1
|
Baker CC, Sessenwein JL, Wood HM, Yu Y, Tsang Q, Alward TA, Jimenez Vargas NN, Omar AA, McDonnel A, Segal JP, Sjaarda CP, Bunnett NW, Schmidt BL, Caminero A, Boev N, Bannerman CA, Ghasemlou N, Sheth PM, Vanner SJ, Reed DE, Lomax AE. Protease-Induced Excitation of Dorsal Root Ganglion Neurons in Response to Acute Perturbation of the Gut Microbiota Is Associated With Visceral and Somatic Hypersensitivity. Cell Mol Gastroenterol Hepatol 2024; 18:101334. [PMID: 38494056 PMCID: PMC11350452 DOI: 10.1016/j.jcmgh.2024.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 03/08/2024] [Accepted: 03/11/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND & AIMS Abdominal pain is a major symptom of diseases that are associated with microbial dysbiosis, including irritable bowel syndrome and inflammatory bowel disease. Germ-free mice are more prone to abdominal pain than conventionally housed mice, and reconstitution of the microbiota in germ-free mice reduces abdominal pain sensitivity. However, the mechanisms underlying microbial modulation of pain remain elusive. We hypothesized that disruption of the intestinal microbiota modulates the excitability of peripheral nociceptive neurons. METHODS In vivo and in vitro assays of visceral sensation were performed on mice treated with the nonabsorbable antibiotic vancomycin (50 μg/mL in drinking water) for 7 days and water-treated control mice. Bacterial dysbiosis was verified by 16s rRNA analysis of stool microbial composition. RESULTS Treatment of mice with vancomycin led to an increased sensitivity to colonic distension in vivo and in vitro and hyperexcitability of dorsal root ganglion (DRG) neurons in vitro, compared with controls. Interestingly, hyperexcitability of DRG neurons was not restricted to those that innervated the gut, suggesting a widespread effect of gut dysbiosis on peripheral pain circuits. Consistent with this, mice treated with vancomycin were more sensitive than control mice to thermal stimuli applied to hind paws. Incubation of DRG neurons from naive mice in serum from vancomycin-treated mice increased DRG neuron excitability, suggesting that microbial dysbiosis alters circulating mediators that influence nociception. The cysteine protease inhibitor E64 (30 nmol/L) and the protease-activated receptor 2 (PAR-2) antagonist GB-83 (10 μmol/L) each blocked the increase in DRG neuron excitability in response to serum from vancomycin-treated mice, as did the knockout of PAR-2 in NaV1.8-expressing neurons. Stool supernatant, but not colonic supernatant, from mice treated with vancomycin increased DRG neuron excitability via cysteine protease activation of PAR-2. CONCLUSIONS Together, these data suggest that gut microbial dysbiosis alters pain sensitivity and identify cysteine proteases as a potential mediator of this effect.
Collapse
Affiliation(s)
- Corey C Baker
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Jessica L Sessenwein
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Hannah M Wood
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Yang Yu
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Quentin Tsang
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Taylor A Alward
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | | | - Amal Abu Omar
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Abby McDonnel
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Julia P Segal
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Calvin P Sjaarda
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, Neuroscience Institute, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, Neuroscience Institute, New York University, New York, New York
| | - Alberto Caminero
- Department of Medicine, Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Nadejda Boev
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Courtney A Bannerman
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada
| | - Nader Ghasemlou
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Anesthesiology and Perioperative Medicine, Queen's Unversity, Kingston, Ontario, Canada
| | - Prameet M Sheth
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Stephen J Vanner
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University, Kingston, Ontario, Canada; Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
2
|
Shukla AK, Kumari A, Kumar A. Gut brain regulation using psychobiotics for improved neuropsychological illness. Dev Psychobiol 2023; 65:e22404. [PMID: 37338246 DOI: 10.1002/dev.22404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 06/21/2023]
Abstract
"Psychobiotics" are a novel class of probiotics that are beneficial to the health and functional efficiency of our brain and psychology. The main hold on command in ill conditions of the brain and psychology is overtaken by these psychobiotic bacteria (a dietary supplement) via the action/determined role of bacterial neurochemicals or neuroactive substances that are released by them in the intestinal epithelium after their ingestion. Although these psychobiotics flourish in the gut of the host consuming them, the effect is widely spread to the brain due to the communication between the gut and the brain via the bidirectional gut-brain axis. The nervous system involved in this directional process includes both the enteric nervous system and the central nervous system. With time, several corroborations have proved the effectiveness of psychobiotics in terms of mental illnesses and brain disorders. In the prevailing situation of the coronavirus pandemic, psychobiotics may serve as an aid because a majority of the population worldwide is already suffering from psychological issues due to changes in lifestyle and dietary habits, and in need of an immediate solution to cope with it. Moreover, the in silico approach is also vital for the development of biological relevance to neurosubstances.
Collapse
Affiliation(s)
- Adarsh Kumar Shukla
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| | - Anita Kumari
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| |
Collapse
|
3
|
Drljača J, Milošević N, Milanović M, Abenavoli L, Milić N. When the microbiome helps the brain-current evidence. CNS Neurosci Ther 2023; 29 Suppl 1:43-58. [PMID: 36601680 PMCID: PMC10314113 DOI: 10.1111/cns.14076] [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: 08/22/2022] [Revised: 12/05/2022] [Accepted: 12/17/2022] [Indexed: 01/06/2023] Open
Abstract
The gut microbiota-brain axis has been recognized as a network of connections that provides communication between the gut microflora and both central and autonomic nervous system. The gut microbiota alteration has been targeted for therapy in various neurodegenerative and psychiatric disbalances. Psychobiotics are probiotics that contribute beneficially to the brain function and the host mental health as a result of an interaction with the commensal gut bacteria, although their mechanism of action has not been completely revealed. In this state-of-art review, the findings about the potential therapeutic effects of the psychobiotics alone or in combination with conventional medicine in the treatment of neurodegenerative diseases such as Alzheimer's disease and Parkinson's disease, as well as in some psychiatric diseases like depression, schizophrenia, and bipolar disorder, have been summarized. The evidence of the psychobiotics therapeutic outcomes obtained in preclinical and clinical trials have been given respectively for the observed neurodegenerative and psychiatric disorders.
Collapse
Affiliation(s)
- Jovana Drljača
- Faculty of Medicine, Department of PharmacyUniversity of Novi SadNovi SadSerbia
| | - Nataša Milošević
- Faculty of Medicine, Department of PharmacyUniversity of Novi SadNovi SadSerbia
| | - Maja Milanović
- Faculty of Medicine, Department of PharmacyUniversity of Novi SadNovi SadSerbia
| | - Ludovico Abenavoli
- Department of Health SciencesUniversity Magna Graecia Campus “Salvatore Venuta”CatanzaroItaly
| | - Nataša Milić
- Faculty of Medicine, Department of PharmacyUniversity of Novi SadNovi SadSerbia
| |
Collapse
|
4
|
Tian S, Zhang H, Chen S, Wu P, Chen M. Global research progress of visceral hypersensitivity and irritable bowel syndrome: bibliometrics and visualized analysis. Front Pharmacol 2023; 14:1175057. [PMID: 37201020 PMCID: PMC10185792 DOI: 10.3389/fphar.2023.1175057] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/18/2023] [Indexed: 05/20/2023] Open
Abstract
Background: Irritable bowel syndrome (IBS) is a group of functional intestinal disorders characterized by abdominal pain, bloating, and changes in bowel habits, and/or stool characteristics. Recent studies have shown that there has been a significant advancement in the study of visceral hypersensitivity in IBS. Through the use of bibliometrics, this study aims to provide a comprehensive overview of the knowledge structure and research hotpots of visceral hypersensitivity in IBS. Methods: Publications related to visceral hypersensitivity in IBS from 2012 to 2022 were searched on the web of science core collection (WoSCC) database. CiteSpace.6.1. R2 and Vosviewer 1.6.17 were used to perform bibliometric analysis. Results: A total of 974 articles led by China and the United States from 52 countries were included. Over the past decade, the number of articles on visceral hypersensitivity and IBS has steadily increased year by year. China, the United States, and Belgium are the main countries in this field. Univ Oklahoma, Univ Gothenburg, and Zhejiang University are the main research institutions. Simren, Magnus, Greenwood-van meerveld, Beverley, and Tack, Jan are the most published authors in this research field. The research on the causes, genes, and pathways involved in visceral hypersensitivity in IBS and the mechanism of IBS are the main topics and hotspots in this field. This study also found that gut microbiota may be related to the occurrence of visceral hypersensitivity, and probiotics may be a new method for the treatment of visceral hypersensitivity and pain, which may become a new direction for research in this field. Conclusion: This is the first bibliometric study to comprehensively summarize the research trends and developments of visceral hypersensitivity in IBS. This information provides the research frontier and hot topics in this field in recent years, which will provide a reference for scholars studying this field.
Collapse
Affiliation(s)
- Siyu Tian
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Hang Zhang
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Siqi Chen
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Pengning Wu
- School of Clinical Medicine, Chengdu University of TCM, Chengdu, China
| | - Min Chen
- Department of Colorectal Diseases, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Min Chen,
| |
Collapse
|
5
|
Dono A, Esquenazi Y, Choi HA. Gut microbiome and neurocritically ill patients. JOURNAL OF NEUROCRITICAL CARE 2022. [DOI: 10.18700/jnc.220058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Since the times of Rokitansky and Cushing, we have been fascinated by the connections between the gut and the brain. Recent advances in next-generation sequencing techniques have shown that this relationship is even more complex and integral to our sense of self than previously imagined. As these techniques refine our understanding of the abundance and diversity of the gut bacterial microbiome, the relationship between the gut and the brain has been redefined. Now, this is understood as a complex symbiotic network with bidirectional communication, the gut-brain axis. The implication of this communication involves an intense focus of research on a variety of chronic psychiatric, neurological, neurodegenerative, and neuro-oncological diseases. Recently, the gut-brain axis has been studied in neurologically ill patients requiring intensive care. Preliminary studies have shown that acute brain injury changes the bacterial phenotype from one that is symbiotic with the host human to one that is pathologic, termed the “pathobiome.” This can contribute to nosocomial pneumonia and sepsis. The first studies in neurologically ill patients in the neurointensive care unit (NeuroICU) demonstrated changes in the gut microbiome between neuroICU patients and healthy matched subjects. Specifically, a decrease in short-chain fatty acid-producing bacteria and increase in harmful gut microbes have been associated with mortality and decreased function at discharge. Although these preliminary findings are exciting and have opened a new field of research in the complex NeuroICU population, there are several limitations and challenges. Further investigation is needed to confirm these correlations and understand their implications on patients in a complex intensive care environment.
Collapse
|
6
|
Dono A, Nickles J, Rodriguez-Armendariz AG, Mcfarland BC, Ajami NJ, Ballester LY, Wargo JA, Esquenazi Y. Glioma and the Gut-Brain Axis: Opportunities and Future Perspectives. Neurooncol Adv 2022; 4:vdac054. [PMID: 35591978 PMCID: PMC9113089 DOI: 10.1093/noajnl/vdac054] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The gut–brain axis has presented a valuable new dynamic in the treatment of cancer and central nervous system (CNS) diseases. However, little is known about the potential role of this axis in neuro-oncology. The goal of this review is to highlight potential implications of the gut–brain axis in neuro-oncology, in particular gliomas, and future areas of research. The gut–brain axis is a well-established biochemical signaling axis that has been associated with various CNS diseases. In neuro-oncology, recent studies have described gut microbiome differences in tumor-bearing mice and glioma patients compared to controls. These differences in the composition of the microbiome are expected to impact the metabolic functionality of each microbiome. The effects of antibiotics on the microbiome may affect tumor growth and modulate the immune system in tumor-bearing mice. Preliminary studies have shown that the gut microbiome might influence PD-L1 response in glioma-bearing mice, as previously observed in other non-CNS cancers. Groundbreaking studies have identified intratumoral bacterial DNA in several cancers including high-grade glioma. The gut microbiome and its manipulation represent a new and relatively unexplored area that could be utilized to enhance the effectiveness of therapy in glioma. Further mechanistic studies of this therapeutic strategy are needed to assess its clinical relevance.
Collapse
Affiliation(s)
- Antonio Dono
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jack Nickles
- Northeastern University, College of Science, Boston, MA, USA
- Memorial Hermann Hospital, Houston, TX, USA
- Tufts Medical Center, Boston, MA, USA
| | - Ana G Rodriguez-Armendariz
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, USA
- Escuela de Medicina y Ciencias de la Salud, Tecnológico de Monterrey, Monterrey, Nuevo Leon, México
| | - Braden C Mcfarland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nadim J Ajami
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Leomar Y Ballester
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
- Memorial Hermann Hospital, Houston, TX, USA
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, University of Texas Health Science Center at Houston, Houston, TX, USA
- McGovern Medical School and Center of Precision Health, School of Biomedical Informatics, University of Texas Health Science Center at Houston, Houston, TX, USA
- Memorial Hermann Hospital, Houston, TX, USA
| |
Collapse
|
7
|
Jomehzadeh N, Amin M, Javaherizadeh H, Rashno M. MOLECULAR ASSESSMENT OF FECAL LACTOBACILLI POPULATIONS IN CHILDREN WITH FUNCTIONAL CONSTIPATION. ARQUIVOS DE GASTROENTEROLOGIA 2022; 59:244-250. [PMID: 35830036 DOI: 10.1590/s0004-2803.202202000-44] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 03/29/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Investigation of the gut-specific bacterial strains including lactobacilli is essential for understanding the bacterial etiology of constipation. OBJECTIVE This study aimed to compare the prevalence and quantity of intestinal lactobacilli in constipated children and healthy controls. METHODS Forty children fulfilling Rome IV criteria for functional constipation and 40 healthy controls were recruited. Fecal samples were analyzed using species-specific polymerase chain reaction followed by random amplified polymorphic DNA-PCR and quantitative real-time PCR. RESULTS Totally, seven different species of lactobacilli were detected. Out of 80 volunteers, 65 (81.3%) were culture and species-specific PCR positive from which 25 (38.46%) constipated children and 40 (61.54%) healthy subjects. The most prevalent species were L. paracasei 21 (32.3%) followed by L. plantarum 18 (27.7%) among both healthy and patient groups. Analysis of the RAPD dendrograms displayed that strains isolated from constipated and non-constipated children have similarity coefficients of more than 90%. The qPCR assays demonstrated constipated children had a lower amount of total lactobacilli population (per gram of feces) than healthy controls. CONCLUSION Our findings showed that the mere existence of various species of Lactobacillus in the gut does not enough to prevent some gastrointestinal disorders such as functional constipation, and their quantity plays a more important role.
Collapse
Affiliation(s)
- Nabi Jomehzadeh
- Department of Microbiology, School of Medicine, Abadan University of Medical Sciences, Abadan, Iran
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mansour Amin
- Infectious and Tropical Diseases Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hazhir Javaherizadeh
- Alimentary Tract Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Rashno
- Department of Microbiology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
8
|
One Giant Leap from Mouse to Man: The Microbiota-Gut-Brain Axis in Mood Disorders and Translational Challenges Moving towards Human Clinical Trials. Nutrients 2022; 14:nu14030568. [PMID: 35276927 PMCID: PMC8840472 DOI: 10.3390/nu14030568] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
The microbiota–gut–brain axis is a bidirectional communication pathway that enables the gut microbiota to communicate with the brain through direct and indirect signaling pathways to influence brain physiology, function, and even behavior. Research has shown that probiotics can improve several aspects of health by changing the environment within the gut, and several lines of evidence now indicate a beneficial effect of probiotics on mental and brain health. Such evidence has prompted the arrival of a new term to the world of biotics research: psychobiotics, defined as any exogenous influence whose effect on mental health is bacterially mediated. Several taxonomic changes in the gut microbiota have been reported in neurodevelopmental disorders, mood disorders such as anxiety and depression, and neurodegenerative disorders such as Alzheimer’s disease. While clinical evidence supporting the role of the gut microbiota in mental and brain health, and indeed demonstrating the beneficial effects of probiotics is rapidly accumulating, most of the evidence to date has emerged from preclinical studies employing different animal models. The purpose of this review is to focus on the role of probiotics and the microbiota–gut–brain axis in relation to mood disorders and to review the current translational challenges from preclinical to clinical research.
Collapse
|
9
|
Gut microbiome effects on neuronal excitability & activity: Implications for epilepsy. Neurobiol Dis 2022; 165:105629. [PMID: 35033659 DOI: 10.1016/j.nbd.2022.105629] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 01/10/2022] [Indexed: 12/19/2022] Open
Abstract
It is now well established that the bacterial population of the gastrointestinal system, known as the gut microbiome, is capable of influencing the brain and its dependent functions. Links have been demonstrated between the microbiome and a variety of normal and pathological neural functions, including epilepsy. Many of these microbiome-brain links involve the direct or indirect modulation of the excitability and activity of individual neurons by the gut microbiome. Such links may be particularly significant when it comes to microbiome modulation of epilepsy, often considered a disorder of neuronal excitability. In this review we consider the current evidence of a relationship between the gut microbiome and the excitability or activity of neurons in the context of epilepsy. The review focuses particularly on evidence of direct, causal microbiome effects on neuronal excitability or activity, but also considers demonstrations of microbiome to host interactions that are likely to have an indirect influence. While we identify a few common themes, it is apparent that deriving general mechanistic principles of microbiome influence on these parameters in epilepsy will require considerable further study to tease out the many interacting factors, systems, and conditions.
Collapse
|
10
|
Kuwahara A, Matsuda K, Kuwahara Y, Asano S, Inui T, Marunaka Y. Microbiota-gut-brain axis: enteroendocrine cells and the enteric nervous system form an interface between the microbiota and the central nervous system. Biomed Res 2021; 41:199-216. [PMID: 33071256 DOI: 10.2220/biomedres.41.199] [Citation(s) in RCA: 52] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The microbiota-gut-brain axis transmits bidirectional communication between the gut and the central nervous system and links the emotional and cognitive centers of the brain with peripheral gut functions. This communication occurs along the axis via local, paracrine, and endocrine mechanisms involving a variety of gut-derived peptide/amine produced by enteroendocrine cells. Neural networks, such as the enteric nervous system, and the central nervous system, including the autonomic nervous system, also transmit information through the microbiota-gut-brain axis. Recent advances in research have described the importance of the gut microbiota in influencing normal physiology and contributing to disease. We are only beginning to understand this bidirectional communication system. In this review, we summarize the available data supporting the existence of these interactions, highlighting data related to the contribution of enteroendocrine cells and the enteric nervous system as an interface between the gut microbiota and brain.
Collapse
Affiliation(s)
- Atsukazu Kuwahara
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University
| | - Kyoko Matsuda
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | - Yuko Kuwahara
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University
| | - Shinji Asano
- Department of Molecular Physiology, College of Pharmaceutical Sciences, Ritsumeikan University
| | | | - Yoshinori Marunaka
- Research Unit for Epithelial Physiology and Research Center for Drug Discovery and Pharmaceutical Development Science, Research Organization of Science and Technology, Ritsumeikan University.,Department of Molecular Cell Physiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine.,Research Institute for Clinical Physiology, Kyoto Industrial Health Association
| |
Collapse
|
11
|
Berding K, Vlckova K, Marx W, Schellekens H, Stanton C, Clarke G, Jacka F, Dinan TG, Cryan JF. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv Nutr 2021; 12:1239-1285. [PMID: 33693453 PMCID: PMC8321864 DOI: 10.1093/advances/nmaa181] [Citation(s) in RCA: 129] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the gut microbiota has emerged as a key component in regulating brain processes and behavior. Diet is one of the major factors involved in shaping the gut microbiota composition across the lifespan. However, whether and how diet can affect the brain via its effects on the microbiota is only now beginning to receive attention. Several mechanisms for gut-to-brain communication have been identified, including microbial metabolites, immune, neuronal, and metabolic pathways, some of which could be prone to dietary modulation. Animal studies investigating the potential of nutritional interventions on the microbiota-gut-brain axis have led to advancements in our understanding of the role of diet in this bidirectional communication. In this review, we summarize the current state of the literature triangulating diet, microbiota, and host behavior/brain processes and discuss potential underlying mechanisms. Additionally, determinants of the responsiveness to a dietary intervention and evidence for the microbiota as an underlying modulator of the effect of diet on brain health are outlined. In particular, we emphasize the understudied use of whole-dietary approaches in this endeavor and the need for greater evidence from clinical populations. While promising results are reported, additional data, specifically from clinical cohorts, are required to provide evidence-based recommendations for the development of microbiota-targeted, whole-dietary strategies to improve brain and mental health.
Collapse
Affiliation(s)
| | | | - Wolfgang Marx
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
| | - Harriet Schellekens
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - Felice Jacka
- Deakin University, iMPACT – the Institute for Mental and Physical Health and Clinical Translation, Food & Mood Centre, School of Medicine, Barwon Health, Geelong, VIC,Australia
- Centre for Adolescent Health, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Black Dog Institute, Randwick, NSW, Australia
- College of Public Health, Medical & Veterinary Sciences, James Cook University, Douglas, QLD, Australia
| | - Timothy G Dinan
- APC Microbiome Ireland, Cork, Ireland
- Department of Psychiatry and Neurobehavioural Sciences, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
12
|
Satia I, Cusack R, Stevens C, Schlatman A, Wattie J, Mian F, Killian KJ, O'Byrne PM, Bienenstock J, Forsythe P, Gauvreau GM. Limosilactobacillus reuteri DSM-17938 for preventing cough in adults with mild allergic asthma: A double-blind randomized placebo-controlled cross-over study. Clin Exp Allergy 2021; 51:1133-1143. [PMID: 34192396 DOI: 10.1111/cea.13976] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Cough is a common troublesome symptom in asthma which is neuronally mediated. Limosilactobacillus reuteri DSM-17938 (L. reuteri DSM-17938) is a probiotic shown to be effective in pre-clinical models at suppressing neuronal responses to capsaicin, a transient receptor potential vanilloid agonist (TRPV1). OBJECTIVE Investigate the effects of DSM-17938 versus matched placebo on capsaicin-evoked coughs in mild allergic asthmatics. METHODS We performed a 4-visit, randomized, double-blind, placebo-controlled, two-way cross-over study comparing full dose cough responses with inhaled capsaicin in mild allergic asthmatics after 1 month of treatment with DSM-17938 compared with matched placebo. Randomization and allocation to trial group were carried out by a central computer system. Histamine skin prick testing, airway hyper-responsiveness and inflammatory cells in induced sputum were measured at every visit. Blood was collected to extract PBMCs and stimulated with CD3/CD28 to ascertain the effects of DSM-17938 /placebo on T-cell cytokine responses. RESULTS Seventeen subjects were recruited and 15 completed the study (8 females, mean age 27.3 years). There was no difference in the change in maximum capsaicin-evoked coughs (Emax) after treatment with L. reuteri DSM-17938 compared with placebo [mean difference 2.07 coughs (95% CI -2.77 to 6.91, p = .38) or relative changes in geometric mean ratios for the dose evoking at least half the Emax (ED50) [1.05 (95% CI 0.31-3.58, p = .94)], concentration evoking 2 coughs (C2) [0.63 (0.26-1.53), p = .28] and 5 coughs (C5) [0.79 (0.25-2.50), p = .67]. There was no effect on histamine skin prick wheal size, intensity of itch sensation, methacholine PC20, airway inflammation or T-cell responses after stimulation with CD3/CD28. There were no serious adverse events. One subject developed a mild upper respiratory tract infection and another mild transient nausea whilst on DSM-17938. CONCLUSION In this small study in adults with mild allergic asthma, we found no evidence that L. reuteri DSM-17938 has any systemic effects on airway nerves, smooth muscle, sputum inflammatory cells, skin responses or T-cell responses after oral consumption. TRIAL REGISTRATION Clinicaltrials.gov Identifier: NCT03603522.
Collapse
Affiliation(s)
- Imran Satia
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - Ruth Cusack
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Catie Stevens
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Abbey Schlatman
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Jennifer Wattie
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Firoz Mian
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Kieran J Killian
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul M O'Byrne
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada.,Firestone Institute for Respiratory Health, St Joseph's Healthcare, Hamilton, ON, Canada
| | - John Bienenstock
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Paul Forsythe
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| | - Gail M Gauvreau
- Division of Respirology, Department of Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
13
|
Simon E, Călinoiu LF, Mitrea L, Vodnar DC. Probiotics, Prebiotics, and Synbiotics: Implications and Beneficial Effects against Irritable Bowel Syndrome. Nutrients 2021; 13:nu13062112. [PMID: 34203002 PMCID: PMC8233736 DOI: 10.3390/nu13062112] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is still a common functional gastrointestinal disease that presents chronic abdominal symptoms but with a pathophysiology that is not yet fully elucidated. Moreover, the use of the synergistic combination of prebiotics and probiotics, known as synbiotics, for IBS therapy is still in the early stages. Advancements in technology led to determining the important role played by probiotics in IBS, whereas the present paper focuses on the detailed review of the various pathophysiologic mechanisms of action of probiotics, prebiotics, and synbiotics via multidisciplinary domains involving the gastroenterology (microbiota modulation, alteration of gut barrier function, visceral hypersensitivity, and gastrointestinal dysmotility) immunology (intestinal immunological modulation), and neurology (microbiota–gut–brain axis communication and co-morbidities) in mitigating the symptoms of IBS. In addition, this review synthesizes literature about the mechanisms involved in the beneficial effects of prebiotics and synbiotics for patients with IBS, discussing clinical studies testing the efficiency and outcomes of synbiotics used as therapy for IBS.
Collapse
Affiliation(s)
- Elemer Simon
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
| | - Lavinia Florina Călinoiu
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
| | - Laura Mitrea
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
| | - Dan Cristian Vodnar
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
- Correspondence: ; Tel.: +40-747-341-881
| |
Collapse
|
14
|
Hegde S, Lin YM, Fu Y, Savidge T, Shi XZ. Precision Lactobacillus reuteri therapy attenuates luminal distension-associated visceral hypersensitivity by inducing peripheral opioid receptors in the colon. Pain 2020; 161:2737-2749. [PMID: 32569084 PMCID: PMC7669621 DOI: 10.1097/j.pain.0000000000001967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Luminal distension and abdominal pain are major clinical hallmarks of obstructive bowel disorders and functional bowel disorders linked to gut dysbiosis. Our recent studies found that chronic lumen distension increased visceral sensitivity and decreased abundance of gut commensal Lactobacillus reuteri in a rodent model of partial colon obstruction (OB). To establish causation, we performed precision microbial therapy to assess whether recolonization of L. reuteri prevents visceral hypersensitivity in lumen distension, and if so, to identify the gut-microbiota mechanism. Lumen distension was induced in Sprague-Dawley rats by implanting an OB band in the distal colon for up to 7 days. L. reuteri strains or vehicle were gavage ingested 1 × 10 colony-forming units/g daily starting 2 days before OB. L. reuteri rat strains that were able to recolonize obstructed colon significantly improved food intake and body weight in OB rats, and attenuated referred visceral hyperalgesia measured by the withdrawal response to von Frey filament applications to the abdomen. Mechanistically, L. reuteri treatment attenuated hyperexcitability of the dorsal root ganglia neurons projecting to the distended colon by promoting opioid receptor function in affected tissues. The expression of µ, δ, and κ opioid receptors was significantly downregulated in colonic muscularis externae and sensory neurons in OB rats. However, L. reuteri treatment prevented the loss of opioid receptors. Furthermore, administration of peripheral opioid receptor antagonist naloxone methiodide abolished the analgesic effect of L. reuteri in OB. In conclusion, precision L. reuteri therapy prevents lumen distension-associated visceral hypersensitivity by local bacterial induction of opioid receptors.
Collapse
Affiliation(s)
- Shrilakshmi Hegde
- Dept. of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - You-Min Lin
- Dept. of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Yu Fu
- Dept. of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Tor Savidge
- Dept. of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Xuan-Zheng Shi
- Dept. of Internal Medicine, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
15
|
Al-Balawi M, Morsy FM. Enterococcus faecalis Is a Better Competitor Than Other Lactic Acid Bacteria in the Initial Colonization of Colon of Healthy Newborn Babies at First Week of Their Life. Front Microbiol 2020; 11:2017. [PMID: 33133027 PMCID: PMC7550472 DOI: 10.3389/fmicb.2020.02017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/30/2020] [Indexed: 11/18/2022] Open
Abstract
Initial colonization of human gut by bacteria is an important step in controlling its microbiota and health status. This study followed the initial colonization by lactic acid bacteria (LAB) in colon of new born babies through following its occurrence in their stool at first week of their life. The LAB occurrence in the neonates' stool was followed on MRS agar medium. The isolated LAB from male and female newborn babies of normal birth and cesarean section surgical delivery were molecular biologically identified by phylogenetic analysis of 16S rRNA gene sequence. From the 24 investigated newborn babies, three LAB taxa, Lactobacillaceae, Enterococcus, and Streptococcus, were detected in their stool at first week of their life. Lactobacillaceae represented 20.8% of total colonized LAB in newborn babies in the culture-dependent approach used in this study and included three species namely Limosilactobacillus reuteri (previously known as Lactobacillus reuteri), Lacticaseibacillus rhamnosus (previously known as Lactobacillus rhamnosus) and Ligilactobacillus agilis (previously known as Lactobacillus agilis). Enterococcus faecalis and E. faecium were detected where E. faecalis was the highest dominant, representing 62.5% of total LAB colonizing newborn babies. This result suggests that this bacterium has high potency for colonization and might be important for controlling the initial settlement of microbiota in healthy newborn babies. Only one species of Streptococcus namely Streptococcus agalactiae was detected in 8.33% total of the investigated newborn babies indicating high competency by other LAB for colonization and that this bacteria, in spite of its pathogenicity, is commensal in its low existence in healthy babies. The explored potency of natural initial colonization of the LAB species E. faecalis, E. faecium, L. reuteri, L. rhamnosus, and L. agilis of which many health beneficial strains were previously reported, would be important for future applications. Despite the controversy in evaluating its health benefits, E. faecalis as a potent competitor to other LAB refers to its importance in initial colonization of healthy babies colon at first week of their life. Further future studies, with more number of samples and characterization, would be of importance for evaluating the potential use of beneficial Enterococcus strains which could improve intestinal ecosystem.
Collapse
Affiliation(s)
- Mohammad Al-Balawi
- Biology Department, Faculty of Science, Taibah University, Medina, Saudi Arabia
| | - Fatthy Mohamed Morsy
- Biology Department, Faculty of Science, Taibah University, Medina, Saudi Arabia
- Bacteriology Section, Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut, Egypt
| |
Collapse
|
16
|
Evrensel A, Ünsalver BÖ, Ceylan ME. Psychobiotics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1192:565-581. [PMID: 31705514 DOI: 10.1007/978-981-32-9721-0_28] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Psychobiotics are live bacteria that directly and indirectly produce positive effects on neuronal functions by colonizing into the intestinal flora. Preliminary studies, although in limited numbers, have found that these bacteria have anxiolytic and antidepressant activities. No research has yet been published on the antipsychotic efficacy of psychobiotics. However, these preliminary studies have opened up new horizons and raised the idea that a new class is emerging in psychopharmacology. About 70 years have passed since the discovery of chlorpromazine, and while the synaptic transmission is understood in almost all details, there seems to be a paradigm shift in psychopharmacology. In recent years, the perspective has shifted from synapse to intestinal microbiota. In this respect, germ-free and conventional animal experiments and few human studies were examined in a comprehensive manner. In this article, after a brief look at the history of contemporary psychopharmacology, the mechanisms of the gut-brain relationship and the evidence of metabolic, systemic, and neuropsychiatric activities of psychobiotics were discussed in detail. In conclusion, psychobiotics seem to have the potential for treatment of neuropsychiatric disorders in the future. However, there are many questions and we do not know the answers yet. We anticipate that the answer to these questions will be given in the near future.
Collapse
Affiliation(s)
- Alper Evrensel
- Department of Psychiatry, Uskudar University, NP Brain Hospital, Saray Mah. Ahmet Tevfik İleri Cad. No: 18 PK, 34768, Umraniye, Istanbul, Turkey.
| | - Barış Önen Ünsalver
- Department of Medical Documentation and Secretariat, Vocational School of Health Services, Uskudar University, Istanbul, Turkey
| | - Mehmet Emin Ceylan
- Departments of Psychology and Philosophy, Uskudar University, Istanbul, Turkey
| |
Collapse
|
17
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 1243] [Impact Index Per Article: 248.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
18
|
Xie Y, Zhou G, Xu Y, He B, Wang Y, Ma R, Chang Y, He D, Xu C, Xiao Z. Effects of Diet Based on IgG Elimination Combined with Probiotics on Migraine Plus Irritable Bowel Syndrome. Pain Res Manag 2019; 2019:7890461. [PMID: 31531150 PMCID: PMC6721378 DOI: 10.1155/2019/7890461] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 07/17/2019] [Accepted: 07/28/2019] [Indexed: 12/31/2022]
Abstract
Several research studies have revealed that migraine has a solid link with gastrointestinal diseases especially irritable bowel syndrome (IBS). This study was carried out to investigate therapeutic potential of diet based on IgG elimination combined with probiotics on migraine plus irritable bowel syndrome. A total of 60 patients diagnosed with migraine plus IBS were recruited for the study. IgG antibodies against 266 food varieties were detected by ELISA. Then, the subjects were randomized into three groups for treatment of IgG elimination diet or probiotics or diet combined with probiotics. Migraine symptom, gut function score, medication use, and serum serotonin level were measured at baseline, 7 weeks, and 14 weeks. Improvement of migraine and gut symptom was achieved at a certain time point. Reduced use of over-the-counter- (OTC-) analgesics was seen in all groups. However, use of triptans did not show significant difference. An increased serum serotonin level was seen in subjects treated with elimination diet and elimination diet combined with probiotics. IgG elimination diet combined with probiotics may be beneficial to migraine plus IBS. It may provide new insight by understanding the intricate relationship between migraine and gastrointestinal diseases.
Collapse
Affiliation(s)
- Yangzhi Xie
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
- Leiyang People's Hospital, Leiyang, Hengyang, Hunan, China
| | - Guijuan Zhou
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Yan Xu
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Bing He
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Yilin Wang
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Rundong Ma
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Yunqian Chang
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Duanqun He
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Chenlin Xu
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| | - Zijian Xiao
- The First Affiliated Hospital of University of South China, University of South China, Hengyang, Hunan, China
| |
Collapse
|
19
|
Abstract
In recent years, interest in the relationship between gut microbiota and disease states has grown considerably. Indeed, several strategies have been employed to modify the microbiome through the administration of different diets, by the administration of antibiotics or probiotics, or even by transplantation of feces. In the present manuscript, we focus specifically on the potential application of probiotics, which seem to be a safe strategy, in the management of digestive, pain, and emotional disorders. We present evidence from animal models and human studies, notwithstanding that translation to clinic still deserves further investigation. The microbiome influences gut functions as well as neurological activity by a variety of mechanisms, which are also discussed. The design and performance of larger trials is urgently needed to verify whether these new strategies might be useful not only for the treatment of disorders affecting the gastrointestinal tract but also in the management of emotional and pain disorders not directly related to the gut.
Collapse
|
20
|
Therapeutic Potential of the Microbiome in the Treatment of Neuropsychiatric Disorders. Med Sci (Basel) 2019; 7:medsci7020021. [PMID: 30709065 PMCID: PMC6410187 DOI: 10.3390/medsci7020021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/26/2019] [Accepted: 01/29/2019] [Indexed: 02/07/2023] Open
Abstract
The search for rational treatment of neuropsychiatric disorders began with the discovery of chlorpromazine in 1951 and continues to evolve. Day by day, new details of the intestinal microbiota–brain axis are coming to light. As the role of microbiota in the etiopathogenesis of neuropsychiatric disorders is more clearly understood, microbiota-based (or as we propose, “fecomodulation”) treatment options are increasingly discussed in the context of treatment. Although their history dates back to ancient times, the importance of psychobiotics and fecal microbiota transplantation (FMT) has only recently been recognized. Despite there being few preclinical and clinical studies, the evidence gathered to this point suggests that consideration of the microbiome in the treatment of neuropsychiatric disorders represents an area of significant therapeutic potential. It is increasingly hoped that such treatment options will be more reliable in terms of their side effects, cost, and ease of implementation. However, there remains much to be researched. Questions will be answered through germ-free animal experiments and randomized controlled trials. In this article, the therapeutic potential of microbiota-based options in the treatment of neuropsychiatric disorders is discussed in light of recent research.
Collapse
|
21
|
Onal EM, Afsar B, Covic A, Vaziri ND, Kanbay M. Gut microbiota and inflammation in chronic kidney disease and their roles in the development of cardiovascular disease. Hypertens Res 2018; 42:123-140. [PMID: 30504819 DOI: 10.1038/s41440-018-0144-z] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 02/06/2023]
Abstract
The health and proper functioning of the cardiovascular and renal systems largely depend on crosstalk in the gut-kidney-heart/vessel triangle. Recent evidence suggests that the gut microbiota has an integral function in this crosstalk. Mounting evidence indicates that the development of chronic kidney and cardiovascular diseases follows chronic inflammatory processes that are affected by the gut microbiota via various immune, metabolic, endocrine, and neurologic pathways. Additionally, deterioration of the function of the cardiovascular and renal systems has been reported to disrupt the original gut microbiota composition, further contributing to the advancement of chronic cardiovascular and renal diseases. Considering the interaction between the gut microbiota and the renal and cardiovascular systems, we can infer that interventions for the gut microbiota through diet and possibly some medications can prevent/stop the vicious cycle between the gut microbiota and the cardiovascular/renal systems, leading to a decrease in chronic cardiovascular and renal diseases.
Collapse
Affiliation(s)
- Emine M Onal
- Department of Medicine, Koc University School of Medicine, Istanbul, Turkey
| | - Baris Afsar
- Department of Medicine, Division of Nephrology, Suleyman Demirel University School of Medicine, Isparta, Turkey
| | - Adrian Covic
- Nephrology Clinic, Dialysis and Renal Transplant Center, 'C.I. PARHON' University Hospital, and 'Grigore T. Popa' University of Medicine, Iasi, Romania
| | - Nosratola D Vaziri
- Division of Nephrology and Hypertension, Schools of Medicine and Biological Science, University of California, California, CA, USA
| | - Mehmet Kanbay
- Department of Medicine, Division of Nephrology, Koc University School of Medicine, Istanbul, Turkey.
| |
Collapse
|
22
|
Bonilla S, Nurko S. Focus on the use of antidepressants to treat pediatric functional abdominal pain: current perspectives. Clin Exp Gastroenterol 2018; 11:365-372. [PMID: 30310301 PMCID: PMC6166750 DOI: 10.2147/ceg.s146646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Chronic abdominal pain is frequently encountered in pediatric practice. A large proportion of cases meet Rome criteria for abdominal pain-functional gastrointestinal disorders (AP-FGIDs). These disorders are costly and, in some cases, lead to impairment of daily functioning and overall quality of life. Pathophysiologic mechanisms include early stressful events, visceral hypersensitivity, dysmotility, changes in intestinal microbiota, and altered central nervous system processing. They are considered disorders of the brain-gut interaction. The diagnosis is made on clinical grounds using symptom-based criteria (Rome criteria). Anxiety and depressive symptoms are more prevalent in patients with AP-FGIDs. Therefore, attention has been directed to the use of neuromodulators as potential interventions for AP-FGIDs. Antidepressants are one type of neuromodulators, and one of the most studied drugs for the management of AP-FGIDs in adult and pediatric population. Data available in pediatric population have significant limitations including nonuniform methodology with different study designs and primary endpoints. Evidence of the efficacy of antidepressants in the management of pediatric AP-FGIDs is not consistent. There is an urgent need for well-designed randomized clinical trials using age-appropriate validated outcome measures. Careful consideration must be given to adverse effects, particularly increased suicidal ideation.
Collapse
Affiliation(s)
- Silvana Bonilla
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital. Boston, MA, USA,
| | - Samuel Nurko
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital. Boston, MA, USA,
| |
Collapse
|
23
|
Mu Q, Tavella VJ, Luo XM. Role of Lactobacillus reuteri in Human Health and Diseases. Front Microbiol 2018; 9:757. [PMID: 29725324 PMCID: PMC5917019 DOI: 10.3389/fmicb.2018.00757] [Citation(s) in RCA: 393] [Impact Index Per Article: 65.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri (L. reuteri) is a well-studied probiotic bacterium that can colonize a large number of mammals. In humans, L. reuteri is found in different body sites, including the gastrointestinal tract, urinary tract, skin, and breast milk. The abundance of L. reuteri varies among different individuals. Several beneficial effects of L. reuteri have been noted. First, L. reuteri can produce antimicrobial molecules, such as organic acids, ethanol, and reuterin. Due to its antimicrobial activity, L. reuteri is able to inhibit the colonization of pathogenic microbes and remodel the commensal microbiota composition in the host. Second, L. reuteri can benefit the host immune system. For instance, some L. reuteri strains can reduce the production of pro-inflammatory cytokines while promoting regulatory T cell development and function. Third, bearing the ability to strengthen the intestinal barrier, the colonization of L. reuteri may decrease the microbial translocation from the gut lumen to the tissues. Microbial translocation across the intestinal epithelium has been hypothesized as an initiator of inflammation. Therefore, inflammatory diseases, including those located in the gut as well as in remote tissues, may be ameliorated by increasing the colonization of L. reuteri. Notably, the decrease in the abundance of L. reuteri in humans in the past decades is correlated with an increase in the incidences of inflammatory diseases over the same period of time. Direct supplementation or prebiotic modulation of L. reuteri may be an attractive preventive and/or therapeutic avenue against inflammatory diseases.
Collapse
Affiliation(s)
| | | | - Xin M. Luo
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
24
|
Martin CR, Osadchiy V, Kalani A, Mayer EA. The Brain-Gut-Microbiome Axis. Cell Mol Gastroenterol Hepatol 2018; 6:133-148. [PMID: 30023410 PMCID: PMC6047317 DOI: 10.1016/j.jcmgh.2018.04.003] [Citation(s) in RCA: 689] [Impact Index Per Article: 114.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 04/04/2018] [Indexed: 12/12/2022]
Abstract
Preclinical and clinical studies have shown bidirectional interactions within the brain-gut-microbiome axis. Gut microbes communicate to the central nervous system through at least 3 parallel and interacting channels involving nervous, endocrine, and immune signaling mechanisms. The brain can affect the community structure and function of the gut microbiota through the autonomic nervous system, by modulating regional gut motility, intestinal transit and secretion, and gut permeability, and potentially through the luminal secretion of hormones that directly modulate microbial gene expression. A systems biological model is proposed that posits circular communication loops amid the brain, gut, and gut microbiome, and in which perturbation at any level can propagate dysregulation throughout the circuit. A series of largely preclinical observations implicates alterations in brain-gut-microbiome communication in the pathogenesis and pathophysiology of irritable bowel syndrome, obesity, and several psychiatric and neurologic disorders. Continued research holds the promise of identifying novel therapeutic targets and developing treatment strategies to address some of the most debilitating, costly, and poorly understood diseases.
Collapse
Key Words
- 2BA, secondary bile acid
- 5-HT, serotonin
- ANS, autonomic nervous system
- ASD, autism spectrum disorder
- BBB, blood-brain barrier
- BGM, brain-gut-microbiome
- CNS, central nervous system
- ECC, enterochromaffin cell
- EEC, enteroendocrine cell
- FFAR, free fatty acid receptor
- FGF, fibroblast growth factor
- FXR, farnesoid X receptor
- GF, germ-free
- GI, gastrointestinal
- GLP-1, glucagon-like peptide-1
- GPR, G-protein–coupled receptor
- IBS, irritable bowel syndrome
- Intestinal Permeability
- Irritable Bowel Syndrome
- LPS, lipopolysaccharide
- SCFA, short-chain fatty acid
- SPF, specific-pathogen-free
- Serotonin
- Stress
- TGR5, G protein-coupled bile acid receptor
- Trp, tryptophan
Collapse
Affiliation(s)
| | | | | | - Emeran A. Mayer
- Correspondence Address correspondence to: Emeran A. Mayer, MD, G. Oppenheimer Center for Neurobiology of Stress and Resilience, University of California at Los Angeles, MC737818-10833 Le Conte Avenue, Los Angeles, California 90095-7378. fax: (310) 825-1919.
| |
Collapse
|
25
|
Ait-Belgnaoui A, Payard I, Rolland C, Harkat C, Braniste V, Théodorou V, Tompkins TA. Bifidobacterium longum and Lactobacillus helveticus Synergistically Suppress Stress-related Visceral Hypersensitivity Through Hypothalamic-Pituitary-Adrenal Axis Modulation. J Neurogastroenterol Motil 2018; 24:138-146. [PMID: 29291614 PMCID: PMC5753912 DOI: 10.5056/jnm16167] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 06/13/2017] [Accepted: 07/12/2017] [Indexed: 12/13/2022] Open
Abstract
Background/Aims Visceral pain and hypothalamic-pituitary-adrenal axis (HPA) dysregulation is a common characteristic in irritable bowel syndrome (IBS) patients. Previously, we reported that a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) prevents chronic stress-mediated brain function abnormalities by attenuating the HPA axis response. Here, we compared the effect between different probiotic treatments on the perception of visceral pain during colorectal distension (CRD) following a chronic stress and the consequences to the activity of the HPA axis. Methods After a 2-week treatment with a combined probiotic formulation, or L. helveticus or B. longum alone in stressed mice, the visceral pain in response to CRD was recorded. The expression of glucocorticoid receptors was determined in the different brain areas involved in the stress response (hypothalamus, hippocampus, and prefrontal cortex). The plasma levels of stress hormones were also measured. Results A pretreatment using the combination of probiotic formulation significantly reduces the chronic stress-induced visceral hypersensitivity respectively at 0.06, 0.08, and 0.10 mL CRD volume. However, a single probiotic (B. longum or L. helveticus) administration is less effective in reducing visceral pain in stressed mice. Moreover, the expression of the glucocorticoid receptor mRNA was consistently up-regulated in several brain areas after pretreatment with a combined probiotic, which correlated with the normalization of stress response compared to the inconsistent effects of a single probiotic. Conclusion The combination of L. helveticus and B. longum is more effective in regulating glucocorticoid negative feedback on the HPA axis than probiotic alone and subsequently in treating stress-induced visceral pain.
Collapse
Affiliation(s)
- Afifa Ait-Belgnaoui
- Neuro-Gastroenterology and Nutrition team, TOXALIM, UMR 1331-INRA/INP/UPS, Toulouse, France.,Lallemand Health Solutions, Montreal, Canada
| | - Isabelle Payard
- Neuro-Gastroenterology and Nutrition team, TOXALIM, UMR 1331-INRA/INP/UPS, Toulouse, France
| | | | - Cherryl Harkat
- Neuro-Gastroenterology and Nutrition team, TOXALIM, UMR 1331-INRA/INP/UPS, Toulouse, France
| | - Viorica Braniste
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Vassillia Théodorou
- Neuro-Gastroenterology and Nutrition team, TOXALIM, UMR 1331-INRA/INP/UPS, Toulouse, France
| | | |
Collapse
|
26
|
Sun Z, Wei W, Liu H, Ma J, Hu M, Huang H. Acute Response of Neurons: An Early Event of Neuronal Cell Death After Facial Nerve Injury. World Neurosurg 2018; 109:e252-e257. [DOI: 10.1016/j.wneu.2017.09.157] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 09/22/2017] [Accepted: 09/23/2017] [Indexed: 01/22/2023]
|
27
|
Maragkoudaki M, Chouliaras G, Orel R, Horvath A, Szajewska H, Papadopoulou A. Lactobacillus reuteri DSM 17938 and a placebo both significantly reduced symptoms in children with functional abdominal pain. Acta Paediatr 2017; 106:1857-1862. [PMID: 28712129 DOI: 10.1111/apa.13992] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 06/06/2017] [Accepted: 07/12/2017] [Indexed: 02/06/2023]
Abstract
AIM Lactobacillus reuteri is a Gram-positive bacterium that naturally inhabits the human intestinal tract. This study assessed how effectively the probiotic L. reuteri DSM 17938 managed childhood functional abdominal pain (FAP). METHODS We recruited 54 children with a mean age 9.1 ± 3.8 years, who were diagnosed with FAP in the outpatient clinics of three university hospitals in Greece, Slovenia and Poland, according to the Rome III criteria, from January 2013 to December 2015. They were randomly assigned to receive either 2 × 108 colony-forming units of L. reuteri (n = 27) or a placebo (n = 27) for four weeks. RESULTS Both L. reuteri and the placebo significantly reduced the frequency and intensity of abdominal pain episodes at four and eight weeks compared to baseline (all p < 0.001). L. reuteri decreased the use of pain relieving drugs at four weeks and the number of child school and adult work absences at four and eight weeks, unlike the placebo, which achieved nonsignificant results. However, the difference between the groups did not reach significance. No side effects were recorded. CONCLUSION Both L. reuteri and the placebo were effective in alleviating pain in children with FAP, but only L. reuteri improved the child's and family's normal activities.
Collapse
Affiliation(s)
- M Maragkoudaki
- Division of Gastroenterology and Hepatology; First Department of Pediatrics; University of Athens; Children's Hospital Agia Sofia; Athens Greece
| | - G Chouliaras
- Division of Gastroenterology and Hepatology; First Department of Pediatrics; University of Athens; Children's Hospital Agia Sofia; Athens Greece
| | - R Orel
- Department of Gastroenterology, Hepatology and Nutrition; Children's Hospital; University Medical Centre; Medical Faculty; University of Ljubljana; Ljubljana Slovenia
| | - A Horvath
- Department of Paediatrics; The Medical University of Warsaw; Warsaw Poland
| | - H Szajewska
- Department of Paediatrics; The Medical University of Warsaw; Warsaw Poland
| | - A Papadopoulou
- Division of Gastroenterology and Hepatology; First Department of Pediatrics; University of Athens; Children's Hospital Agia Sofia; Athens Greece
| |
Collapse
|
28
|
Shiro Y, Arai YC, Ikemoto T, Hayashi K. Stool consistency is significantly associated with pain perception. PLoS One 2017; 12:e0182859. [PMID: 28793322 PMCID: PMC5549932 DOI: 10.1371/journal.pone.0182859] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 07/25/2017] [Indexed: 12/12/2022] Open
Abstract
Background Commensal as well as pathogenic bacteria can influence a variety of gut functions, thereby leading to constipation and diarrhea in severe cases. In fact, several researchers have reported evidence supporting the association between stool consistency or constipation and the Gut microbiome (GM) composition and dysbiosis. GM influences the human health and disease via the gut-brain axis. We thus hypothesized that the pathogenic bacteria increases pain perception to some extent, which means that there could be an association between stool consistency or constipation and pain perception of healthy subjects. Design Observational study. Objectives The aim of the present study was to investigate the association between stool consistency or constipation and pain perception of healthy subjects. Methods Thirty-eight healthy subjects participated in this study. The participants were assessed on their usual stool form (the Bristol Stool Form Scale: BSFS), constipation (the Cleveland Clinic Constipation score: CCS), degree of obesity, pain perception by mechanical stimulus, cold pain threshold, and a questionnaire on psychological state. Results The BSFS was significantly and positively associated with pain perception, and showed a significant association with anxiety states. Furthermore, pain perception was significantly associated with anxiety states. However, there were no significant associations between the CCS and any independent variables. In addition, we found that a significant predictor to the pain perception was BSFS. Moreover, there were significant relationships among the psychological states, BSFS and obesity. Conclusion These results suggest that the stool form is associated with pain perception and anxiety status.
Collapse
Affiliation(s)
- Yukiko Shiro
- Department of Physical Therapy, Faculty of Rehabilitation Sciences, Nagoya Gakuin University, Nagoya, Japan
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Japan
| | - Young-Chang Arai
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Japan
- Institute of Physical Fitness, Sports Medicine and Rehabilitation, School of Medicine, Aichi Medical University, Nagakute, Japan
- * E-mail:
| | - Tatsunori Ikemoto
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Japan
- Institute of Physical Fitness, Sports Medicine and Rehabilitation, School of Medicine, Aichi Medical University, Nagakute, Japan
| | - Kazuhiro Hayashi
- Multidisciplinary Pain Center, Aichi Medical University, Nagakute, Japan
- Institute of Rehabilitation, Aichi Medical University Hospital, Nagakute, Japan
| |
Collapse
|
29
|
Lai NY, Mills K, Chiu IM. Sensory neuron regulation of gastrointestinal inflammation and bacterial host defence. J Intern Med 2017; 282:5-23. [PMID: 28155242 PMCID: PMC5474171 DOI: 10.1111/joim.12591] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Sensory neurons in the gastrointestinal tract have multifaceted roles in maintaining homeostasis, detecting danger and initiating protective responses. The gastrointestinal tract is innervated by three types of sensory neurons: dorsal root ganglia, nodose/jugular ganglia and intrinsic primary afferent neurons. Here, we examine how these distinct sensory neurons and their signal transducers participate in regulating gastrointestinal inflammation and host defence. Sensory neurons are equipped with molecular sensors that enable neuronal detection of diverse environmental signals including thermal and mechanical stimuli, inflammatory mediators and tissue damage. Emerging evidence shows that sensory neurons participate in host-microbe interactions. Sensory neurons are able to detect pathogenic and commensal bacteria through specific metabolites, cell-wall components, and toxins. Here, we review recent work on the mechanisms of bacterial detection by distinct subtypes of gut-innervating sensory neurons. Upon activation, sensory neurons communicate to the immune system to modulate tissue inflammation through antidromic signalling and efferent neural circuits. We discuss how this neuro-immune regulation is orchestrated through transient receptor potential ion channels and sensory neuropeptides including substance P, calcitonin gene-related peptide, vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide. Recent studies also highlight a role for sensory neurons in regulating host defence against enteric bacterial pathogens including Salmonella typhimurium, Citrobacter rodentium and enterotoxigenic Escherichia coli. Understanding how sensory neurons respond to gastrointestinal flora and communicate with immune cells to regulate host defence enhances our knowledge of host physiology and may form the basis for new approaches to treat gastrointestinal diseases.
Collapse
Affiliation(s)
- N Y Lai
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - K Mills
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| | - I M Chiu
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Kelly JR, Allen AP, Temko A, Hutch W, Kennedy PJ, Farid N, Murphy E, Boylan G, Bienenstock J, Cryan JF, Clarke G, Dinan TG. Lost in translation? The potential psychobiotic Lactobacillus rhamnosus (JB-1) fails to modulate stress or cognitive performance in healthy male subjects. Brain Behav Immun 2017; 61:50-59. [PMID: 27865949 DOI: 10.1016/j.bbi.2016.11.018] [Citation(s) in RCA: 220] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 10/28/2016] [Accepted: 11/16/2016] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Preclinical studies have identified certain probiotics as psychobiotics - live microorganisms with a potential mental health benefit. Lactobacillus rhamnosus (JB-1) has been shown to reduce stress-related behaviour, corticosterone release and alter central expression of GABA receptors in an anxious mouse strain. However, it is unclear if this single putative psychobiotic strain has psychotropic activity in humans. Consequently, we aimed to examine if these promising preclinical findings could be translated to healthy human volunteers. OBJECTIVES To determine the impact of L. rhamnosus on stress-related behaviours, physiology, inflammatory response, cognitive performance and brain activity patterns in healthy male participants. METHODS An 8week, randomized, placebo-controlled, cross-over design was employed. Twenty-nine healthy male volunteers participated. Participants completed self-report stress measures, cognitive assessments and resting electroencephalography (EEG). Plasma IL10, IL1β, IL6, IL8 and TNFα levels and whole blood Toll-like 4 (TLR-4) agonist-induced cytokine release were determined by multiplex ELISA. Salivary cortisol was determined by ELISA and subjective stress measures were assessed before, during and after a socially evaluated cold pressor test (SECPT). RESULTS There was no overall effect of probiotic treatment on measures of mood, anxiety, stress or sleep quality and no significant effect of probiotic over placebo on subjective stress measures, or the HPA response to the SECPT. Visuospatial memory performance, attention switching, rapid visual information processing, emotion recognition and associated EEG measures did not show improvement over placebo. No significant anti-inflammatory effects were seen as assessed by basal and stimulated cytokine levels. CONCLUSIONS L. rhamnosus was not superior to placebo in modifying stress-related measures, HPA response, inflammation or cognitive performance in healthy male participants. These findings highlight the challenges associated with moving promising preclinical studies, conducted in an anxious mouse strain, to healthy human participants. Future interventional studies investigating the effect of this psychobiotic in populations with stress-related disorders are required.
Collapse
Affiliation(s)
- John R Kelly
- APC Microbiome Institute, University College Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - Andrew P Allen
- APC Microbiome Institute, University College Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - Andriy Temko
- Department of Electrical and Electronic Engineering, University College Cork, Ireland
| | - William Hutch
- INFANT Research Centre and Department of Pediatrics & Child Health, University College Cork, Ireland
| | - Paul J Kennedy
- APC Microbiome Institute, University College Cork, Ireland
| | - Niloufar Farid
- Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - Eileen Murphy
- Alimentary Health Ltd., Cork Airport Business Park, Cork, Ireland
| | - Geraldine Boylan
- INFANT Research Centre and Department of Pediatrics & Child Health, University College Cork, Ireland
| | - John Bienenstock
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Canada
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Institute, University College Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Institute, University College Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Ireland.
| |
Collapse
|
31
|
West C, Stanisz AM, Wong A, Kunze WA. Effects of Saccharomyces cerevisiae or boulardii yeasts on acute stress induced intestinal dysmotility. World J Gastroenterol 2016; 22:10532-10544. [PMID: 28082805 PMCID: PMC5192264 DOI: 10.3748/wjg.v22.i48.10532] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Revised: 09/15/2016] [Accepted: 10/10/2016] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the capacity of Saccharomyces cerevisiae (S. cerevisiae) and Saccharomyces boulardii (S. boulardii) yeasts to reverse or to treat acute stress-related intestinal dysmotility.
METHODS Adult Swiss Webster mice were stressed for 1 h in a wire-mesh restraint to induce symptoms of intestinal dysmotility and were subsequently killed by cervical dislocation. Jejunal and colon tissue were excised and placed within a tissue perfusion bath in which S. cerevisiae, S. boulardii, or their supernatants were administered into the lumen. Video recordings of contractility and gut diameter changes were converted to spatiotemporal maps and the velocity, frequency, and amplitude of propagating contractile clusters (PCC) were measured. Motility pre- and post-treatment was compared between stressed animals and unstressed controls.
RESULTS S. boulardii and S. cerevisiae helped to mediate the effects of stress on the small and large intestine. Restraint stress reduced jejunal transit velocity (mm/s) from 2.635 ± 0.316 to 1.644 ± 0.238, P < 0.001 and jejunal transit frequency (Hz) from 0.032 ± 0.008 to 0.016 ± 0.005, P < 0.001. Restraint stress increased colonic transit velocity (mm/s) from 0.864 ± 0.183 to 1.432 ± 0.329, P < 0.001 and frequency to a lesser degree. Luminal application of S. boulardii helped to restore jejunal and colonic velocity towards the unstressed controls; 1.833 ± 0.688 to 2.627 ± 0.664, P < 0.001 and 1.516 ± 0.263 to 1.036 ± 0.21, P < 0.001, respectively. S. cerevisiae also had therapeutic effects on the stressed gut, but was most apparent in the jejunum. S. cerevisiae increased PCC velocity in the stressed jejunum from 1.763 ± 0.397 to 2.017 ± 0.48, P = 0.0031 and PCC frequency from 0.016 ± 0.009 to 0.027 ± 0.007, P < 0.001. S. cerevisiae decreased colon PCC velocity from 1.647 ± 0.187 to 1.038 ± 0.222, P < 0.001. Addition of S. boulardii or S. cerevisiae supernatants also helped to restore motility to unstressed values in similar capacity.
CONCLUSION There is a potential therapeutic role for S. cerevisiae and S. boulardii yeasts and their supernatants in the treatment of acute stress-related gut dysmotility.
Collapse
|
32
|
Sarkar A, Lehto SM, Harty S, Dinan TG, Cryan JF, Burnet PWJ. Psychobiotics and the Manipulation of Bacteria-Gut-Brain Signals. Trends Neurosci 2016; 39:763-781. [PMID: 27793434 PMCID: PMC5102282 DOI: 10.1016/j.tins.2016.09.002] [Citation(s) in RCA: 581] [Impact Index Per Article: 72.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 09/01/2016] [Accepted: 09/06/2016] [Indexed: 02/07/2023]
Abstract
Psychobiotics were previously defined as live bacteria (probiotics) which, when ingested, confer mental health benefits through interactions with commensal gut bacteria. We expand this definition to encompass prebiotics, which enhance the growth of beneficial gut bacteria. We review probiotic and prebiotic effects on emotional, cognitive, systemic, and neural variables relevant to health and disease. We discuss gut–brain signalling mechanisms enabling psychobiotic effects, such as metabolite production. Overall, knowledge of how the microbiome responds to exogenous influence remains limited. We tabulate several important research questions and issues, exploration of which will generate both mechanistic insights and facilitate future psychobiotic development. We suggest the definition of psychobiotics be expanded beyond probiotics and prebiotics to include other means of influencing the microbiome. Psychobiotics are beneficial bacteria (probiotics) or support for such bacteria (prebiotics) that influence bacteria–brain relationships. Psychobiotics exert anxiolytic and antidepressant effects characterised by changes in emotional, cognitive, systemic, and neural indices. Bacteria–brain communication channels through which psychobiotics exert effects include the enteric nervous system and the immune system. Current unknowns include dose-responses and long-term effects. The definition of psychobiotics should be expanded to any exogenous influence whose effect on the brain is bacterially-mediated.
Collapse
Affiliation(s)
- Amar Sarkar
- Department of Experimental Psychology, University of Oxford, Oxford OX1 3UD, UK
| | - Soili M Lehto
- Institute of Clinical Medicine/Psychiatry, University of Eastern Finland, FI-70211, Kuopio, Finland; Department of Psychiatry, Kuopio University Hospital, FI-70211, Kuopio, Finland
| | - Siobhán Harty
- Department of Experimental Psychology, University of Oxford, Oxford OX1 3UD, UK
| | - Timothy G Dinan
- Department of Psychiatry, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | |
Collapse
|
33
|
de Meij TGJ, de Groot EFJ, Eck A, Budding AE, Kneepkens CMF, Benninga MA, van Bodegraven AA, Savelkoul PHM. Characterization of Microbiota in Children with Chronic Functional Constipation. PLoS One 2016; 11:e0164731. [PMID: 27760208 PMCID: PMC5070844 DOI: 10.1371/journal.pone.0164731] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/29/2016] [Indexed: 12/18/2022] Open
Abstract
Objectives Disruption of the intestinal microbiota is considered an etiological factor in pediatric functional constipation. Scientifically based selection of potential beneficial probiotic strains in functional constipation therapy is not feasible due to insufficient knowledge of microbiota composition in affected subjects. The aim of this study was to describe microbial composition and diversity in children with functional constipation, compared to healthy controls. Study Design Fecal samples from 76 children diagnosed with functional constipation according to the Rome III criteria (median age 8.0 years; range 4.2–17.8) were analyzed by IS-pro, a PCR-based microbiota profiling method. Outcome was compared with intestinal microbiota profiles of 61 healthy children (median 8.6 years; range 4.1–17.9). Microbiota dissimilarity was depicted by principal coordinate analysis (PCoA), diversity was calculated by Shannon diversity index. To determine the most discriminative species, cross validated logistic ridge regression was performed. Results Applying total microbiota profiles (all phyla together) or per phylum analysis, no disease-specific separation was observed by PCoA and by calculation of diversity indices. By ridge regression, however, functional constipation and controls could be discriminated with 82% accuracy. Most discriminative species were Bacteroides fragilis, Bacteroides ovatus, Bifidobacterium longum, Parabacteroides species (increased in functional constipation) and Alistipes finegoldii (decreased in functional constipation). Conclusions None of the commonly used unsupervised statistical methods allowed for microbiota-based discrimination of children with functional constipation and controls. By ridge regression, however, both groups could be discriminated with 82% accuracy. Optimization of microbiota-based interventions in constipated children warrants further characterization of microbial signatures linked to clinical subgroups of functional constipation.
Collapse
Affiliation(s)
- Tim G. J. de Meij
- Department of Pediatric Gastroenterology, VU University Medical Center, Amsterdam, The Netherlands
- * E-mail:
| | - Evelien F. J. de Groot
- Department of Gastroenterology and Hepatology, VU University Medical Center, Amsterdam, The Netherlands
| | - Anat Eck
- Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - Andries E. Budding
- Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - C. M. Frank Kneepkens
- Department of Pediatric Gastroenterology, VU University Medical Center, Amsterdam, The Netherlands
| | - Marc A. Benninga
- Department of Pediatric Gastroenterology, Emma Children’s Hospital, Academic Medical Center, Amsterdam, The Netherlands
| | - Adriaan A. van Bodegraven
- Department of Gastroenterology and Hepatology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Gastroenterology, Geriatrics, Internal and Intensive Care Medicine (Co-MIK), Zuyderland Medical Center, Heerlen-Sittard-Geleen, The Netherlands
| | - Paul H. M. Savelkoul
- Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
- Department of Medical Microbiology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
34
|
Kao ACC, Harty S, Burnet PWJ. The Influence of Prebiotics on Neurobiology and Behavior. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 131:21-48. [PMID: 27793220 DOI: 10.1016/bs.irn.2016.08.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Manipulating the intestinal microbiota for the benefit of the brain is a concept that has become widely acknowledged. Prebiotics are nondigestible nutrients (i.e., fibers, carbohydrates, or various saccharides) that proliferate intrinsic, beneficial gut bacteria, and so provide an alternative strategy for effectively altering the enteric ecosystem, and thence brain function. Rodent studies demonstrating neurobiological changes following prebiotic intake are slowly emerging, and have thus far revealed significant benefits in disease models, including antiinflammatory and neuroprotective actions. There are also compelling data showing the robust and favorable effects of prebiotics on several behavioral paradigms including, anxiety, learning, and memory. At present, studies in humans are limited, though there is strong evidence for prebiotics modulating emotional processes and the neuroendocrine stress response that may underlie the pathophysiology of anxiety. While the mechanistic details linking the enteric microbiota to the central nervous system remain to be elucidated, there are a number of considerations that can guide future studies. These include the modulation of intestinal endocrine systems and inflammatory cascades, as well as direct interaction with the enteric nervous system and gut mucosa. Our knowledge of gut microbiome-brain communication is steadily progressing, and thorough investigations validating the use of prebiotics in the treatment of neuropsychiatric disorders would be highly valued and are encouraged.
Collapse
Affiliation(s)
- A C C Kao
- University of Oxford, Oxford, United Kingdom
| | - S Harty
- University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
35
|
Perez-Burgos A, Wang L, McVey Neufeld KA, Mao YK, Ahmadzai M, Janssen LJ, Stanisz AM, Bienenstock J, Kunze WA. The TRPV1 channel in rodents is a major target for antinociceptive effect of the probiotic Lactobacillus reuteri DSM 17938. J Physiol 2015; 593:3943-57. [PMID: 26084409 DOI: 10.1113/jp270229] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022] Open
Abstract
Certain probiotic bacteria have been shown to reduce distension-dependent gut pain, but the mechanisms involved remain obscure. Live luminal Lactobacillus reuteri (DSM 17938) and its conditioned medium dose dependently reduced jejunal spinal nerve firing evoked by distension or capsaicin, and 80% of this response was blocked by a specific TRPV1 channel antagonist or in TRPV1 knockout mice. The specificity of DSM action on TRPV1 was further confirmed by its inhibition of capsaicin-induced intracellular calcium increases in dorsal root ganglion neurons. Another lactobacillus with ability to reduce gut pain did not modify this response. Prior feeding of rats with DSM inhibited the bradycardia induced by painful gastric distension. These results offer a system for the screening of new and improved candidate bacteria that may be useful as novel therapeutic adjuncts in gut pain. Certain bacteria exert visceral antinociceptive activity, but the mechanisms involved are not determined. Lactobacillus reuteri DSM 17938 was examined since it may be antinociceptive in children. Since transient receptor potential vanilloid 1 (TRPV1) channel activity may mediate nociceptive signals, we hypothesized that TRPV1 current is inhibited by DSM. We tested this by examining the effect of DSM on the firing frequency of spinal nerve fibres in murine jejunal mesenteric nerve bundles following serosal application of capsaicin. We also measured the effects of DSM on capsaicin-evoked increase in intracellular Ca(2+) or ionic current in dorsal root ganglion (DRG) neurons. Furthermore, we tested the in vivo antinociceptive effects of oral DSM on gastric distension in rats. Live DSM reduced the response of capsaicin- and distension-evoked firing of spinal nerve action potentials (238 ± 27.5% vs. 129 ± 17%). DSM also reduced the capsaicin-evoked TRPV1 ionic current in DRG neuronal primary culture from 83 ± 11% to 41 ± 8% of the initial response to capsaicin only. Another lactobacillus (Lactobacillus rhamnosus JB-1) with known visceral anti-nociceptive activity did not have these effects. DSM also inhibited capsaicin-evoked Ca(2+) increase in DRG neurons; an increase in Ca(2+) fluorescence intensity ratio of 2.36 ± 0.31 evoked by capsaicin was reduced to 1.25 ± 0.04. DSM releasable products (conditioned medium) mimicked DSM inhibition of capsaicin-evoked excitability. The TRPV1 antagonist 6-iodonordihydrocapsaicin or the use of TRPV1 knock-out mice revealed that TRPV1 channels mediate about 80% of the inhibitory effect of DSM on mesenteric nerve response to high intensity gut distension. Finally, feeding with DSM inhibited perception in rats of painful gastric distension. Our results identify a specific target channel for a probiotic with potential therapeutic properties.
Collapse
Affiliation(s)
- Azucena Perez-Burgos
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6
| | - Lu Wang
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
| | - Karen-Anne McVey Neufeld
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6
| | - Yu-Kang Mao
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6
| | - Mustafa Ahmadzai
- Firestone Institute for Respiratory Health, St Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Luke J Janssen
- Firestone Institute for Respiratory Health, St Joseph's Hospital, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Andrew M Stanisz
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6
| | - John Bienenstock
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Wolfgang A Kunze
- McMaster Brain-Body Institute, St Joseph's Healthcare, Hamilton, 50 Charlton Avenue East, Hamilton, Ontario, Canada, L8N 4A6
| |
Collapse
|
36
|
Ten years of subproteome investigations in lactic acid bacteria: A key for food starter and probiotic typing. J Proteomics 2015; 127:332-9. [PMID: 25957532 DOI: 10.1016/j.jprot.2015.04.028] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 04/22/2015] [Accepted: 04/26/2015] [Indexed: 12/29/2022]
Abstract
The definition of safety and efficacy of food-employed bacteria as well as probiotic strains is a continuous, often unattended, challenge. Proteomic techniques such as 2DE, DIGE and LC/LC-MS/MS are suitable and powerful tools to reveal new aspects (positive and negative) of "known" and "unknown" strains that can be employed in food making and as nutraceutical supplements for human health. Unfortunately, these techniques are not used as extensively as it should be wise. The present report describes the most significant results obtained by our research group in 10years of study on subproteomes in bacteria, chiefly lactic acid bacteria. Production of desired and undesired metabolites, differences between strains belonging to same species but isolated from different ecological niches, the effect of cryoprotectants on survival to lyophilization as well as the adhesive capability of strains, were elucidated by analysis of cytosolic, membrane-enriched, surface and extracellular proteomes. The present review opens a window on a yet largely underexplored field and highlights the huge potential of subproteome investigations for more rational choice of microbial strains as food starters, probiotics and for production of nutraceuticals. These analyses will hopefully contribute to manufacturing safer and healthier food and food supplements in the near future. This article is part of a Special Issue entitled: HUPO 2014.
Collapse
|
37
|
Chichlowski M, Rudolph C. Visceral pain and gastrointestinal microbiome. J Neurogastroenterol Motil 2015; 21:172-81. [PMID: 25829337 PMCID: PMC4398233 DOI: 10.5056/jnm15025] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 03/10/2015] [Accepted: 03/11/2015] [Indexed: 12/20/2022] Open
Abstract
A complex set of interactions between the microbiome, gut and brain modulate responses to visceral pain. These interactions occur at the level of the gastrointestinal mucosa, and via local neural, endocrine or immune activity; as well as by the production of factors transported through the circulatory system, like bacterial metabolites or hormones. Various psychological, infectious and other stressors can disrupt this harmonious relationship and alter both the microbiome and visceral pain responses. There are critical sensitive periods that can impact visceral pain responses in adulthood. In this review we provide a brief background of the intestinal microbiome and emerging concepts of the bidirectional interactions between the microbiome, gut and brain. We also discuss recent work in animal models, and human clinical trials using prebiotics and probiotics that alter the microbiome with resultant alterations in visceral pain responses.
Collapse
Affiliation(s)
| | - Colin Rudolph
- Mead Johnson Nutrition, Evansville, IN, USA.,Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California, San Francisco, CA, USA
| |
Collapse
|
38
|
Abstract
Tremendous progress has been made in characterizing the bidirectional interactions between the central nervous system, the enteric nervous system, and the gastrointestinal tract. A series of provocative preclinical studies have suggested a prominent role for the gut microbiota in these gut-brain interactions. Based on studies using rodents raised in a germ-free environment, the gut microbiota appears to influence the development of emotional behavior, stress- and pain-modulation systems, and brain neurotransmitter systems. Additionally, microbiota perturbations by probiotics and antibiotics exert modulatory effects on some of these measures in adult animals. Current evidence suggests that multiple mechanisms, including endocrine and neurocrine pathways, may be involved in gut microbiota-to-brain signaling and that the brain can in turn alter microbial composition and behavior via the autonomic nervous system. Limited information is available on how these findings may translate to healthy humans or to disease states involving the brain or the gut/brain axis. Future research needs to focus on confirming that the rodent findings are translatable to human physiology and to diseases such as irritable bowel syndrome, autism, anxiety, depression, and Parkinson's disease.
Collapse
|
39
|
Gong L, Gao F, Li J, Li J, Yu X, Ma X, Zheng W, Cui S, Liu K, Zhang M, Kunze W, Liu CY. Oxytocin-induced membrane hyperpolarization in pain-sensitive dorsal root ganglia neurons mediated by Ca(2+)/nNOS/NO/KATP pathway. Neuroscience 2015; 289:417-28. [PMID: 25617653 DOI: 10.1016/j.neuroscience.2014.12.058] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 12/11/2014] [Accepted: 12/16/2014] [Indexed: 12/27/2022]
Abstract
Oxytocin (OT) plays an important role in pain modulation and antinociception in the central nervous system. However, little is known about its peripheral effects. This study was conducted to investigate the effect of OT on the electrical properties of neurons in the dorsal root ganglia (DRG) and the underlying mechanisms. DRG neurons from adult rats were acutely dissociated and cultured. Intracellular Ca(2+) was determined by fluorescent microscopy using an indicator dye. The electrical properties of DRG neurons were tested by patch-clamp recording. The oxytocin receptor (OTR) and neuronal nitric oxide synthase (nNOS) on DRG neurons were assessed with immunofluorescence assays. OTR co-localized with nNOS in most of Isolectin B4 (IB4)-binding cultured DRG neurons in rats. OT decreased the excitability, increased the outward current, and evoked the membrane hyperpolarization in cultured DRG neurons. Sodium nitroprusside (SNP), the donor of nitric oxide (NO), exerted similar effects as OT on the membrane potential of cultured DRG neurons. OT increased the production of NO in DRGs and cultured DRG neurons. Pre-treatment of the OTR antagonist atosiban or the selective nNOS inhibitor N-Propyl-l-arginine (NPLA) significantly attenuated the hyperpolarization effect evoked by OT. OT produced a concentration-dependent increase in intracellular Ca(2+) in DRG neurons that responds to capsaicin, which can be attenuated by atosiban, but not by NPLA. OT-evoked membrane hyperpolarization and increase of outward current were distinctly attenuated by glibenclamide, a blocker of ATP-sensitive K(+) (KATP) channel. OT might be an endogenous antinociceptive agent and the peripheral antinociceptive effects of OT are mediated by activation of the Ca(2+)/nNOS/NO/KATP pathway in DRG neurons.
Collapse
Affiliation(s)
- L Gong
- Department of Physiology, Shandong University School of Medicine, China
| | - F Gao
- Department of Physiology, Shandong University School of Medicine, China
| | - J Li
- Department of Physiology, Shandong University School of Medicine, China
| | - J Li
- Department of Physiology, Shandong University School of Medicine, China
| | - X Yu
- Department of Physiology, Shandong University School of Medicine, China
| | - X Ma
- Department of Physiology, Shandong University School of Medicine, China
| | - W Zheng
- Department of Physiology, Shandong University School of Medicine, China
| | - S Cui
- Department of Physiology, Shandong University School of Medicine, China
| | - K Liu
- Department of Physiology, Shandong University School of Medicine, China
| | - M Zhang
- Department of Biology, McMaster University, Canada
| | - W Kunze
- The McMaster Brain-Body Institutes, St Joseph's Healthcare, Hamilton, Ontario, Canada
| | - C Y Liu
- Department of Physiology, Shandong University School of Medicine, China; Provincial Key Lab of Mental Disorder, Shandong University School of Medicine, China.
| |
Collapse
|
40
|
Kannampalli P, Pochiraju S, Chichlowski M, Berg BM, Rudolph C, Bruckert M, Miranda A, Sengupta JN. Probiotic Lactobacillus rhamnosus GG (LGG) and prebiotic prevent neonatal inflammation-induced visceral hypersensitivity in adult rats. Neurogastroenterol Motil 2014; 26:1694-704. [PMID: 25298006 DOI: 10.1111/nmo.12450] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 09/09/2014] [Indexed: 02/06/2023]
Abstract
BACKGROUND Increasing evidence indicates a positive effect of probiotics on the nervous system. The objective of this study was to determine if probiotic Lactobacillus rhamnosus GG (LGG) and/or prebiotics polydextrose/galactooligosaccharide (PDX/GOS) can alter the colonic sensitivity in a neonatal rat model of chronic visceral hyperalgesia and to determine whether altered sensitivity is associated with changes in neurotransmitter levels in the brain. METHODS Chronic visceral hyperalgesia was induced in rats by intracolonic administration of zymosan for 3 days during postnatal day 14-16 (P14-P16). After weaning (P21), these pups were divided into groups that received either (1) control diet (CD), (2) PDX/GOS, (3) LGG, or (4) PDX/GOS + LGG. These diets were continued until visceral sensitivity was tested at P60. The viscero-motor response (VMR) to graded colorectal distension (CRD) was determined by measuring the electromyographic (EMG) activity from the abdominal external oblique muscles. The levels of neurotransmitters and biogenic amines were quantified in the frontal cortex, subcortex, brain stem, and cerebellum. KEY RESULTS At P60, the VMR to CRD in the neonatal zymosan-treated rats was significantly higher than neonatal saline-treated rats. In contrast, neonatal zymosan-treated rats that received PDX/GOS or LGG did not exhibit visceral hyperalgesia. The levels of serotonin, noradrenaline, and dopamine were significantly altered in LGG-treated rats compared to other groups. CONCLUSIONS & INFERENCES Results document that in rats LGG can attenuate neonatally induced chronic visceral pain measured in adulthood. Prolonged intake of LGG alters some key brain neurotransmitters and biogenic amines that could be involved in pain modulation.
Collapse
Affiliation(s)
- P Kannampalli
- Division of Gastroenterology and Hepatology, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Savignac HM, Kiely B, Dinan TG, Cryan JF. Bifidobacteria exert strain-specific effects on stress-related behavior and physiology in BALB/c mice. Neurogastroenterol Motil 2014; 26:1615-27. [PMID: 25251188 DOI: 10.1111/nmo.12427] [Citation(s) in RCA: 286] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 08/17/2014] [Indexed: 02/08/2023]
Abstract
BACKGROUND Accumulating evidence suggests that commensal bacteria consumption has the potential to have a positive impact on stress-related psychiatric disorders. However, the specific bacteria influencing behaviors related to anxiety and depression remain unclear. To this end, we compared the effects of two different Bifidobacteria on anxiety and depression-like behavior; an antidepressant was also used as a comparator. METHODS Innately anxious BALB/c mice received daily Bifidobacterium longum (B.) 1714, B. breve 1205, the antidepressant escitalopram or vehicle treatment for 6 weeks. Behavior was assessed in stress-induced hyperthermia test, marble burying, elevated plus maze, open field, tail suspension test, and forced swim test. Physiological responses to acute stress were also assessed. KEY RESULTS Both Bifidobacteria and escitalopram reduced anxiety in the marble burying test; however, only B. longum 1714 decreased stress-induced hyperthermia. B. breve 1205 induced lower anxiety in the elevated plus maze whereas B. longum 1714 induced antidepressant-like behavior in the tail suspension test. However, there was no difference in corticosterone levels between groups. CONCLUSIONS & INFERENCES These data show that these two Bifidobacteria strains reduced anxiety in an anxious mouse strain. These results also suggest that each bacterial strain has intrinsic effects and may be beneficially specific for a given disorder. These findings strengthen the role of gut microbiota supplementation as psychobiotic-based strategies for stress-related brain-gut axis disorders, opening new avenues in the field of neurogastroenterology.
Collapse
Affiliation(s)
- H M Savignac
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | | | | | | |
Collapse
|
42
|
Romano C, Ferrau' V, Cavataio F, Iacono G, Spina M, Lionetti E, Comisi F, Famiani A, Comito D. Lactobacillus reuteri in children with functional abdominal pain (FAP). J Paediatr Child Health 2014; 50:E68-71. [PMID: 20626584 DOI: 10.1111/j.1440-1754.2010.01797.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
AIM Functional abdominal pain (FAP) is a frequent condition affecting 10-20% of children and can be considered within the classification of functional gastrointestinal disorders (FGID). The objective of this study was to determine the effect of daily supplementation with the probiotic Lactobacillus reuteri DSM 17938 in children with FAP. METHODS The children (aged 6-16 years) were screened for FAP as defined in the Rome III criteria and 60 patients were recruited in this double-blind, randomised, placebo-controlled trial. The children were randomly allocated to receive either L. reuteri (2×10(8) CFU/day) or identical placebo for 4 weeks followed by a 4-week follow-up period without supplementation. Frequency and intensity of pain was self-recorded by the subjects. RESULTS The L. reuteri-supplemented children had significantly lower pain intensity compared with the placebo controls. CONCLUSIONS Supplementation with L. reuteri reduced perceived abdominal pain intensity, which may encourage clinicians to use this probiotic in children with FAP.
Collapse
Affiliation(s)
- Claudio Romano
- Pediatric Department, University of Messina, Messina, Children's Hospital G. Di Cristina, Palermo, Pediatric Department, University of Catania, Catania and Pediatric Unit, Vittoria Hospital, Vittoria, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kianifar H, Ahanchian H, Grover Z, Jafari S, Noorbakhsh Z, Khakshour A, Sedaghat M, Kiani M. Synbiotic in the management of infantile colic: a randomised controlled trial. J Paediatr Child Health 2014; 50:801-5. [PMID: 24962875 DOI: 10.1111/jpc.12640] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/06/2014] [Indexed: 01/09/2023]
Abstract
AIM Infant colic is a frequent problem affecting up to 10-30% of infants in first 3 months of life. Results from previous trials have shown that manipulation of gut microbiota can lead to symptomatic improvements. In a randomised clinical trial, we aimed to determine efficacy of synbiotic in reducing average infant crying time at day 7 and day 30 after starting intervention. METHODS Fifty breastfed infants aged 15-120 days with infantile colic randomly assigned to receive either the synbiotic sachet containing 1 billion CFU of: Lactobacillus casei, L. rhamnosus, Streptococcus thermophilus, Bifidobacterium breve, L. acidophilus, B. infantis, L. bulgaricus and fructooligosacharide (Protexin Healthcare, Somerset, UK), or placebo daily for 30 days. Parents were asked to record details of crying times in a symptoms diary. The primary outcome measure was the treatment success (reduction in the daily crying time >50%) and the secondary outcome measure was symptom resolution (reduction in the daily crying time >90%). RESULTS The treatment success was significantly higher in synbiotic group (82.6%) compared with placebo (35.7%) at day 7 (P < 0.005). At day30, treatment success was 87% and 46% in synbiotic and placebo group, respectively (P < 0.01). Symptom resolution was also higher in synbiotic group (39%) compared with placebo (7%) at day 7 (P < 0.03) but not at day 30 (56% vs.36%, P = 0.24). We encountered no complication related to synbiotic use. CONCLUSION This synbiotic (a mixture of seven probiotic strains plus FOS) significantly improved colic symptoms in comparison with placebo.
Collapse
Affiliation(s)
- Hamidreza Kianifar
- Pediatric Gastroenterology, Mashhad University of Medical Sciences, Mashhad, Khorasan, Iran
| | | | | | | | | | | | | | | |
Collapse
|
44
|
O'Mahony SM, Felice VD, Nally K, Savignac HM, Claesson MJ, Scully P, Woznicki J, Hyland NP, Shanahan F, Quigley EM, Marchesi JR, O'Toole PW, Dinan TG, Cryan JF. Disturbance of the gut microbiota in early-life selectively affects visceral pain in adulthood without impacting cognitive or anxiety-related behaviors in male rats. Neuroscience 2014; 277:885-901. [PMID: 25088912 DOI: 10.1016/j.neuroscience.2014.07.054] [Citation(s) in RCA: 194] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/19/2014] [Accepted: 07/15/2014] [Indexed: 02/08/2023]
Abstract
Disruption of bacterial colonization during the early postnatal period is increasingly being linked to adverse health outcomes. Indeed, there is a growing appreciation that the gut microbiota plays a role in neurodevelopment. However, there is a paucity of information on the consequences of early-life manipulations of the gut microbiota on behavior. To this end we administered an antibiotic (vancomycin) from postnatal days 4-13 to male rat pups and assessed behavioral and physiological measures across all aspects of the brain-gut axis. In addition, we sought to confirm and expand the effects of early-life antibiotic treatment using a different antibiotic strategy (a cocktail of pimaricin, bacitracin, neomycin; orally) during the same time period in both female and male rat pups. Vancomycin significantly altered the microbiota, which was restored to control levels by 8 weeks of age. Notably, vancomycin-treated animals displayed visceral hypersensitivity in adulthood without any significant effect on anxiety responses as assessed in the elevated plus maze or open field tests. Moreover, cognitive performance in the Morris water maze was not affected by early-life dysbiosis. Immune and stress-related physiological responses were equally unaffected. The early-life antibiotic-induced visceral hypersensitivity was also observed in male rats given the antibiotic cocktail. Both treatments did not alter visceral pain perception in female rats. Changes in visceral pain perception in males were paralleled by distinct decreases in the transient receptor potential cation channel subfamily V member 1, the α-2A adrenergic receptor and cholecystokinin B receptor. In conclusion, a temporary disruption of the gut microbiota in early-life results in very specific and long-lasting changes in visceral sensitivity in male rats, a hallmark of stress-related functional disorders of the brain-gut axis such as irritable bowel disorder.
Collapse
Affiliation(s)
- S M O'Mahony
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - V D Felice
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - K Nally
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Biochemistry, University College Cork, Cork, Ireland
| | - H M Savignac
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - M J Claesson
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Microbiology, University College Cork, Cork, Ireland
| | - P Scully
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - J Woznicki
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - N P Hyland
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Pharmacology & Therapeutics, University College Cork, Cork, Ireland
| | - F Shanahan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Medicine, University College Cork, Cork, Ireland
| | - E M Quigley
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - J R Marchesi
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland
| | - P W O'Toole
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Microbiology, University College Cork, Cork, Ireland
| | - T G Dinan
- Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland; Department of Psychiatry, University College Cork, Cork, Ireland
| | - J F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Alimentary Pharmabiotic Centre, University College Cork, Cork, Ireland.
| |
Collapse
|
45
|
The effect of the sympathetic nervous system on splenic natural killer cell activity in mice administered the Lactobacillus pentosus strain S-PT84. Neuroreport 2014; 24:988-91. [PMID: 24165047 PMCID: PMC4047316 DOI: 10.1097/wnr.0000000000000036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Splenic sympathetic nerve activity (SNA) modulates cellular immune functions such as splenic natural killer cell activity. Lactobacillus pentosus strain S-PT84 enhances splenic natural killer cell activity. Here, we examined whether S-PT84 affects splenic natural killer activity through splenic SNA in BALB/c mice. Splenic SNA was significantly decreased following the administration of S-PT84. This phenomenon was inhibited by pretreatment with thioperamide (histamine H3 receptor antagonist), suggesting that S-PT84 directly affected splenic SNA. Thioperamide also inhibited the increase in splenic natural killer activity by S-PT84. Thus, the change in splenic natural killer activity by S-PT84 may be partially modulated through SNA.
Collapse
|
46
|
Sharkey KA, Savidge TC. Reprint of: Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2014; 182:70-82. [PMID: 24674836 DOI: 10.1016/j.autneu.2014.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/11/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
Affiliation(s)
- Keith A Sharkey
- Hotchkiss Brain Institute and Snyder Institute for Chronic Diseases, Department of Physiology & Pharmacology, University of Calgary, Calgary, Alberta, Canada.
| | - Tor C Savidge
- Texas Children's Microbiome Center, Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
47
|
Lu W, Li J, Gong L, Xu X, Han T, Ye Y, Che T, Luo Y, Li J, Zhan R, Yao W, Liu K, Cui S, Liu C. H2 S modulates duodenal motility in male rats via activating TRPV1 and K(ATP) channels. Br J Pharmacol 2014; 171:1534-50. [PMID: 24345161 PMCID: PMC3954491 DOI: 10.1111/bph.12562] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Revised: 11/08/2013] [Accepted: 11/29/2013] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE H2 S induces vasodilatation by opening KATP channels but it may also affect other ion channels. The aim of this study was to investigate the effect of H2 S on intestinal motility in rats and its underlying mechanism. EXPERIMENTAL APPROACH The tension of intestinal muscle strips, afferent firing of intestinal mesenteric nerves, length of duodenal smooth muscle cells and whole-cell membrane potential of dorsal root ganglion (DRG) neurons were monitored. H2 S-producing enzymes were located by immunofluorescence staining. KEY RESULTS NaHS exerted early transient excitation and late long-lasting inhibition on the intestinal contraction. The excitation was attenuated by TRPV1 antagonists capsazepine, A784168, SB-366791 and NK1 receptor antagonist L703606, while the inhibition was attenuated by glibenclamide. NaHS increased duodenal afferent nerve firing and depolarized DRG neurons. These effects were reduced by capsazepine and A784168. NaHS relaxed isolated duodenal smooth muscle cells. The KATP channels were expressed in smooth muscle cells. Cystathionine β-synthase and cystathionine γ-lyase were expressed in rat duodenal myenteric neurons. L-cysteine and S-adenosyl-L-methionine increased the contraction of duodenal muscle strips, an effect attenuated by capsazepine and L703606. CONCLUSIONS AND IMPLICATIONS NaHS induces biphasic effects on intestinal motility in rats while endogenous H2 S only exerts an excitatory effect. This transient excitatory effect might be mediated by activation of TRPV1 channels in sensory nerve terminals with the consequent release of substance P. The long-lasting inhibitory effect might be mediated by activation of KATP channels in the smooth muscle cells. These findings reveal a novel mechanism for the excitatory effect of H2 S on gastrointestinal motility.
Collapse
Affiliation(s)
- Wen Lu
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Jing Li
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Liping Gong
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Xiaomeng Xu
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Ting Han
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Yanfang Ye
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Tongtong Che
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Yan Luo
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Jingxin Li
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Renzhi Zhan
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Wei Yao
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Kejing Liu
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Shuang Cui
- Department of Physiology, Shandong University School of MedicineJinan, China
| | - Chuanyong Liu
- Department of Physiology, Shandong University School of MedicineJinan, China
- Provincial Key Lab of Mental Disorder, Shandong University School of MedicineJinan, China
| |
Collapse
|
48
|
Neuropeptides and the microbiota-gut-brain axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 817:195-219. [PMID: 24997035 DOI: 10.1007/978-1-4939-0897-4_9] [Citation(s) in RCA: 282] [Impact Index Per Article: 28.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropeptides are important mediators both within the nervous system and between neurons and other cell types. Neuropeptides such as substance P, calcitonin gene-related peptide and neuropeptide Y (NPY), vasoactive intestinal polypeptide, somatostatin and corticotropin-releasing factor are also likely to play a role in the bidirectional gut-brain communication. In this capacity they may influence the activity of the gastrointestinal microbiota and its interaction with the gut-brain axis. Current efforts in elucidating the implication of neuropeptides in the microbiota-gut-brain axis address four information carriers from the gut to the brain (vagal and spinal afferent neurons; immune mediators such as cytokines; gut hormones; gut microbiota-derived signalling molecules) and four information carriers from the central nervous system to the gut (sympathetic efferent neurons; parasympathetic efferent neurons; neuroendocrine factors involving the adrenal medulla; neuroendocrine factors involving the adrenal cortex). Apart from operating as neurotransmitters, many biologically active peptides also function as gut hormones. Given that neuropeptides and gut hormones target the same cell membrane receptors (typically G protein-coupled receptors), the two messenger roles often converge in the same or similar biological implications. This is exemplified by NPY and peptide YY (PYY), two members of the PP-fold peptide family. While PYY is almost exclusively expressed by enteroendocrine cells, NPY is found at all levels of the gut-brain and brain-gut axis. The function of PYY-releasing enteroendocrine cells is directly influenced by short chain fatty acids generated by the intestinal microbiota from indigestible fibre, while NPY may control the impact of the gut microbiota on inflammatory processes, pain, brain function and behaviour. Although the impact of neuropeptides on the interaction between the gut microbiota and brain awaits to be analysed, biologically active peptides are likely to emerge as neural and endocrine messengers in orchestrating the microbiota-gut-brain axis in health and disease.
Collapse
|
49
|
Role of enteric neurotransmission in host defense and protection of the gastrointestinal tract. Auton Neurosci 2013; 181:94-106. [PMID: 24412639 DOI: 10.1016/j.autneu.2013.12.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 12/11/2013] [Indexed: 12/24/2022]
Abstract
Host defense is a vital role played by the gastrointestinal tract. As host to an enormous and diverse microbiome, the gut has evolved an elaborate array of chemical and physicals barriers that allow the digestion and absorption of nutrients without compromising the mammalian host. The control of such barrier functions requires the integration of neural, humoral, paracrine and immune signaling, involving redundant and overlapping mechanisms to ensure, under most circumstances, the integrity of the gastrointestinal epithelial barrier. Here we focus on selected recent developments in the autonomic neural control of host defense functions used in the protection of the gut from luminal agents, and discuss how the microbiota may potentially play a role in enteric neurotransmission. Key recent findings include: the important role played by subepithelial enteric glia in modulating intestinal barrier function, identification of stress-induced mechanisms evoking barrier breakdown, neural regulation of epithelial cell proliferation, the role of afferent and efferent vagal pathways in regulating barrier function, direct evidence for bacterial communication to the enteric nervous system, and microbial sources of enteric neurotransmitters. We discuss these new and interesting developments in our understanding of the role of the autonomic nervous system in gastrointestinal host defense.
Collapse
|
50
|
|