaptive MGB axis in the pathophysiology of depression. Depression refers to symptoms that characterize major depressive disorder (MDD), a mood disorder with a disease burden that rivals that of heart diseases. The use of probiotics to treat depression has gained attention in recent years, as evidenced by increasing numbers of animal and human studies that have supported the antidepressive efficacy of probiotics. Physiological changes observed in these studies allow for the elucidation of probiotics antidepressive mechanisms, which ultimately aim to restore proper functioning of the MGB axis. However, the understanding of mechanisms does not yet complete the endeavor in applying probiotics to treat MDD. Other challenges remain which include the heterogeneous nature of both the gut microbiota composition and depressive symptoms in the clinical setting. Nevertheless, probiotics offer some advantages over standard pharmaceutical antidepressants, in terms of residual symptoms, side effects and stigma involved. This review outlines antidepressive mechanisms of probiotics based on the currently available literature and discusses therapeutic potentials of probiotics for depression.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Tommy Tong
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Jactty Chew
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Science and Technology, Sunway University, Bandar Sunway, Malaysia
| |
Collapse
|
38
|
Wang W, Yang Q, Du Y, Zhou X, Du X, Wu Q, Lin L, Song Y, Li F, Yang C, Tan W. Metabolic Labeling of Peptidoglycan with NIR‐II Dye Enables In Vivo Imaging of Gut Microbiota. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.201910555] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Wei Wang
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
| | - Qinglai Yang
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
- Research Center for Advanced Materials and BiotechnologyResearch Institute of Tsinghua University in Shenzhen Shenzhen 518057 China
| | - Yahui Du
- Collaborative Innovation Center of Chemistry for Energy MaterialsThe MOE Key Laboratory of Spectrochemical Analysis and InstrumentationState Key Laboratory of Physical Chemistry of Solid SurfacesDepartment of Chemical BiologyCollege of Chemistry and Chemical EngineeringXiamen University Xiamen 361005 China
| | - Xiaobo Zhou
- Department of ChemistryFudan University Shanghai 200433 China
| | - Xiaochen Du
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
| | - Qiuyue Wu
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
| | - Liyuan Lin
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
| | - Yanling Song
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
| | - Fuyou Li
- Department of ChemistryFudan University Shanghai 200433 China
| | - Chaoyong Yang
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
- Collaborative Innovation Center of Chemistry for Energy MaterialsThe MOE Key Laboratory of Spectrochemical Analysis and InstrumentationState Key Laboratory of Physical Chemistry of Solid SurfacesDepartment of Chemical BiologyCollege of Chemistry and Chemical EngineeringXiamen University Xiamen 361005 China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM)Renji HospitalShanghai Jiao Tong University School of MedicineSchool of Chemistry and Chemical EngineeringShanghai Jiao Tong University Shanghai 200127 China
- Molecular Science and Biomedicine Laboratory (MBL)State Key Laboratory of Chemo/Biosensing and ChemometricsCollege of Chemistry and Chemical EngineeringHunan University Changsha 410082 China
| |
Collapse
|
39
|
Wang W, Yang Q, Du Y, Zhou X, Du X, Wu Q, Lin L, Song Y, Li F, Yang C, Tan W. Metabolic Labeling of Peptidoglycan with NIR-II Dye Enables In Vivo Imaging of Gut Microbiota. Angew Chem Int Ed Engl 2020; 59:2628-2633. [PMID: 31793153 DOI: 10.1002/anie.201910555] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 11/09/2019] [Indexed: 12/23/2022]
Abstract
Deepening our understanding of mammalian gut microbiota has been greatly hampered by the lack of a facile, real-time, and in vivo bacterial imaging method. To address this unmet need in microbial visualization, we herein report the development of a second near-infrared (NIR-II)-based method for in vivo imaging of gut bacteria. Using d-propargylglycine in gavage and then click reaction with an azide-containing NIR-II dye, gut microbiota of a donor mouse was strongly labeled with NIR-II fluorescence on their peptidoglycan. The bacteria could be readily visualized in recipient mouse gut with high spatial resolution and deep tissue penetration under NIR irradiation. The NIR-II-based metabolic labeling strategy reported herein, provides, to the best of our knowledge, the first protocol for facile in vivo visualization of gut microbiota within deep tissues, and offers an instrumental tool for deciphering the complex biology of these gut "dark matters".
Collapse
Affiliation(s)
- Wei Wang
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qinglai Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.,Research Center for Advanced Materials and Biotechnology, Research Institute of Tsinghua University in Shenzhen, Shenzhen, 518057, China
| | - Yahui Du
- Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Xiaobo Zhou
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Xiaochen Du
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Qiuyue Wu
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Liyuan Lin
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Yanling Song
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Fuyou Li
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chaoyong Yang
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.,Collaborative Innovation Center of Chemistry for Energy Materials, The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, China
| | - Weihong Tan
- Institute of Molecular Medicine (IMM), Renji Hospital, Shanghai Jiao Tong University School of Medicine, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200127, China.,Molecular Science and Biomedicine Laboratory (MBL), State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| |
Collapse
|
40
|
Abstract
Stress is a nonspecific response of the body to any demand imposed upon it, disrupting the body homoeostasis and manifested with symptoms such as anxiety, depression or even headache. These responses are quite frequent in the present competitive world. The aim of this review is to explore the effect of stress on gut microbiota. First, we summarize evidence of where the microbiota composition has changed as a response to a stressful situation, and thereby the effect of the stress response. Likewise, we review different interventions that can modulate microbiota and could modulate the stress according to the underlying mechanisms whereby the gut-brain axis influences stress. Finally, we review both preclinical and clinical studies that provide evidence of the effect of gut modulation on stress. In conclusion, the influence of stress on gut microbiota and gut microbiota on stress modulation is clear for different stressors, but although the preclinical evidence is so extensive, the clinical evidence is more limited. A better understanding of the mechanism underlying stress modulation through the microbiota may open new avenues for the design of therapeutics that could boost the pursued clinical benefits. These new designs should not only focus on stress but also on stress-related disorders such as anxiety and depression, in both healthy individuals and different populations.
Collapse
|
41
|
SEURA T, FUKUWATARI T. Differences in gut microbial patterns associated with salivary biomarkers in young Japanese adults. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2020; 39:243-249. [PMID: 33117623 PMCID: PMC7573114 DOI: 10.12938/bmfh.2019-034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 07/11/2020] [Indexed: 12/22/2022]
Abstract
Recent evidence suggests that psychological stress is associated with gut microbiota;
however, there are no reports of its association with gut microbial structure. This
cross-sectional study examined the relationship between psychological stress and gut
microbial patterns in young Japanese adults. Analysis of fecal microbiota was performed
using terminal restriction fragment length polymorphism (T-RFLP). Psychological stress was
assessed using salivary biomarkers, including cortisol, alpha-amylase, and secretory IgA
(S-IgA). Fecal microbial patterns were defined using principal component analysis of the
T-RFLP profile and were classified into two enterotype-like clusters, which were defined
by the B (microbiota dominated by Bacteroides) and BL patterns
(microbiota dominated by Bifidobacterium and
Lactobacillales), respectively. The Simpson index was significantly
higher for the BL pattern than for the B pattern. The salivary cortisol level was
significantly lower for the BL pattern than for the B pattern. Salivary alpha-amylase and
S-IgA levels showed a negative correlation with the Simpson index. Our results raise the
possibility that salivary biomarkers may be involved in the observed differences in
microbial patterns.
Collapse
Affiliation(s)
- Takahiro SEURA
- Department of Sports and Health Sciences, Faculty of Health and Medical Sciences, Aichi Shukutoku University, 2-9 Katahira, Nagakute, Aichi 480-1197, Japan
- Graduate School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka-cho, Hikone, Shiga 522-8533, Japan
- Department of Home Economics and Technology Education, Hokkaido University of Education, 9 Hokumon-cho, Asahikawa, Hokkaido 070-8621, Japan
| | - Tsutomu FUKUWATARI
- Graduate School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka-cho, Hikone, Shiga 522-8533, Japan
| |
Collapse
|
42
|
Louwies T, Johnson AC, Orock A, Yuan T, Greenwood-Van Meerveld B. The microbiota-gut-brain axis: An emerging role for the epigenome. Exp Biol Med (Maywood) 2019; 245:138-145. [PMID: 31805777 DOI: 10.1177/1535370219891690] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Affiliation(s)
- Tijs Louwies
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | | | - Albert Orock
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Tian Yuan
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Beverley Greenwood-Van Meerveld
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.,Oklahoma City VA Medical Center, Oklahoma City, OK 73104, USA.,Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
43
|
Baritaki S, de Bree E, Chatzaki E, Pothoulakis C. Chronic Stress, Inflammation, and Colon Cancer: A CRH System-Driven Molecular Crosstalk. J Clin Med 2019; 8:E1669. [PMID: 31614860 PMCID: PMC6833069 DOI: 10.3390/jcm8101669] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Chronic stress is thought to be involved in the occurrence and progression of multiple diseases, via mechanisms that still remain largely unknown. Interestingly, key regulators of the stress response, such as members of the corticotropin-releasing-hormone (CRH) family of neuropeptides and receptors, are now known to be implicated in the regulation of chronic inflammation, one of the predisposing factors for oncogenesis and disease progression. However, an interrelationship between stress, inflammation, and malignancy, at least at the molecular level, still remains unclear. Here, we attempt to summarize the current knowledge that supports the inseparable link between chronic stress, inflammation, and colorectal cancer (CRC), by modulation of a cascade of molecular signaling pathways, which are under the regulation of CRH-family members expressed in the brain and periphery. The understanding of the molecular basis of the link among these processes may provide a step forward towards personalized medicine in terms of CRC diagnosis, prognosis and therapeutic targeting.
Collapse
Affiliation(s)
- Stavroula Baritaki
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Eelco de Bree
- Division of Surgery, School of Medicine, University of Crete, Heraklion, 71500 Crete, Greece.
| | - Ekaterini Chatzaki
- Laboratory of Pharmacology, Medical School, Democritus University of Thrace, 68100 Alexandroupolis, Greece.
| | - Charalabos Pothoulakis
- IBD Center, Division of Digestive Diseases, David Geffen School of Medicine at UCLA, Los Angeles, CA 10833, USA.
| |
Collapse
|
44
|
Bassett SA, Young W, Fraser K, Dalziel JE, Webster J, Ryan L, Fitzgerald P, Stanton C, Dinan TG, Cryan JF, Clarke G, Hyland N, Roy NC. Metabolome and microbiome profiling of a stress-sensitive rat model of gut-brain axis dysfunction. Sci Rep 2019; 9:14026. [PMID: 31575902 PMCID: PMC6773725 DOI: 10.1038/s41598-019-50593-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 09/09/2019] [Indexed: 12/16/2022] Open
Abstract
Stress negatively impacts gut and brain health. Individual differences in response to stress have been linked to genetic and environmental factors and more recently, a role for the gut microbiota in the regulation of stress-related changes has been demonstrated. However, the mechanisms by which these factors influence each other are poorly understood, and there are currently no established robust biomarkers of stress susceptibility. To determine the metabolic and microbial signatures underpinning physiological stress responses, we compared stress-sensitive Wistar Kyoto (WKY) rats to the normo-anxious Sprague Dawley (SD) strain. Here we report that acute stress-induced strain-specific changes in brain lipid metabolites were a prominent feature in WKY rats. The relative abundance of Lactococcus correlated with the relative proportions of many brain lipids. In contrast, plasma lipids were significantly elevated in response to stress in SD rats, but not in WKY rats. Supporting these findings, we found that the greatest difference between the SD and WKY microbiomes were the predicted relative abundance of microbial genes involved in lipid and energy metabolism. Our results provide potential insights for developing novel biomarkers of stress vulnerability, some of which appear genotype specific.
Collapse
Affiliation(s)
- Shalome A Bassett
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Wayne Young
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Karl Fraser
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Julie E Dalziel
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand. .,Riddet Institute, Massey University, Palmerston North, New Zealand.
| | - Jim Webster
- Farm Systems North, AgResearch Ltd., Ruakura Research Centre, Hamilton, New Zealand
| | - Leigh Ryan
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand
| | - Patrick Fitzgerald
- Laboratory of Neurogastroenterology, APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- Laboratory of Neurogastroenterology, APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - Niall Hyland
- Laboratory of Neurogastroenterology, APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Physiology, University College Cork, Cork, Ireland
| | - Nicole C Roy
- Food Nutrition & Health, AgResearch Ltd., Grasslands Research Centre, Tennent Drive, Palmerston North, 4442, New Zealand.,Riddet Institute, Massey University, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| |
Collapse
|
45
|
Wang SL, Shao BZ, Zhao SB, Chang X, Wang P, Miao CY, Li ZS, Bai Y. Intestinal autophagy links psychosocial stress with gut microbiota to promote inflammatory bowel disease. Cell Death Dis 2019; 10:391. [PMID: 31564717 PMCID: PMC6766473 DOI: 10.1038/s41419-019-1634-x] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 04/14/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022]
Abstract
Psychosocial stress is a critical inducing factor of inflammatory bowel diseases (IBD), while autophagy is a novel central issue of IBD development. The present study investigated the potential role of autophagy in stress-related IBD in patients and animal model. The correlation between psychosocial stress and intestinal autophagy was determined in 23 patients with IBD. Corticotropin-releasing hormone (CRH), a well-established inducer of psychosocial stress, was administrated in dextran sulfate sodium (DSS)-induced IBD mice and lipopolysaccharide (LPS)-stimulated bone marrow-derived macrophages (BMDM). In IBD patients, the autophagy markers beclin-1, LC3-II/I ratio, Atg16L1, and Atg4B were significantly enhanced. The psychosocial stress score was positively associated with the levels of beclin-1 and the LC3II/I ratio in intestinal biopsy specimens. In IBD mouse model, CRH significantly aggravated intestinal inflammation, increased Paneth cell metaplasia, and enhanced intestinal autophagy (beclin-1, Atg16L1, PIK3R4, and Atg4B upregulation; GAA, CTSD, and PPKAA1 downregulation). Additionally, the CRH-induced gut microbial dysbiosis was evidenced by a marked increase in the number of detrimental bacteria. In LPS-stimulated BMDM, CRH substantially increased M1/M2 polarization and thus promoted inflammation. In both IBD mice and LPS-treated BMDM, blockade of autophagy by chloroquine abrogated the unbeneficial effects of CRH, whereas autophagy inducer rapamycin resulted in a pronounced protective effect against IBD lesion. Our data demonstrate that psychosocial stress may link the enhanced intestinal autophagy by modulating gut microbiota and inflammation to aggravate IBD. These data indicate autophagy as a promising therapeutic target for psychosocial stress-related IBD.
Collapse
Affiliation(s)
- Shu-Ling Wang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Bo-Zong Shao
- General Hospital of the Chinese People's Liberation Army, Beijing, China
| | - Sheng-Bing Zhao
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Xin Chang
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Pei Wang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, China.
| | - Zhao-Shen Li
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China.
| | - Yu Bai
- Department of Gastroenterology, Changhai Hospital, Second Military Medical University/Naval Medical University, Shanghai, China.
| |
Collapse
|
46
|
Wei L, Li Y, Tang W, Sun Q, Chen L, Wang X, Liu Q, Yu S, Yu S, Liu C, Ma X. Chronic Unpredictable Mild Stress in Rats Induces Colonic Inflammation. Front Physiol 2019; 10:1228. [PMID: 31616319 PMCID: PMC6764080 DOI: 10.3389/fphys.2019.01228] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 09/09/2019] [Indexed: 01/01/2023] Open
Abstract
Chronic psychological stress is associated with an increased risk for relapse of inflammatory bowel diseases (IBD) and impedes the treatment of this condition. However, the impact of stress on the risk of IBD onset remains unclear. The goal of the present study was to examine whether chronic unpredictable mild stress (CUMS) could initiate or aggravate the onset of colon inflammation in rats which, in turn, would be capable of triggering bowel disease. We found that CUMS exposure increased infiltration of CD-45 positive cells and MPO activity, as well as augmented the expression of the inflammatory cytokines, IFN-γ and IL-6 within the colon of these rats. In addition, CUMS treatment changed the composition and diversity of gut microbiota and enhanced intestinal epithelial permeability, indicating the presence of a defect in the intestinal barrier. This CUMS-induced disruption of mucosal barrier integrity was associated with a reduction in expression of the tight junction protein, occludin 1, and an inhibition in mucosal layer functioning via reductions in goblet cells. Results from bacterial cultures revealed an increased presence of bacterial invasion after CUMS treatment as compared with that observed in controls. Thus, our data indicate that CUMS treatment induces alterations of the fecal microbiome and intestinal barrier defects, which facilitates bacterial invasion into colonic mucosa and further exacerbates inflammatory reactions within the colon. Accordingly, chronic stress may predispose patients to gastrointestinal infection and increase the risk of inflammation-related gut diseases.
Collapse
Affiliation(s)
- Lina Wei
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Ye Li
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Wenjun Tang
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qian Sun
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Lixin Chen
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xia Wang
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Qingyi Liu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Siqi Yu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Shuyan Yu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Chuanyong Liu
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Xuelian Ma
- Department of Physiology, School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
47
|
Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, Helbling JC, Lévy É, Chevalier G, Leboyer M, Eberl G, Layé S, Capuron L, Vergnolle N, Neunlist M, Boudin H, Lepage P, Darnaudéry M. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun 2019; 80:179-192. [PMID: 30872090 DOI: 10.1016/j.bbi.2019.03.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
Abstract
The accumulation of adverse events in utero and during childhood differentially increases the vulnerability to psychiatric diseases in men and women. Gut microbiota is highly sensitive to the early environment and has been recently hypothesized to affect brain development. However, the impact of early-life adversity on gut microbiota, notably with regards to sex differences, remains to be explored. We examined the effects of multifactorial early-life adversity on behavior and microbiota composition in C3H/HeN mice of both sexes exposed to a combination of maternal immune activation (lipopolysaccharide injection on embryonic day 17, 120 µg/kg, i.p.), maternal separation (3hr per day from postnatal day (PND)2 to PND14) and maternal unpredictable chronic mild stress. At adulthood, offspring exposed to multi-hit early adversity showed sex-specific behavioral phenotypes with males exhibiting deficits in social behavior and females showing increased anxiety in the elevated plus maze and increased compulsive behavior in the marble burying test. Early adversity also differentially regulated gene expression in the medial prefrontal cortex (mPFC) according to sex. Interestingly, several genes such as Arc, Btg2, Fosb, Egr4 or Klf2 were oppositely regulated by early adversity in males versus females. Finally, 16S-based microbiota profiling revealed sex-dependent gut dysbiosis. In males, abundance of taxa belonging to Lachnospiraceae and Porphyromonadaceae families or other unclassified Firmicutes, but also Bacteroides, Lactobacillus and Alloprevotella genera was regulated by early adversity. In females, the effects of early adversity were limited and mainly restricted to Lactobacillus and Mucispirillum genera. Our work reveals marked sex differences in a multifactorial model of early-life adversity, both on emotional behaviors and gut microbiota, suggesting that sex should systematically be considered in preclinical studies both in neurogastroenterology and psychiatric research.
Collapse
Affiliation(s)
- Marion Rincel
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Philippe Aubert
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Julien Chevalier
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Pierre-Antoine Grohard
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Lilian Basso
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Camille Monchaux de Oliveira
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Jean Christophe Helbling
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Élodie Lévy
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | | | - Marion Leboyer
- Université Paris-est-Créteil, Laboratoire Psychiatrie translationnelle, INSERM U955, Hôpital Chenevier-Mondor, Créteil, France
| | - Gérard Eberl
- Unité Microenvironnement et Immunité, Institut Pasteur, Paris, France
| | - Sophie Layé
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Lucile Capuron
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Nathalie Vergnolle
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Hélène Boudin
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, Univ. Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Darnaudéry
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
48
|
Abstract
The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.
Collapse
|
49
|
Pittman QJ. Stress co-opts the gut to affect epileptogenesis. Commentary on "Facilitation of kindling epileptogenesis by chronic stress may be mediated by intestinal microbiome". Epilepsia Open 2019; 4:230-231. [PMID: 31168487 PMCID: PMC6546071 DOI: 10.1002/epi4.12326] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/19/2022] Open
Affiliation(s)
- Quentin J Pittman
- Department of Physiology and Pharmacology, Hotchkiss Brain Institute Cumming School of Medicine University of Calgary Calgary Alberta Canada
| |
Collapse
|
50
|
Rincel M, Olier M, Minni A, Monchaux de Oliveira C, Matime Y, Gaultier E, Grit I, Helbling JC, Costa AM, Lépinay A, Moisan MP, Layé S, Ferrier L, Parnet P, Theodorou V, Darnaudéry M. Pharmacological restoration of gut barrier function in stressed neonates partially reverses long-term alterations associated with maternal separation. Psychopharmacology (Berl) 2019; 236:1583-1596. [PMID: 31147734 DOI: 10.1007/s00213-019-05252-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
Abstract
RATIONALE Intestinal permeability plays an important role in gut-brain axis communication. Recent studies indicate that intestinal permeability increases in neonate pups during maternal separation (MS). OBJECTIVES The present study aims to determine whether pharmacological inhibition of myosin light chain kinase (MLCK), which regulates tight junction contraction and controls intestinal permeability, in stressed neonates, protects against the long-term effects of MS. METHODS Male Wistar rats were exposed to MS (3 h per day from post-natal day (PND)2 to PND14) or left undisturbed and received daily intraperitoneal injection of a MLCK inhibitor (ML-7, 5 mg/kg) or vehicle during the same period. At adulthood, emotional behaviors, corticosterone response to stress, and gut microbiota composition were analyzed. RESULTS ML-7 restored gut barrier function in MS rats specifically during the neonatal period. Remarkably, ML-7 prevented MS-induced sexual reward-seeking impairment and reversed the alteration of corticosterone response to stress at adulthood. The effects of ML-7 were accompanied by the normalization of the abundance of members of Lachnospiraceae, Clostridiales, Desulfovibrio, Bacteroidales, Enterorhabdus, and Bifidobacterium in the feces of MS rats at adulthood. CONCLUSIONS Altogether, our work suggests that improvement of intestinal barrier defects during development may alleviate some of the long-term effects of early-life stress and provides new insight on brain-gut axis communication in a context of stress.
Collapse
Affiliation(s)
- Marion Rincel
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Maïwenn Olier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Amandine Minni
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | | | - Yann Matime
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Eric Gaultier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Isabelle Grit
- UMR 1280, Institut des maladies de l'appareil digestif, PhAN, INRA, University of Nantes, Nantes, France
| | | | - Anna Maria Costa
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Amandine Lépinay
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Sophie Layé
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Laurent Ferrier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Patricia Parnet
- UMR 1280, Institut des maladies de l'appareil digestif, PhAN, INRA, University of Nantes, Nantes, France
| | - Vassilia Theodorou
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Muriel Darnaudéry
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France.
| |
Collapse
|