101
|
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: 207.2] [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
|
102
|
Early life stress induces submissive behavior in adult rats. Behav Brain Res 2019; 372:112025. [DOI: 10.1016/j.bbr.2019.112025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Revised: 04/18/2019] [Accepted: 06/08/2019] [Indexed: 12/16/2022]
|
103
|
Long-Smith C, O'Riordan KJ, Clarke G, Stanton C, Dinan TG, Cryan JF. Microbiota-Gut-Brain Axis: New Therapeutic Opportunities. Annu Rev Pharmacol Toxicol 2019; 60:477-502. [PMID: 31506009 DOI: 10.1146/annurev-pharmtox-010919-023628] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The traditional fields of pharmacology and toxicology are beginning to consider the substantial impact our gut microbiota has on host physiology. The microbiota-gut-brain axis is emerging as a particular area of interest and a potential new therapeutic target for effective treatment of central nervous system disorders, in addition to being a potential cause of drug side effects. Microbiota-gut-brain axis signaling can occur via several pathways, including via the immune system, recruitment of host neurochemical signaling, direct enteric nervous system routes and the vagus nerve, and the production of bacterial metabolites. Altered gut microbial profiles have been described in several psychiatric and neurological disorders. Psychobiotics, live biotherapeutics or substances whose beneficial effects on the brain are bacterially mediated, are currently being investigated as direct and/or adjunctive therapies for psychiatric and neurodevelopmental disorders and possibly for neurodegenerative disease, and they may emerge as new therapeutic options in the clinical management of brain disorders.
Collapse
Affiliation(s)
| | | | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; .,Department of Psychiatry & Neurobehavioral Science, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; .,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; .,Department of Psychiatry & Neurobehavioral Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; .,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
104
|
Serra D, Almeida LM, Dinis TCP. Polyphenols in the management of brain disorders: Modulation of the microbiota-gut-brain axis. ADVANCES IN FOOD AND NUTRITION RESEARCH 2019; 91:1-27. [PMID: 32035595 DOI: 10.1016/bs.afnr.2019.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
The modulation of the microbiota-gut-brain axis with a view to preventing and treating brain disorders became recently a hot topic for the scientific community. Dietary polyphenols are multifaceted compounds that have demonstrated to be highly advantageous to counteract inflammation, oxidative stress, and neurodegeneration, among other pathological conditions, being useful in the prevention and treatment of several chronic disorders. The potential of these compounds to prevent and treat brain disorders has not been only related to their capacity to reach the brain, depending on their chemical structure, and interact directly with brain cells, but also to their ability to modulate the communication between the brain and the gut, interfering with multiple branches of this axis. Preclinical studies have demonstrated the potential of these food bioactive compounds in brain diseases, namely, neurodevelopmental, such as Down's syndrome and Autism spectrum disorder, neurodegenerative, such as Parkinson's disease and Alzheimer's disease, and psychiatric disorders, such as depression and anxiety. Until now, dietary polyphenols have been recognized as promising nutraceuticals to combat brain disorders. However, the impact of these compounds on the gut-brain interconnection remains poorly elucidated. Also, clinical assays are crucial to further support the beneficial effects of these compounds as demonstrated in preclinical research.
Collapse
Affiliation(s)
- Diana Serra
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal.
| | - Leonor M Almeida
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Teresa C P Dinis
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
105
|
O'Mahony SM, McVey Neufeld KA, Waworuntu RV, Pusceddu MM, Manurung S, Murphy K, Strain C, Laguna MC, Peterson VL, Stanton C, Berg BM, Dinan TG, Cryan JF. The enduring effects of early-life stress on the microbiota-gut-brain axis are buffered by dietary supplementation with milk fat globule membrane and a prebiotic blend. Eur J Neurosci 2019; 51:1042-1058. [PMID: 31339598 DOI: 10.1111/ejn.14514] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2018] [Revised: 06/12/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022]
Abstract
Nutritional interventions targeting the microbiota-gut-brain axis are proposed to modulate stress-induced dysfunction of physiological processes and brain development. Maternal separation (MS) in rats induces long-term alterations to behaviour, pain responses, gut microbiome and brain neurochemistry. In this study, the effects of dietary interventions (milk fat globule membrane [MFGM] and a polydextrose/galacto-oligosaccharide prebiotic blend) were evaluated. Diets were provided from postnatal day 21 to both non-separated and MS offspring. Spatial memory, visceral sensitivity and stress reactivity were assessed in adulthood. Gene transcripts associated with cognition and stress and the caecal microbiota composition were analysed. MS-induced visceral hypersensitivity was ameliorated by MFGM and to greater extent with the combination of MFGM and prebiotic blend. Furthermore, spatial learning and memory were improved by prebiotics and MFGM alone and with the combination. The prebiotic blend and the combination of the prebiotics and MFGM appeared to facilitate return to baseline with regard to HPA axis response to the restraint stress, which can be beneficial in times where coping mechanisms to stressful events are required. Interestingly, the combination of MFGM and prebiotic reduced the long-term impact of MS on a marker of myelination in the prefrontal cortex. MS affected the microbiota at family level only, while MFGM, the prebiotic blend and the combination influenced abundance at family and genus level as well as influencing beta-diversity levels. In conclusion, intervention with MFGM and prebiotic blend significantly impacted the composition of the microbiota as well as ameliorating some of the long-term effects of early-life stress.
Collapse
Affiliation(s)
- Siobhain M O'Mahony
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | | | | | | | - Kiera Murphy
- Teagasc Food Research Centre, Moorepark, Co., Cork, Ireland
| | - Conall Strain
- Teagasc Food Research Centre, Moorepark, Co., Cork, Ireland
| | - Mamen C Laguna
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | | | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Co., Cork, Ireland
| | - Brian M Berg
- Mead Johnson Pediatric Nutrition Institute, Evansville, IN, USA
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
106
|
Bonaz B, Sinniger V, Pellissier S. Vagus Nerve Stimulation at the Interface of Brain-Gut Interactions. Cold Spring Harb Perspect Med 2019; 9:cshperspect.a034199. [PMID: 30201788 DOI: 10.1101/cshperspect.a034199] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The vagus nerve, a key component of the cross-communication between the gut and the brain, is a major element of homeostasis sensing the "milieu intérieur" and boosting the nervous and endocrine responses to maintain the gastrointestinal health status. This nerve has anti-inflammatory properties regulating the gut through the activation of the hypothalamic-pituitary-adrenal axis and the release of cortisol and through a vagovagal reflex, which has an anti-tumor necrosis factor (TNF) effect called the cholinergic anti-inflammatory pathway. Stimulating this nerve is an interesting tool as a nondrug therapy for the treatment of gastrointestinal diseases in which brain-gut communication is dysfunctional, such as inflammatory bowel disorders and others. This review presents the rationale of vagal gastrointestinal physiology and diseases and the most recent advances in vagus nerve stimulation. It also highlights the main issues to be addressed in the future to improve this bioelectronic therapy for gastrointestinal disorders.
Collapse
Affiliation(s)
- Bruno Bonaz
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Valérie Sinniger
- Division of Hepato-Gastroenterology, Grenoble University Hospital, 38043 Grenoble Cedex 09, France.,U1216, INSERM, GIN, Grenoble Institute of Neurosciences, University Grenoble Alpes, Grenoble, France
| | - Sonia Pellissier
- University Grenoble Alpes, University Savoie Mont Blanc, 38000 Grenoble, France
| |
Collapse
|
107
|
Rincel M, Aubert P, Chevalier J, Grohard PA, Basso L, Monchaux de Oliveira C, Helbling JC, Lévy É, Chevalier G, Leboyer M, Eberl G, Layé S, Capuron L, Vergnolle N, Neunlist M, Boudin H, Lepage P, Darnaudéry M. Multi-hit early life adversity affects gut microbiota, brain and behavior in a sex-dependent manner. Brain Behav Immun 2019; 80:179-192. [PMID: 30872090 DOI: 10.1016/j.bbi.2019.03.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 02/22/2019] [Accepted: 03/09/2019] [Indexed: 12/15/2022] Open
Abstract
The accumulation of adverse events in utero and during childhood differentially increases the vulnerability to psychiatric diseases in men and women. Gut microbiota is highly sensitive to the early environment and has been recently hypothesized to affect brain development. However, the impact of early-life adversity on gut microbiota, notably with regards to sex differences, remains to be explored. We examined the effects of multifactorial early-life adversity on behavior and microbiota composition in C3H/HeN mice of both sexes exposed to a combination of maternal immune activation (lipopolysaccharide injection on embryonic day 17, 120 µg/kg, i.p.), maternal separation (3hr per day from postnatal day (PND)2 to PND14) and maternal unpredictable chronic mild stress. At adulthood, offspring exposed to multi-hit early adversity showed sex-specific behavioral phenotypes with males exhibiting deficits in social behavior and females showing increased anxiety in the elevated plus maze and increased compulsive behavior in the marble burying test. Early adversity also differentially regulated gene expression in the medial prefrontal cortex (mPFC) according to sex. Interestingly, several genes such as Arc, Btg2, Fosb, Egr4 or Klf2 were oppositely regulated by early adversity in males versus females. Finally, 16S-based microbiota profiling revealed sex-dependent gut dysbiosis. In males, abundance of taxa belonging to Lachnospiraceae and Porphyromonadaceae families or other unclassified Firmicutes, but also Bacteroides, Lactobacillus and Alloprevotella genera was regulated by early adversity. In females, the effects of early adversity were limited and mainly restricted to Lactobacillus and Mucispirillum genera. Our work reveals marked sex differences in a multifactorial model of early-life adversity, both on emotional behaviors and gut microbiota, suggesting that sex should systematically be considered in preclinical studies both in neurogastroenterology and psychiatric research.
Collapse
Affiliation(s)
- Marion Rincel
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Philippe Aubert
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Julien Chevalier
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Pierre-Antoine Grohard
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Lilian Basso
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Camille Monchaux de Oliveira
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Jean Christophe Helbling
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Élodie Lévy
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | | | - Marion Leboyer
- Université Paris-est-Créteil, Laboratoire Psychiatrie translationnelle, INSERM U955, Hôpital Chenevier-Mondor, Créteil, France
| | - Gérard Eberl
- Unité Microenvironnement et Immunité, Institut Pasteur, Paris, France
| | - Sophie Layé
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Lucile Capuron
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France
| | - Nathalie Vergnolle
- Institut de Recherche en Santé Digestive, INSERM UMR1220, INRA UMR1416, ENVT, UPS, Toulouse, France
| | - Michel Neunlist
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Hélène Boudin
- The Enteric Nervous System in Gut and Brain Disorders, INSERM UMR1235, IMAD, Nantes, France
| | - Patricia Lepage
- Micalis Institute, INRA, AgroParisTech, Univ. Paris-Saclay, Jouy-en-Josas, France
| | - Muriel Darnaudéry
- Univ. Bordeaux, INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France; INRA, Nutrition and Integrative Neurobiology, UMR 1286, 33076 Bordeaux, France.
| |
Collapse
|
108
|
Abstract
The developmental period constitutes a critical window of sensitivity to stress. Indeed, early-life adversity increases the risk to develop psychiatric diseases, but also gastrointestinal disorders such as the irritable bowel syndrome at adulthood. In the past decade, there has been huge interest in the gut-brain axis, especially as regards stress-related emotional behaviours. Animal models of early-life adversity, in particular, maternal separation (MS) in rodents, demonstrate lasting deleterious effects on both the gut and the brain. Here, we review the effects of MS on both systems with a focus on stress-related behaviours. In addition, we discuss more recent findings showing the impact of gut-directed interventions, including nutrition with pre- and probiotics, illustrating the role played by gut microbiota in mediating the long-term effects of MS. Overall, preclinical studies suggest that nutritional approaches with pro- and prebiotics may constitute safe and efficient strategies to attenuate the effects of early-life stress on the gut-brain axis. Further research is required to understand the complex mechanisms underlying gut-brain interaction dysfunctions after early-life stress as well as to determine the beneficial impact of gut-directed strategies in a context of early-life adversity in human subjects.
Collapse
|
109
|
Ambrosini YM, Borcherding D, Kanthasamy A, Kim HJ, Willette AA, Jergens A, Allenspach K, Mochel JP. The Gut-Brain Axis in Neurodegenerative Diseases and Relevance of the Canine Model: A Review. Front Aging Neurosci 2019; 11:130. [PMID: 31275138 PMCID: PMC6591269 DOI: 10.3389/fnagi.2019.00130] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Accepted: 05/16/2019] [Indexed: 12/13/2022] Open
Abstract
Identifying appropriate animal models is critical in developing translatable in vitro and in vivo systems for therapeutic drug development and investigating disease pathophysiology. These animal models should have direct biological and translational relevance to the underlying disease they are supposed to mimic. Aging dogs not only naturally develop a cognitive decline in many aspects including learning and memory deficits, but they also exhibit human-like individual variability in the aging process. Neurodegenerative processes that can be observed in both human and canine brains include the progressive accumulation of β-amyloid (Aβ) found as diffuse plaques in the prefrontal cortex (PFC), including the gyrus proreus (i.e., medial orbital PFC), as well as the hippocampus and the cerebral vasculature. Tau pathology, a marker of neurodegeneration and dementia progression, was also found in canine hippocampal synapses. Various epidemiological data show that human patients with neurodegenerative diseases have concurrent intestinal lesions, and histopathological changes in the gastrointestinal (GI) tract occurs decades before neurodegenerative changes. Gut microbiome alterations have also been reported in many neurodegenerative diseases including Alzheimer's (AD) and Parkinson's diseases, as well as inflammatory central nervous system (CNS) diseases. Interestingly, the dog gut microbiome more closely resembles human gut microbiome in composition and functional overlap compared to rodent models. This article reviews the physiology of the gut-brain axis (GBA) and its involvement with neurodegenerative diseases in humans. Additionally, we outline the advantages and weaknesses of current in vitro and in vivo models and discuss future research directions investigating major human neurodegenerative diseases such as AD and Parkinson's diseases using dogs.
Collapse
Affiliation(s)
- Yoko M. Ambrosini
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Dana Borcherding
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Anumantha Kanthasamy
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| | - Hyun Jung Kim
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, United States
| | - Auriel A. Willette
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
- Department of Food Science and Human Nutrition, College of Agriculture and Life Sciences, Iowa State University, Ames, IA, United States
| | - Albert Jergens
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Karin Allenspach
- Department of Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Jonathan P. Mochel
- Department of Biomedical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA, United States
| |
Collapse
|
110
|
Rincel M, Olier M, Minni A, Monchaux de Oliveira C, Matime Y, Gaultier E, Grit I, Helbling JC, Costa AM, Lépinay A, Moisan MP, Layé S, Ferrier L, Parnet P, Theodorou V, Darnaudéry M. Pharmacological restoration of gut barrier function in stressed neonates partially reverses long-term alterations associated with maternal separation. Psychopharmacology (Berl) 2019; 236:1583-1596. [PMID: 31147734 DOI: 10.1007/s00213-019-05252-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 04/22/2019] [Indexed: 02/06/2023]
Abstract
RATIONALE Intestinal permeability plays an important role in gut-brain axis communication. Recent studies indicate that intestinal permeability increases in neonate pups during maternal separation (MS). OBJECTIVES The present study aims to determine whether pharmacological inhibition of myosin light chain kinase (MLCK), which regulates tight junction contraction and controls intestinal permeability, in stressed neonates, protects against the long-term effects of MS. METHODS Male Wistar rats were exposed to MS (3 h per day from post-natal day (PND)2 to PND14) or left undisturbed and received daily intraperitoneal injection of a MLCK inhibitor (ML-7, 5 mg/kg) or vehicle during the same period. At adulthood, emotional behaviors, corticosterone response to stress, and gut microbiota composition were analyzed. RESULTS ML-7 restored gut barrier function in MS rats specifically during the neonatal period. Remarkably, ML-7 prevented MS-induced sexual reward-seeking impairment and reversed the alteration of corticosterone response to stress at adulthood. The effects of ML-7 were accompanied by the normalization of the abundance of members of Lachnospiraceae, Clostridiales, Desulfovibrio, Bacteroidales, Enterorhabdus, and Bifidobacterium in the feces of MS rats at adulthood. CONCLUSIONS Altogether, our work suggests that improvement of intestinal barrier defects during development may alleviate some of the long-term effects of early-life stress and provides new insight on brain-gut axis communication in a context of stress.
Collapse
Affiliation(s)
- Marion Rincel
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Maïwenn Olier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Amandine Minni
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | | | - Yann Matime
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Eric Gaultier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Isabelle Grit
- UMR 1280, Institut des maladies de l'appareil digestif, PhAN, INRA, University of Nantes, Nantes, France
| | | | - Anna Maria Costa
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Amandine Lépinay
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Marie-Pierre Moisan
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Sophie Layé
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France
| | - Laurent Ferrier
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Patricia Parnet
- UMR 1280, Institut des maladies de l'appareil digestif, PhAN, INRA, University of Nantes, Nantes, France
| | - Vassilia Theodorou
- Laboratoire Toxalim, UMR 1331, University of Toulouse III (UPS), INP-EI-Purpan, INRA, Toulouse, France
| | - Muriel Darnaudéry
- Univ. Bordeaux, INRA, Bordeaux INP, NutriNeuro, UMR 1286, 33076, Bordeaux, France.
| |
Collapse
|
111
|
Cowan CSM, Richardson R. A Brief Guide to Studying Fear in Developing Rodents: Important Considerations and Common Pitfalls. ACTA ACUST UNITED AC 2019; 83:e44. [PMID: 30040208 DOI: 10.1002/cpns.44] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Development is a time of rapid change that sets the pathway to adult functioning across all aspects of physical and mental health. Developmental studies can therefore offer insight into the unique needs of individuals at different stages of normal development as well as the etiology of various disease states. The aim of this overview is to provide an introduction to the practical implementation of developmental studies in rats and mice, with an emphasis on the study of learned fear. We first discuss how developmental factors may influence experimental outcomes for any study. This is followed by a discussion of methodological issues to consider when conducting studies of developing rodents, highlighting examples from the literature on learned fear. Throughout, we offer some recommendations to guide researchers on best practice in developmental studies. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Rick Richardson
- School of Psychology, The University of New South Wales, Sydney, Australia
| |
Collapse
|
112
|
Kolacz J, Kovacic KK, Porges SW. Traumatic stress and the autonomic brain-gut connection in development: Polyvagal Theory as an integrative framework for psychosocial and gastrointestinal pathology. Dev Psychobiol 2019; 61:796-809. [PMID: 30953358 DOI: 10.1002/dev.21852] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 02/12/2019] [Accepted: 02/23/2019] [Indexed: 12/14/2022]
Abstract
A range of psychiatric disorders such as anxiety, depression, and post-traumatic stress disorder frequently co-occur with functional gastrointestinal (GI) disorders. Risk of these pathologies is particularly high in those with a history of trauma, abuse, and chronic stress. These scientific findings and rising awareness within the healthcare profession give rise to a need for an integrative framework to understand the developmental mechanisms that give rise to these observations. In this paper, we introduce a plausible explanatory framework, based on the Polyvagal Theory (Porges, Psychophysiology, 32, 301-318, 1995; Porges, International Journal of Psychophysiology, 42, 123-146, 2001; Porges, Biological Psychology, 74, 116-143, 2007), which describes how evolution impacted the structure and function of the autonomic nervous system (ANS). The Polyvagal Theory provides organizing principles for understanding the development of adaptive diversity in homeostatic, threat-response, and psychosocial functions that contribute to pathology. Using these principles, we outline possible mechanisms that promote and maintain socioemotional and GI dysfunction and review their implications for therapeutic targets.
Collapse
Affiliation(s)
- Jacek Kolacz
- Traumatic Stress Research Consortium at the Kinsey Institute, Indiana University, Bloomington, Indiana
| | - Katja K Kovacic
- Division of Pediatric Gastroenterology, Hepatology & Nutrition, Department of Pediatrics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Stephen W Porges
- Traumatic Stress Research Consortium at the Kinsey Institute, Indiana University, Bloomington, Indiana.,Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| |
Collapse
|
113
|
Dettmer AM, Allen JM, Jaggers RM, Bailey MT. A descriptive analysis of gut microbiota composition in differentially reared infant rhesus monkeys (Macaca mulatta) across the first 6 months of life. Am J Primatol 2019; 81:e22969. [PMID: 30941799 DOI: 10.1002/ajp.22969] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/13/2019] [Accepted: 02/26/2019] [Indexed: 12/12/2022]
Abstract
The gastrointestinal microbiome is recognized as a critical component in host immune function, physiology, and behavior. Early life experiences that alter diet and social contact also influence these outcomes. Despite the growing number of studies in this area, no studies to date have examined the contribution of early life experiences on the gut microbiome in infants across development. Such studies are important for understanding the biological and environmental factors that contribute to optimal gut microbial colonization and subsequent health. We studied infant rhesus monkeys (Macaca mulatta) across the first 6 months of life that were pseudo-randomly assigned to one of two different rearing conditions at birth: mother-peer-reared (MPR), in which infants were reared in social groups with many other adults and peers and nursed on their mothers, or nursery-reared (NR), in which infants were reared by human caregivers, fed formula, and given daily social contact with peers. We analyzed the microbiome from rectal swabs (total N = 97; MPR = 43, NR = 54) taken on the day of birth and at postnatal Days 14, 30, 90, and 180 using 16S rRNA gene sequencing. Bacterial composition differences were evident as early as 14 days, with MPR infants exhibiting a lower abundance of Bifidobacterium and a higher abundance of Bacteroides than NR infants. The most marked differences were observed at 90 days, when Bifidobacterium, Lactobacillus, Streptococcus, Bacteroides, Clostridium, and Prevotella differed across rearing groups. By Day 180, no differences in the relative abundances of the bacteria of interest were observed. These novel findings in developing primate neonates indicate that the early social environment as well as diet influence gut microbiota composition very early in life. These results also lay the groundwork for mechanistic studies examining the effects of early experiences on gut microbiota across development with the ultimate goal of understanding the clinical significance of developmental changes.
Collapse
Affiliation(s)
- Amanda M Dettmer
- Laboratory of Comparative Ethology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Poolesville, Maryland.,Yale Child Study Center, Yale School of Medicine, New Haven, Connecticut
| | - Jacob M Allen
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Robert M Jaggers
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Michael T Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio.,Department of Pediatrics, The Ohio State University College of Medicine, Columbus, Ohio
| |
Collapse
|
114
|
Glutamatergic Signaling Along The Microbiota-Gut-Brain Axis. Int J Mol Sci 2019; 20:ijms20061482. [PMID: 30934533 PMCID: PMC6471396 DOI: 10.3390/ijms20061482] [Citation(s) in RCA: 196] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/04/2019] [Accepted: 03/21/2019] [Indexed: 12/21/2022] Open
Abstract
A complex bidirectional communication system exists between the gastrointestinal tract and the brain. Initially termed the “gut-brain axis” it is now renamed the “microbiota-gut-brain axis” considering the pivotal role of gut microbiota in maintaining local and systemic homeostasis. Different cellular and molecular pathways act along this axis and strong attention is paid to neuroactive molecules (neurotransmitters, i.e., noradrenaline, dopamine, serotonin, gamma aminobutyric acid and glutamate and metabolites, i.e., tryptophan metabolites), sustaining a possible interkingdom communication system between eukaryota and prokaryota. This review provides a description of the most up-to-date evidence on glutamate as a neurotransmitter/neuromodulator in this bidirectional communication axis. Modulation of glutamatergic receptor activity along the microbiota-gut-brain axis may influence gut (i.e., taste, visceral sensitivity and motility) and brain functions (stress response, mood and behavior) and alterations of glutamatergic transmission may participate to the pathogenesis of local and brain disorders. In this latter context, we will focus on two major gut disorders, such as irritable bowel syndrome and inflammatory bowel disease, both characterized by psychiatric co-morbidity. Research in this area opens the possibility to target glutamatergic neurotransmission, either pharmacologically or by the use of probiotics producing neuroactive molecules, as a therapeutic approach for the treatment of gastrointestinal and related psychiatric disorders.
Collapse
|
115
|
Liao JF, Hsu CC, Chou GT, Hsu JS, Liong MT, Tsai YC. Lactobacillus paracasei PS23 reduced early-life stress abnormalities in maternal separation mouse model. Benef Microbes 2019; 10:425-436. [PMID: 30882243 DOI: 10.3920/bm2018.0077] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maternal separation (MS) has been developed as a model for inducing stress and depression in studies using rodents. The concept of the gut-brain axis suggests that gut health is essential for brain health. Here, we present the effects of administration of a probiotic, Lactobacillus paracasei PS23 (PS23), to MS mice against psychological traits including anxiety and depression. The administration of live and heat-killed PS23 cells showed positive behavioural effects on MS animals, where exploratory tendencies and mobility were increased in behavioural tests, indicating reduced anxiety and depression compared to the negative control mice (P<0.05). Mice administered with both live and heat-killed PS23 cells also showed lower serum corticosterone levels accompanied by higher serum anti-inflammatory interleukin 10 (IL-10) levels, compared to MS separated mice (P<0.05), indicating a stress-elicited response affiliated with increased immunomodulatory properties. Assessment of neurotransmitters in the brain hippocampal region revealed that PS23 affected the concentrations of dopaminergic metabolites differently than the control, suggesting that PS23 may have improved MS-induced stress levels via neurotransmitter pathways, such as dopamine or other mechanisms not addressed in the current study. Our study illustrates the potential of a probiotic in reversing abnormalities induced by early life stress and could be an alternative for brain health along the gut-brain axis.
Collapse
Affiliation(s)
- J F Liao
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No.155, Sec.2, Linong Street, Beitou Dist., Taipei 11221, Taiwan ROC
| | - C C Hsu
- 2 Bened Biomedical Co., Ltd., 2F-2, No.129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei 10448, Taiwan ROC
| | - G T Chou
- 1 Institute of Biochemistry and Molecular Biology, National Yang-Ming University, No.155, Sec.2, Linong Street, Beitou Dist., Taipei 11221, Taiwan ROC
| | - J S Hsu
- 2 Bened Biomedical Co., Ltd., 2F-2, No.129, Sec. 2, Zhongshan N. Rd., Zhongshan Dist., Taipei 10448, Taiwan ROC
| | - M T Liong
- 3 School of Industrial Technology, Universiti Sains Malaysia, 11800 Penang, Malaysia
| | - Y C Tsai
- 4 Microbiome Research Center, National Yang-Ming University, No.155, Sec.2, Linong Street, Beitou Dist., Taipei 11221, Taiwan ROC
| |
Collapse
|
116
|
Pharmacological rescue of nociceptive hypersensitivity and oxytocin analgesia impairment in a rat model of neonatal maternal separation. Pain 2019; 159:2630-2640. [PMID: 30169420 DOI: 10.1097/j.pain.0000000000001375] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Oxytocin (OT), known for its neurohormonal effects around birth, has recently been suggested for being a critical determinant in neurodevelopmental disorders. This hypothalamic neuropeptide exerts a potent analgesic effect through an action on the nociceptive system. This endogenous control of pain has an important adaptive value but might be altered by early life stress, possibly contributing to its long-term consequences on pain responses and associated comorbidities. We tested this hypothesis using a rat model of neonatal maternal separation (NMS) known to induce long-term consequences on several brain functions including chronic stress, anxiety, altered social behavior, and visceral hypersensitivity. We found that adult rats with a history of NMS were hypersensitive to noxious mechanical/thermal hot stimuli and to inflammatory pain. We failed to observe OT receptor-mediated stress-induced analgesia and OT antihyperalgesia after carrageenan inflammation. These alterations were partially rescued if NMS pups were treated by intraperitoneal daily injection during NMS with OT or its downstream second messenger allopregnanolone. The involvement of epigenetic changes in these alterations was confirmed since neonatal treatment with the histone deacetylase inhibitor SAHA, not only normalized nociceptive sensitivities but also restored OT receptor-mediated stress-induced analgesia and the endogenous antihyperalgesia in inflamed NMS rats. There is growing evidence in the literature that early life stress might impair the nociceptive system ontogeny and function. This study suggests that these alterations might be restored while stimulating OT receptor signaling or histone deacetylase inhibitors, using molecules that are currently available or part of clinical trials for other pathologies.
Collapse
|
117
|
Huang TT, Lai JB, Du YL, Xu Y, Ruan LM, Hu SH. Current Understanding of Gut Microbiota in Mood Disorders: An Update of Human Studies. Front Genet 2019; 10:98. [PMID: 30838027 PMCID: PMC6389720 DOI: 10.3389/fgene.2019.00098] [Citation(s) in RCA: 151] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 01/29/2019] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota plays an important role in the bidirectional communication between the gut and the central nervous system. Mounting evidence suggests that gut microbiota can influence the brain function via neuroimmune and neuroendocrine pathways as well as the nervous system. Advances in gene sequencing techniques further facilitate investigating the underlying relationship between gut microbiota and psychiatric disorders. In recent years, researchers have preliminarily explored the gut microbiota in patients with mood disorders. The current review aims to summarize the published human studies of gut microbiota in mood disorders. The findings showed that microbial diversity and taxonomic compositions were significantly changed compared with healthy individuals. Most of these findings revealed that short-chain fatty acids-producing bacterial genera were decreased, while pro-inflammatory genera and those involved in lipid metabolism were increased in patients with depressive episodes. Interestingly, the abundance of Actinobacteria, Enterobacteriaceae was increased and Faecalibacterium was decreased consistently in patients with either bipolar disorder or major depressive disorder. Some studies further indicated that specific bacteria were associated with clinical characteristics, inflammatory profiles, metabolic markers, and pharmacological treatment. These studies present preliminary evidence of the important role of gut microbiota in mood disorders, through the brain-gut-microbiota axis, which emerges as a promising target for disease diagnosis and therapeutic interventions in the future.
Collapse
Affiliation(s)
- Ting-Ting Huang
- Department of Psychiatry, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jian-Bo Lai
- Department of Psychiatry, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China.,Brain Research Institute of Zhejiang University, Hangzhou, China
| | - Yan-Li Du
- Department of Psychiatry, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yi Xu
- Department of Psychiatry, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China.,Brain Research Institute of Zhejiang University, Hangzhou, China
| | - Lie-Min Ruan
- Department of Mental Health, Ningbo First Hospital, Ningbo, China
| | - Shao-Hua Hu
- Department of Psychiatry, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,The Key Laboratory of Mental Disorder's Management of Zhejiang Province, Hangzhou, China.,Brain Research Institute of Zhejiang University, Hangzhou, China
| |
Collapse
|
118
|
Niccolai E, Boem F, Russo E, Amedei A. The Gut⁻Brain Axis in the Neuropsychological Disease Model of Obesity: A Classical Movie Revised by the Emerging Director "Microbiome". Nutrients 2019; 11:E156. [PMID: 30642052 PMCID: PMC6356219 DOI: 10.3390/nu11010156] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/21/2018] [Accepted: 01/08/2019] [Indexed: 02/06/2023] Open
Abstract
The worldwide epidemic of obesity has become an important public health issue, with serious psychological and social consequences. Obesity is a multifactorial disorder in which various elements (genetic, host, and environment), play a definite role, even if none of them satisfactorily explains its etiology. A number of neurological comorbidities, such as anxiety and depression, charges the global obesity burden, and evidence suggests the hypothesis that the brain could be the seat of the initial malfunction leading to obesity. The gut microbiome plays an important role in energy homeostasis regulating energy harvesting, fat deposition, as well as feeding behavior and appetite. Dietary patterns, like the Western diet, are known to be a major cause of the obesity epidemic, probably promoting a dysbiotic drift in the gut microbiota. Moreover, the existence of a "gut⁻brain axis" suggests a role for microbiome on hosts' behavior according to different modalities, including interaction through the nervous system, and mutual crosstalk with the immune and the endocrine systems. In the perspective of obesity as a real neuropsychological disease and in light of the discussed considerations, this review focuses on the microbiome role as an emerging director in the development of obesity.
Collapse
Affiliation(s)
- Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Federico Boem
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Largo Brambilla 3, 50134 Florence, Italy.
- Department of Biomedicine, Azienda Ospedaliera Universitaria Careggi (AOUC), Largo Brambilla 3, 50134 Florence, Italy.
| |
Collapse
|
119
|
Bastiaanssen TFS, Cowan CSM, Claesson MJ, Dinan TG, Cryan JF. Making Sense of … the Microbiome in Psychiatry. Int J Neuropsychopharmacol 2019; 22:37-52. [PMID: 30099552 PMCID: PMC6313131 DOI: 10.1093/ijnp/pyy067] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 07/10/2018] [Accepted: 08/02/2018] [Indexed: 12/13/2022] Open
Abstract
Microorganisms can be found almost anywhere, including in and on the human body. The collection of microorganisms associated with a certain location is called a microbiota, with its collective genetic material referred to as the microbiome. The largest population of microorganisms on the human body resides in the gastrointestinal tract; thus, it is not surprising that the most investigated human microbiome is the human gut microbiome. On average, the gut hosts microbes from more than 60 genera and contains more cells than the human body. The human gut microbiome has been shown to influence many aspects of host health, including more recently the brain.Several modes of interaction between the gut and the brain have been discovered, including via the synthesis of metabolites and neurotransmitters, activation of the vagus nerve, and activation of the immune system. A growing body of work is implicating the microbiome in a variety of psychological processes and neuropsychiatric disorders. These include mood and anxiety disorders, neurodevelopmental disorders such as autism spectrum disorder and schizophrenia, and even neurodegenerative disorders such as Alzheimer's and Parkinson's diseases. Moreover, it is probable that most psychotropic medications have an impact on the microbiome.Here, an overview will be provided for the bidirectional role of the microbiome in brain health, age-associated cognitive decline, and neurological and psychiatric disorders. Furthermore, a primer on the common microbiological and bioinformatics techniques used to interrogate the microbiome will be provided. This review is meant to equip the reader with a primer to this exciting research area that is permeating all areas of biological psychiatry research.
Collapse
Affiliation(s)
- Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | | | - Marcus J Claesson
- APC Microbiome Ireland, University College Cork, Ireland
- School of Microbiology, University College Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Ireland
- Department of Psychiatry, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| |
Collapse
|
120
|
Tian P, Zou R, Song L, Zhang X, Jiang B, Wang G, Lee YK, Zhao J, Zhang H, Chen W. Ingestion of Bifidobacterium longum subspecies infantis strain CCFM687 regulated emotional behavior and the central BDNF pathway in chronic stress-induced depressive mice through reshaping the gut microbiota. Food Funct 2019; 10:7588-7598. [DOI: 10.1039/c9fo01630a] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Probiotics which enhance the biosynthesis of 5-hydroxytryptamine in enterochromaffin cells could alleviate depression symptoms through regulating the CREB-BDNF pathway in the brain.
Collapse
|
121
|
Qian L, Lu L, Huang L, Wen Q, Xie J, Jin W, Li H, Jiang L. The effect of neonatal maternal separation on short-chain fatty acids and airway inflammation in adult asthma mice. Allergol Immunopathol (Madr) 2019; 47:2-11. [PMID: 30458973 DOI: 10.1016/j.aller.2018.05.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/23/2018] [Accepted: 05/10/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND To investigate neonatal maternal separation (NMS) effects on airway inflammation of asthma and potential mechanism using a mouse model. METHODS 80 Balb/c neonatal male mice were randomly assigned to NMS and non-NMS groups. Feces were collected on PND21, 28, 35 and 42 to analyze microbiota and short-chain fatty acids (SCFAs). Non-NMS group were then divided into control (group A) and asthma groups (group B), while NMS group was assigned to NMS+asthma (group C) and NMS+SCFAs+asthma groups (group D). Inflammatory cells and eosinophils (EOS) in bronchoalveolar lavage fluid (BALF) were assessed. Pathological changes and cytokines in lung tissue were observed. Protein expression of Occludin and E-cadherin in airway epithelial was examined. RESULTS The number of S', diversity index H' and dominance index D', as well as content butyric acid in NMS group C were significantly lower than non-NMS group B (p<0.05). Mice in group C had a higher level of inflammatory cells and EOS compared with group A, B and D. EOS moderate infiltration was found in mice of group B, C and D. Mice in group C had significantly higher levels of cytokines and showed slightly increased bronchial epithelium goblet cells and a small amount of visceral secretions. Occludin and E-cadherin expression in lung in B, C and D groups was depressed, and protein level in group C was significantly lower than group B and D. CONCLUSIONS NMS is associated with exacerbated inflammation of adult asthma by changing intestinal microflora resulting in butanoic acid decline and airway epithelial barrier damage.
Collapse
|
122
|
Tillmann S, Abildgaard A, Winther G, Wegener G. Altered fecal microbiota composition in the Flinders sensitive line rat model of depression. Psychopharmacology (Berl) 2019; 236:1445-1457. [PMID: 30470860 PMCID: PMC6599185 DOI: 10.1007/s00213-018-5094-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/25/2018] [Indexed: 12/12/2022]
Abstract
RATIONALE The gut microbiota is increasingly recognized as a potential mediator of psychiatric diseases. Depressed patients have been shown to have a different microbiota composition compared with healthy controls, and several lines of research now aim to restore this dysbiosis. To develop novel treatments, preclinical models may provide novel mechanistic insights. OBJECTIVE AND METHODS We characterized the gut microbiota of male adult Flinders sensitive line (FSL) rats, an animal model of depression, and their controls, Flinders resistant line (FRL) rats using 16S rRNA amplicon sequencing. Moreover, we performed fecal microbiota transplantation (using saline or pooled FRL/FSL feces) to study if the potential strain-specific differences could be transferred from one strain to the other, and if these differences were reflected in their depressive-like behavior in the forced swim test. RESULTS FSL rats tended to have lower bacterial richness and altered relative abundances of several bacterial phyla, families, and species, including higher Proteobacteria and lower Elusimicrobia and Saccharibacteria. There was a clear separation between FRL and FSL rat strains, but no effect of treatment, i.e., the bacterial composition of FSL rats receiving FRL feces was still more similar to FSL and not FRL rats. Similarly, the transplantation did not reverse behavioral differences in the forced swim test, although FSL feces significantly increased immobility compared with saline. CONCLUSIONS Our study showed that the gut microbiota composition of the depressive-like rats markedly differed from their controls, which may be of value for future microbiota-targeted work in this and similar animal models.
Collapse
Affiliation(s)
- Sandra Tillmann
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Anders Abildgaard
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Clinical Biochemistry, Aarhus University Hospital, Aarhus, Denmark
| | - Gudrun Winther
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
123
|
Al-Orf N, El-Ansary A, Bjørklund G, Moubayed N, Bhat RS, Bacha AB. Therapeutic effects of probiotics on neurotoxicity induced by clindamycin and propionic acid in juvenile hamsters. Metab Brain Dis 2018; 33:1811-1820. [PMID: 30019266 DOI: 10.1007/s11011-018-0284-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 06/29/2018] [Indexed: 10/28/2022]
Abstract
The present study investigated the therapeutic effects of probiotics on brain intoxication induced by clindamycin and propionic acid (PPA) in hamsters. Fifty golden Syrian hamsters were randomly divided into five experimental groups of ten animals each: (A) control group receiving phosphate buffered saline; (B) oral buffered PPA-treated group being administered with a neurotoxic dose of 250 mg/kg PPA during three days; (C) oral clindamycin-treated group receiving a single dose of 30 mg clindamycin/kg; and (D, E) the two therapeutic groups being administered the same doses of clindamycin and PPA followed by probiotics for three weeks at a daily dose of 0.2 g/kg. Biochemical parameters of energy metabolism and oxidative stress were examined in brain homogenates from all hamsters. The development of pathogenic bacteria was monitored on stool samples from all hamsters. Descriptive changes in fecal microbiota and overgrowth of Clostridium species in clindamycin and PPA treated hamsters were recorded. Interestingly, probiotics were shown effective to restore normal gut microbiota. Clindamycin and PPA treatments caused an elevation in lipid peroxidation and catalase activity, as oxidative stress markers, together with a reduction in GST activity and GSH level. Energy metabolism impairment was ascertained via the activation of creatine kinase and a decrease of lactate dehydrogenase. These findings suggest that bacteria overgrowth caused by PPA and clindamycin was efficient to illustrate signs of neuronal toxicity. The present study indicates that probiotic treatment can improve poor detoxification, oxidative stress, and altered gut microbiota as mechanisms implicated in the etiology of many neurological disorders.
Collapse
Affiliation(s)
- Nora Al-Orf
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
- The Materials Science Research Institute, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Afaf El-Ansary
- Central Laboratory, King Saud University, Riyadh, Saudi Arabia
| | - Geir Bjørklund
- Council for Nutritional and Environmental Medicine, Toften 24, 8610, Mo i Rana, Norway.
| | - Nadine Moubayed
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Ramesa Shafi Bhat
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abir Ben Bacha
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
- Laboratory of Plant Biotechnology Applied to Crop Improvement, Faculty of Science of Sfax, University of Sfax, Sfax, Tunisia
| |
Collapse
|
124
|
A randomized clinical trial examining the impact of LGG probiotic supplementation on psychological status in middle-aged and older adults. Contemp Clin Trials Commun 2018; 12:192-197. [PMID: 30511028 PMCID: PMC6251018 DOI: 10.1016/j.conctc.2018.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 10/01/2018] [Accepted: 11/13/2018] [Indexed: 12/26/2022] Open
Abstract
Cognitive decline is common in older adults and more than 5 million Americans suffer from Alzheimer's disease (AD). A number of physiological processes including systemic inflammation, excess adiposity, and impaired glucoregulation arise from a combination of genetic and behavioral factors and increase risk for developing AD or other forms of dementia. Recent research suggests that the gut microbiome may moderate these pathological processes and possibly influence cognitive outcomes. This paper reviews the methodology for a double-blind, randomized clinical trial examining the influence of Lactobacillus GG (LGG) probiotic supplementation on mood and cognitive functioning in middle-aged and older adults. Our two primary hypotheses include: 1) Participants randomized to the probiotic group will show greater improvements in psychological status compared to participants in the placebo group; 2) Participants randomized to the probiotic group will show greater improvements in executive functioning and processing speed, as evidenced through performance on neuropsychological testing, than participants in the placebo group. We anticipate these results will inform future efforts on the feasibility of LGG probiotic supplementation as an intervention for psychological status and cognitive functioning and further elucidate the link between the gut microbiome and cognitive health.
Collapse
|
125
|
Lucking EF, O'Connor KM, Strain CR, Fouhy F, Bastiaanssen TFS, Burns DP, Golubeva AV, Stanton C, Clarke G, Cryan JF, O'Halloran KD. Chronic intermittent hypoxia disrupts cardiorespiratory homeostasis and gut microbiota composition in adult male guinea-pigs. EBioMedicine 2018; 38:191-205. [PMID: 30446434 PMCID: PMC6306383 DOI: 10.1016/j.ebiom.2018.11.010] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/26/2018] [Accepted: 11/05/2018] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Carotid body (peripheral oxygen sensor) sensitisation is pivotal in the development of chronic intermittent hypoxia (CIH)-induced hypertension. We sought to determine if exposure to CIH, modelling human sleep apnoea, adversely affects cardiorespiratory control in guinea-pigs, a species with hypoxia-insensitive carotid bodies. We reasoned that CIH-induced disruption of gut microbiota would evoke cardiorespiratory morbidity. METHODS Adult male guinea-pigs were exposed to CIH (6.5% O2 at nadir, 6 cycles.hour-1) for 8 h.day-1 for 12 consecutive days. FINDINGS CIH-exposed animals established reduced faecal microbiota species richness, with increased relative abundance of Bacteroidetes and reduced relative abundance of Firmicutes bacteria. Urinary corticosterone and noradrenaline levels were unchanged in CIH-exposed animals, but brainstem noradrenaline concentrations were lower compared with sham. Baseline ventilation was equivalent in CIH-exposed and sham animals; however, respiratory timing variability, sigh frequency and ventilation during hypoxic breathing were all lower in CIH-exposed animals. Baseline arterial blood pressure was unaffected by exposure to CIH, but β-adrenoceptor-dependent tachycardia and blunted bradycardia during phenylephrine-induced pressor responses was evident compared with sham controls. INTERPRETATION Increased carotid body chemo-afferent signalling appears obligatory for the development of CIH-induced hypertension and elevated chemoreflex control of breathing commonly reported in mammals, with hypoxia-sensitive carotid bodies. However, we reveal that exposure to modest CIH alters gut microbiota richness and composition, brainstem neurochemistry, and autonomic control of heart rate, independent of carotid body sensitisation, suggesting modulation of breathing and autonomic homeostasis via the microbiota-gut-brainstem axis. The findings have relevance to human sleep-disordered breathing. FUNDING The Department of Physiology, and APC Microbiome Ireland, UCC.
Collapse
Affiliation(s)
- Eric F Lucking
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Karen M O'Connor
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Conall R Strain
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Fiona Fouhy
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Thomaz F S Bastiaanssen
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - David P Burns
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland
| | - Anna V Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - John F Cryan
- Department of Anatomy & Neuroscience, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ken D O'Halloran
- Department of Physiology, School of Medicine, College of Medicine & Health, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland.
| |
Collapse
|
126
|
Li B, Yu FZ, Minich A, Hock A, Lee C, Pierro A. Neonatal intestinal injury induced by maternal separation: pathogenesis and pharmacological targets 1. Can J Physiol Pharmacol 2018; 97:193-196. [PMID: 30383976 DOI: 10.1139/cjpp-2018-0370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Maternal separation (MS) is a well-studied phenomenon thought to play a role in the pathogenesis of many diseases ranging from neuropsychiatric to early intestinal disorders such as necrotizing enterocolitis. The existing evidence suggests that MS initiates a variety of processes that in turn lead to early intestinal injury. Although there are many theories as to how MS alters normal physiological processes, the exact mechanism of action remains to be elucidated. This review aims to describe some of the pathological processes affecting the intestine that are caused by MS, including (i) brain-gut axis, (ii) intestinal epithelial barrier function, (iii) microbiome, (iv) oxidative stress and endoplasmic reticulum stress, and (v) gut inflammation.
Collapse
Affiliation(s)
- Bo Li
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Fang Zhou Yu
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada.,b School of Medicine, University of St Andrews, St Andrews, Fife, United Kingdom
| | - Adam Minich
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Alison Hock
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Carol Lee
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Agostino Pierro
- a Division of General and Thoracic Surgery, Translation Medicine Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
127
|
Olén O, Stephansson O, Backman AS, Törnblom H, Simrén M, Altman M. Pre- and perinatal stress and irritable bowel syndrome in young adults - A nationwide register-based cohort study. Neurogastroenterol Motil 2018; 30:e13436. [PMID: 30084125 DOI: 10.1111/nmo.13436] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/02/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND The etiology of irritable bowel syndrome (IBS) is poorly understood. Animal and human data suggest that early life stress may induce long-term changes in the nociceptive circuitry, but conclusive studies are lacking. METHODS We identified all Swedish children born between 1973 and 1992 in the Swedish Medical Birth Register. We had access to all diagnostic codes for specialized (nonprimary care) outpatient visits 2001-2009 (the National Patient Register) and identified individuals who were diagnosed with IBS (ICD-10 code: K58) after 18 years of age. We compared incidence of IBS in individuals with and without pre- and perinatal stress using multivariable logistic regression. KEY RESULTS 2 056 430 children were included in the study. After turning 18 years, 14 382 of them were diagnosed with IBS in specialized outpatient care. Neither high, nor low birth weight was a risk factor for IBS in young adults. Preterm birth was associated with lower occurrence of IBS (adjusted OR 0.82 [0.75-0.90]) and vaginal instrumental delivery and Cesarean delivery were associated with slightly increased odds of IBS (adjusted OR 1.14 [1.06-1.24] and 1.09 [1.03-1.16] respectively). Neonatal distress and respiratory distress were not associated with future IBS. Female gender was by far the strongest risk factor for IBS in young adults (adjusted OR 3.48 [3.34-3.63]). CONCLUSIONS & INFERENCES In this large population-based study, we found that mode of delivery was associated with an increased risk for IBS in young adulthood, while other proxies for pre- and perinatal stress were not. Female gender remains the most important risk factor for IBS.
Collapse
Affiliation(s)
- Ola Olén
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden.,Sachs' Children's Hospital, Stockholm South General Hospital, Stockholm, Sweden
| | - Olof Stephansson
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Ann-Sofie Backman
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Hans Törnblom
- Department of Internal Medicine & Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Magnus Simrén
- Department of Internal Medicine & Clinical Nutrition, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Center for Functional Gastrointestinal and Motility Disorders, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Maria Altman
- Clinical Epidemiology Unit, Department of Medicine Solna, Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
128
|
Ihekweazu FD, Versalovic J. Development of the Pediatric Gut Microbiome: Impact on Health and Disease. Am J Med Sci 2018; 356:413-423. [PMID: 30384950 PMCID: PMC6268214 DOI: 10.1016/j.amjms.2018.08.005] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023]
Abstract
The intestinal microbiota are important in human growth and development. Microbial composition may yield insights into the temporal development of microbial communities and vulnerabilities to disorders of microbial ecology such as recurrent Clostridium difficile infection. Discoveries of key microbiome features of carbohydrate and amino acid metabolism are lending new insights into possible therapies or preventative strategies for inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS). In this review, we summarize the current understanding of the development of the pediatric gastrointestinal microbiome, the influence of the microbiome on the developing brain through the gut-brain axis, and the impact of dysbiosis on disease development. Dysbiosis is explored in the context of pediatric allergy and asthma, recurrent C. difficile infection, IBD, IBS, and metabolic disorders. The central premise is that the human intestinal microbiome plays a vital role in health and disease, beginning in the prenatal period and extending throughout childhood.
Collapse
Affiliation(s)
- Faith D. Ihekweazu
- Pediatric Gastroenterology, Hepatology and Nutrition, Baylor College of Medicine, Texas Children’s Hospital, 1102 Bates St., Houston, TX, 77030, USA.
| | - James Versalovic
- Pediatric Pathology and Immunology, Baylor College of Medicine, Texas Children’s Hospital 1102 Bates St., Houston, TX, 77030, USA.
| |
Collapse
|
129
|
|
130
|
Abstract
BACKGROUND Visceral pain is a complex and heterogeneous disorder, which can range from the mild discomfort of indigestion to the agonizing pain of renal colic. Regulation of visceral pain involves the spinal cord as well as higher order brain structures. Recent findings have linked the microbiota to gastrointestinal disorders characterized by abdominal pain suggesting the ability of microbes to modulate visceral hypersensitivity and nociception to pain. MAIN BODY In this review we describe the neuroanatomical basis of visceral pain signaling and the existing evidence of its manipulation exerted by the gut microbiota. We included an updated overview of the potential therapeutic effects of dietary intervention, specifically probiotics and prebiotics, in alleviating hypersensitivity to visceral pain stimuli. CONCLUSIONS The gut microbiota dramatically impacts normal visceral pain sensation and affects the mechanisms mediating visceral nociception. Furthermore, manipulation of the gut microbiota using prebiotics and probiotics plays a potential role in the regulation of visceral pain disorders.
Collapse
Affiliation(s)
- Matteo M Pusceddu
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, One Shield Avenue, Davis, CA, USA.
| | - Melanie G Gareau
- Department of Anatomy, Physiology and Cell Biology, School of Veterinary Medicine, University of California Davis, One Shield Avenue, Davis, CA, USA.
| |
Collapse
|
131
|
Ruiz R, Roque A, Pineda E, Licona-Limón P, José Valdéz-Alarcón J, Lajud N. Early life stress accelerates age-induced effects on neurogenesis, depression, and metabolic risk. Psychoneuroendocrinology 2018; 96:203-211. [PMID: 30048914 DOI: 10.1016/j.psyneuen.2018.07.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/15/2018] [Accepted: 07/07/2018] [Indexed: 01/06/2023]
Abstract
Early life stress (ELS) affects hippocampal neurogenesis, increases depressive-like behavior, and causes mild metabolic imbalance in early adulthood (2 months). However, whether these effects worsen in mid life remains unclear. To test whether age-dependent effects of ELS on hippocampal neurogenesis are related to deficient hypothalamic-pituitary-adrenal (HPA) axis feedback that causes increased comorbidity of depression and metabolic risk, we evaluated the effects of periodic maternal separation (MS180) in young (4-months-old) and middle-aged (10-months-old) adult rats. MS180 caused more severe depressive-like behavior in middle-aged adults than in young animals. There were no behavioral phenotypic differences between young MS180 and control middle-aged groups. MS180 similarly affected glucose tolerance, increased fasting corticosterone, insulin, and the quantitative insulin sensitivity check index (QUICKI) at both ages. However, middle-aged adult MS180 rats showed more severe age-induced obesity (>40% BW) than controls (>22% BW). MS180 differentially affected dorsal and ventral neurogenesis. In young adults, MS180 animals only showed a decrease in dorsal hippocampal neurogenesis as compared to their age-matched counterparts. In contrast, at 10 months of age, MS180 caused a similar decrease in both dorsal and ventral hippocampal neurogenesis as compared to age-matched controls, and a more severe decrease as compared to young animals. Taken together, our data indicate that MS180 animals show an early onset of age-induced alterations on depression and metabolic risk, and these effects relate to alterations in hippocampal neurogenesis.
Collapse
Affiliation(s)
- Roberto Ruiz
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán-Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico; Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Angélica Roque
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán-Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Edel Pineda
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán-Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico
| | - Paula Licona-Limón
- Centro Multidisciplinario de Estudios en Biotecnología, Facultad de Medicina Veterinaria y Zootecnia - Benemérita y Centenaria Universidad Michoacana de San Nicolás de Hidalgo, Mexico
| | - Juan José Valdéz-Alarcón
- Departamento de Biología Celular y del Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Naima Lajud
- División de Neurociencias, Centro de Investigación Biomédica de Michoacán-Instituto Mexicano del Seguro Social, Morelia, Michoacán, Mexico.
| |
Collapse
|
132
|
Gut-brain actions underlying comorbid anxiety and depression associated with inflammatory bowel disease. Acta Neuropsychiatr 2018; 30:275-296. [PMID: 28270247 DOI: 10.1017/neu.2017.3] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
UNLABELLED IntroductionInflammatory bowel disease (IBD) is a chronic relapsing and remitting disorder characterised by inflammation of the gastrointestinal tract. There is a growing consensus that IBD is associated with anxiety- and depression-related symptoms. Psychological symptoms appear to be more prevalent during active disease states with no difference in prevalence between Crohn's disease and ulcerative colitis. Behavioural disturbances including anxiety- and depression-like symptoms have also been observed in animal models of IBD. RESULTS The likely mechanisms underlying the association are discussed with particular reference to communication between the gut and brain. The close bidirectional relationship known as the gut-brain axis includes neural, hormonal and immune communication links. Evidence is provided for a number of interacting factors including activation of the inflammatory response system in the brain, the hypothalamic-pituitary-adrenal axis, and brain areas implicated in altered behaviours, changes in blood brain barrier integrity, and an emerging role for gut microbiota and response to probiotics in IBD.DiscussionThe impact of psychological stress in models of IBD remains somewhat conflicted, however, it is weighted in favour of stress or early stressful life events as risk factors in the development of IBD, stress-induced exacerbation of inflammation and relapse. CONCLUSION It is recommended that patients with IBD be screened for psychological disturbance and treated accordingly as intervention can improve quality of life and may reduce relapse rates.
Collapse
|
133
|
Early life stress in mice is a suitable model for Irritable Bowel Syndrome but does not predispose to colitis nor increase susceptibility to enteric infections. Brain Behav Immun 2018; 73:403-415. [PMID: 29860025 DOI: 10.1016/j.bbi.2018.05.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 05/17/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023] Open
Abstract
Neonatal period is characterized by an immature intestinal barrier. Scattered evidence suggests that early life stressful events induce long lasting alterations of intestinal homeostasis mimicking Irritable Bowel Syndrome (IBS). Those observations highlighting defect of intestinal barrier by early life stress questioned its potential role as a risk factor for gastrointestinal disorders such as colitis and infections. In this study, we aimed to analyze if maternal separation (MS) in mice mimicks IBS main features. We next addressed whether MS could trigger or exacerbate colitis in genetically predisposed mice and/or enhance susceptibility to gastrointestinal infections in wild type mice. MS induced main features of IBS in adult wild type male mice i.e. intestinal hyperpermeability, visceral hypersensitivity, microbiota dysbiosis, bile acid malabsorption and low grade inflammation in intestine associated with a defect of Paneth cells and the ILC3 population. This breach in mucosal barrier functions in adults was associated with a systemic IgG response against commensal E. coli and increased IFNγ secretion by splenocytes. However, in IL10-/- mice, MS did not trigger nor worsen colitis. Furthermore, wild type mice submitted to MS did not show increase susceptibility to gastrointestinal infections (S. Typhimurium, L. monocytogenes or T. gondii) compared to controls. Altogether, our results identify MS in mice as a good experimental model for IBS mimicking all the main features. In addition, early life stress, even though it has long lasting consequences on intestinal homeostasis, does not constitute a facilitating factor to colitis in predisposed individuals nor to gastrointestinal infections in wild type mice.
Collapse
|
134
|
Cussotto S, Sandhu KV, Dinan TG, Cryan JF. The Neuroendocrinology of the Microbiota-Gut-Brain Axis: A Behavioural Perspective. Front Neuroendocrinol 2018; 51:80-101. [PMID: 29753796 DOI: 10.1016/j.yfrne.2018.04.002] [Citation(s) in RCA: 195] [Impact Index Per Article: 27.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/17/2022]
Abstract
The human gut harbours trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. These intestinal microbes are also key components of the gut-brain axis, the bidirectional communication pathway between the gut and the central nervous system (CNS). In addition, the CNS is closely interconnected with the endocrine system to regulate many physiological processes. An expanding body of evidence is supporting the notion that gut microbiota modifications and/or manipulations may also play a crucial role in the manifestation of specific behavioural responses regulated by neuroendocrine pathways. In this review, we will focus on how the intestinal microorganisms interact with elements of the host neuroendocrine system to modify behaviours relevant to stress, eating behaviour, sexual behaviour, social behaviour, cognition and addiction.
Collapse
Affiliation(s)
- Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Kiran V Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland.
| |
Collapse
|
135
|
Kim YK, Shin C. The Microbiota-Gut-Brain Axis in Neuropsychiatric Disorders: Pathophysiological Mechanisms and Novel Treatments. Curr Neuropharmacol 2018; 16:559-573. [PMID: 28925886 PMCID: PMC5997867 DOI: 10.2174/1570159x15666170915141036] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/10/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023] Open
Abstract
Background The human gut microbiome comprise a huge number of microorganisms with co-evolutionary associations with humans. It has been repeatedly revealed that bidirectional communication exists between the brain and the gut and involves neural, hormonal, and immunological pathways. Evidences from neuroscience researches over the past few years suggest that microbiota is essential for the development and maturation of brain systems that are associated to stress responses. Method This review provides that the summarization of the communication among microbiota, gut and brain and the results of preclinical and clinical studies on gut microbiota used in treatments for neuropsychiatric disorders. Result Recent studies have reported that diverse forms of neuropsychiatric disorders (such as autism, depression, anxiety, and schizophrenia) are associated with or modulated by variations in the microbiome, by microbial substrates, and by exogenous prebiotics, antibiotics, and probiotics. Conclusion The microbiota–gut–brain axis might provide novel targets for prevention and treatment of neuropsychiatric disorders. However, further studies are required to substantiate the clinical use of probiotics, prebiotics and FMT.
Collapse
Affiliation(s)
- Yong-Ku Kim
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| | - Cheolmin Shin
- Department of Psychiatry, College of Medicine, Korea University, Seoul, Korea
| |
Collapse
|
136
|
Liang S, Wu X, Jin F. Gut-Brain Psychology: Rethinking Psychology From the Microbiota-Gut-Brain Axis. Front Integr Neurosci 2018; 12:33. [PMID: 30271330 PMCID: PMC6142822 DOI: 10.3389/fnint.2018.00033] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022] Open
Abstract
Mental disorders and neurological diseases are becoming a rapidly increasing medical burden. Although extensive studies have been conducted, the progress in developing effective therapies for these diseases has still been slow. The current dilemma reminds us that the human being is a superorganism. Only when we take the human self and its partner microbiota into consideration at the same time, can we better understand these diseases. Over the last few centuries, the partner microbiota has experienced tremendous change, much more than human genes, because of the modern transformations in diet, lifestyle, medical care, and so on, parallel to the modern epidemiological transition. Existing research indicates that gut microbiota plays an important role in this transition. According to gut-brain psychology, the gut microbiota is a crucial part of the gut-brain network, and it communicates with the brain via the microbiota-gut-brain axis. The gut microbiota almost develops synchronously with the gut-brain, brain, and mind. The gut microbiota influences various normal mental processes and mental phenomena, and is involved in the pathophysiology of numerous mental and neurological diseases. Targeting the microbiota in therapy for these diseases is a promising approach that is supported by three theories: the gut microbiota hypothesis, the "old friend" hypothesis, and the leaky gut theory. The effects of gut microbiota on the brain and behavior are fulfilled by the microbiota-gut-brain axis, which is mainly composed of the nervous pathway, endocrine pathway, and immune pathway. Undoubtedly, gut-brain psychology will bring great enhancement to psychology, neuroscience, and psychiatry. Various microbiota-improving methods including fecal microbiota transplantation, probiotics, prebiotics, a healthy diet, and healthy lifestyle have shown the capability to promote the function of the gut-brain, microbiota-gut-brain axis, and brain. It will be possible to harness the gut microbiota to improve brain and mental health and prevent and treat related diseases in the future.
Collapse
Affiliation(s)
- Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoli Wu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
137
|
Liu YW, Liong MT, Tsai YC. New perspectives of Lactobacillus plantarum as a probiotic: The gut-heart-brain axis. J Microbiol 2018; 56:601-613. [DOI: 10.1007/s12275-018-8079-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/17/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
|
138
|
Neonatal- maternal separation primes zymogenic cells in the rat gastric mucosa through glucocorticoid receptor activity. Sci Rep 2018; 8:9823. [PMID: 29959361 PMCID: PMC6026145 DOI: 10.1038/s41598-018-28223-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 06/19/2018] [Indexed: 12/21/2022] Open
Abstract
Neonatal- Maternal Separation (NMS) deprives mammals from breastfeeding and maternal care, influencing growth during suckling- weaning transition. In the gastric mucosa, Mist1 (encoded by Bhlha15 gene) and moesin organize the secretory apparatus for pepsinogen C in zymogenic cells. Our current hypothesis was that NMS would change corticosterone activity through receptors (GR), which would modify molecules involved in zymogenic cell differentiation in rats. We found that NMS increased corticosterone levels from 18 days onwards, as GR decreased in the gastric mucosa. However, as nuclear GR was detected, we investigated receptor binding to responsive elements (GRE) and observed an augment in NMS groups. Next, we demonstrated that NMS increased zymogenic population (18 and and 30 days), and targeted Mist1 and moesin. Finally, we searched for evolutionarily conserved sequences that contained GRE in genes involved in pepsinogen C secretion, and found that the genomic regions of Bhlha15 and PgC contained sites highly likely to be responsive to glucocorticoids. We suggest that NMS triggers GR- GRE to enhance the expression and to prime genes that organize cellular architecture in zymogenic population for PgC function. As pepsinogen C- pepsin is essential for digestion, disturbance of parenting through NMS might alter functions of gastric mucosa in a permanent manner.
Collapse
|
139
|
Wang YT, Xu WX. Role of stress in pathophysiology of irritable bowel syndrome. Shijie Huaren Xiaohua Zazhi 2018; 26:1064-1070. [DOI: 10.11569/wcjd.v26.i17.1064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Irritable bowel syndrome (IBS), one of the most common functional gastrointestinal disorders in the world, is characterized by chronic intermittent abdominal discomfort and colon dysmotility with altered bowel habits, significantly impacting patients' quality of life. The pathophysiology of IBS remains incompletely understood although some contributing factors have been identified. Increased visceral sensitivity and intestinal permeability may play an important role in the pathophysiology of IBS. Psychological factors, especially stress, play an important role in the occurrence, development, and regulation of IBS. To facilitate further research of IBS, this review focuses on the relationship between stress and IBS in animal models, as well as the role of stress in increased visceral sensitivity and intestinal permeability in IBS.
Collapse
Affiliation(s)
- Yu-Ting Wang
- Shanghai Jiao Tong University School of Medicine, Shanghai 200020, China
| | - Wen-Xie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, China
| |
Collapse
|
140
|
Liang S, Wu X, Hu X, Wang T, Jin F. Recognizing Depression from the Microbiota⁻Gut⁻Brain Axis. Int J Mol Sci 2018; 19:ijms19061592. [PMID: 29843470 PMCID: PMC6032096 DOI: 10.3390/ijms19061592] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 04/19/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022] Open
Abstract
Major depression is one of the leading causes of disability, morbidity, and mortality worldwide. The brain⁻gut axis functions are disturbed, revealed by a dysfunction of the brain, immune system, endocrine system, and gut. Traditional depression treatments all target the brain, with different drugs and/or psychotherapy. Unfortunately, most of the patients have never received any treatment. Studies indicate that gut microbiota could be a direct cause for the disorder. Abnormal microbiota and the microbiota⁻gut⁻brain dysfunction may cause mental disorders, while correcting these disturbance could alleviate depression. Nowadays, the gut microbiota modulation has become a hot topic in treatment research of mental disorders. Depression is closely related with the health condition of the brain⁻gut axis, and maintaining/restoring the normal condition of gut microbiota helps in the prevention/therapy of mental disorders.
Collapse
Affiliation(s)
- Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Xiaoli Wu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
- Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Xu Hu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Tao Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
141
|
Agustí A, García-Pardo MP, López-Almela I, Campillo I, Maes M, Romaní-Pérez M, Sanz Y. Interplay Between the Gut-Brain Axis, Obesity and Cognitive Function. Front Neurosci 2018; 12:155. [PMID: 29615850 PMCID: PMC5864897 DOI: 10.3389/fnins.2018.00155] [Citation(s) in RCA: 178] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 02/26/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity continues to be one of the major public health problems due to its high prevalence and co-morbidities. Common co-morbidities not only include cardiometabolic disorders but also mood and cognitive disorders. Obese subjects often show deficits in memory, learning and executive functions compared to normal weight subjects. Epidemiological studies also indicate that obesity is associated with a higher risk of developing depression and anxiety, and vice versa. These associations between pathologies that presumably have different etiologies suggest shared pathological mechanisms. Gut microbiota is a mediating factor between the environmental pressures (e.g., diet, lifestyle) and host physiology, and its alteration could partly explain the cross-link between those pathologies. Westernized dietary patterns are known to be a major cause of the obesity epidemic, which also promotes a dysbiotic drift in the gut microbiota; this, in turn, seems to contribute to obesity-related complications. Experimental studies in animal models and, to a lesser extent, in humans suggest that the obesity-associated microbiota may contribute to the endocrine, neurochemical and inflammatory alterations underlying obesity and its comorbidities. These include dysregulation of the HPA-axis with overproduction of glucocorticoids, alterations in levels of neuroactive metabolites (e.g., neurotransmitters, short-chain fatty acids) and activation of a pro-inflammatory milieu that can cause neuro-inflammation. This review updates current knowledge about the role and mode of action of the gut microbiota in the cross-link between energy metabolism, mood and cognitive function.
Collapse
Affiliation(s)
- Ana Agustí
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Maria P García-Pardo
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Inmaculada López-Almela
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Isabel Campillo
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - Marina Romaní-Pérez
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Microbial Ecology and Nutrition Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
142
|
Jang DE, Bae JH, Chang YJ, Lee YH, Nam KT, Kim IY, Seong JK, Lee YC, Yeom SC. Neuronal Nitric Oxide Synthase Is a Novel Biomarker for the Interstitial Cells of Cajal in Stress-Induced Diarrhea-Dominant Irritable Bowel Syndrome. Dig Dis Sci 2018; 63:619-627. [PMID: 29372479 DOI: 10.1007/s10620-018-4933-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 01/16/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Irritable bowel syndrome (IBS) is a functional gastrointestinal disorder involving changes in normal bowel movements. The pathophysiology of IBS is not clearly understood owing to the lack of identifiable pathological abnormalities and reliable biomarkers. AIM The aim of this study was to discover the novel and reliable biomarker for IBS. METHOD In this study, neonatal maternal separation (NMS) stress model was used for the IBS mouse model. Further assessment was conducted with whole gastrointestinal transit test, quantitative RT-PCR, histological examination, and western blot. RESULTS Male pups developed symptoms similar to those of human IBS with diarrhea (IBS-D), such as low-grade inflammation, stool irregularity, and increased bowel motility. NMS stress influenced to the interstitial cells of Cajal (ICC) and induced altered bowel motility, resulting in IBS-D-like symptoms. In addition, we found neuronal nitric oxide synthase (nNOS) to be a novel biomarker for ICC under NMS stress. nNOS expression was only observed in the ICC of the submucosal plexus of IBS-D mice, and the inhibition of nNOS changed the phenotype from IBS-D to IBS with constipation. CONCLUSION Our study demonstrates that early-life stress can influence to ICC and modulate bowel activity and that nNOS might be used as a biomarker for ICC stimulation in IBS.
Collapse
Affiliation(s)
- Da Eun Jang
- Graduate School of International Agricultural Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea
| | - Ji Hyun Bae
- Designed Animal and Transplantation Research Institute, Institute of Greenbio Research and Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea
| | - Yoo Jin Chang
- Graduate School of International Agricultural Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea
| | - Yoon Hoo Lee
- Graduate School of International Agricultural Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea
| | - Ki Taek Nam
- Severance Biomedical Science Institute, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul, 25354, Republic of Korea
| | - Il Yong Kim
- Department of Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Je Kyung Seong
- Department of Veterinary Science, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, Republic of Korea
| | - Yong Chan Lee
- Yonsei University, 50 Yonsei-ro Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Su Cheong Yeom
- Graduate School of International Agricultural Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea.
- Designed Animal and Transplantation Research Institute, Institute of Greenbio Research and Technology, Seoul National University, 1447 Pyeongchang-Ro, Daewha, Pyeongchang, Gangwon, 25354, Republic of Korea.
| |
Collapse
|
143
|
Pierce AN, Eller-Smith OC, Christianson JA. Voluntary wheel running attenuates urinary bladder hypersensitivity and dysfunction following neonatal maternal separation in female mice. Neurourol Urodyn 2018; 37:1623-1632. [PMID: 29464752 DOI: 10.1002/nau.23530] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 01/28/2018] [Indexed: 01/08/2023]
Abstract
AIMS Patients with interstitial cystitis/painful bladder syndrome (IC/PBS) commonly suffer from widespread pain and mood disorder, which has been attributed to improper functioning of the hypothalamic-pituitary-adrenal (HPA) axis. Voluntary exercise has been shown to improve HPA axis function, therefore we are determining whether voluntary wheel running can attenuate urological pain and dysfunction following neonatal maternal separation (NMS) in female mice. METHODS Mice underwent NMS for 3 h/day from postnatal Day 1-21, were caged with free access to running wheels at 4 weeks of age, and assessed 4 weeks later for bladder sensitivity, micturition, reward behavior, mast cell degranulation, and HPA axis-related in vitro analysis. RESULTS Increased bladder sensitivity, void frequency, and mast cell degranulation was observed in adult sedentary (-Sed) NMS mice, compared to naïve-Sed controls. Sucrose preference was increased in NMS-Sed mice and corticotropin-releasing factor receptor 1 (CRF1 ) and glucocorticoid receptor mRNA levels were significantly reduced in the hippocampus. Exercise normalized bladder sensitivity, micturition output, and increased brain-derived neurotrophic factor (BDNF) mRNA levels in the hippocampus of NMS mice. Mast cell degranulation was also normalized in NMS bladders following exercise. CONCLUSIONS Voluntary exercise normalized behavioral outcomes resulting from NMS in female mice, increased hippocampal BDNF mRNA levels, and decreased mast cell degranulation in the bladder. Together these results provide novel insight into the efficacy of voluntary exercise to attenuate comorbid outcomes resulting from exposure to early life stress.
Collapse
Affiliation(s)
- Angela N Pierce
- Department of Anatomy and Cell Biology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Olivia C Eller-Smith
- Department of Anatomy and Cell Biology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| | - Julie A Christianson
- Department of Anatomy and Cell Biology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
144
|
Nair AT, Ramachandran V, Joghee NM, Antony S, Ramalingam G. Gut Microbiota Dysfunction as Reliable Non-invasive Early Diagnostic Biomarkers in the Pathophysiology of Parkinson's Disease: A Critical Review. J Neurogastroenterol Motil 2018; 24:30-42. [PMID: 29291606 PMCID: PMC5753901 DOI: 10.5056/jnm17105] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/08/2017] [Accepted: 11/20/2017] [Indexed: 12/13/2022] Open
Abstract
Recent investigations suggest that gut microbiota affects the brain activity through the microbiota-gut-brain axis under both physiological and pathological disease conditions like Parkinson's disease. Further dopamine synthesis in the brain is induced by dopamine producing enzymes that are controlled by gut microbiota via the microbiota-gut-brain axis. Also alpha synuclein deposition and the associated neurodegeneration in the enteric nervous system that increase intestinal permeability, oxidative stress, and local inflammation, accounts for constipation in Parkinson's disease patients. The trigger that causes blood brain barrier leakage, immune cell activation and inflammation, and ultimately neuroinflammation in the central nervous system is believed to be due to the chronic low-grade inflammation in the gut. The non-motor symptoms that appear years before motor symptoms could be reliable early biomarkers, if they could be correlated with the established and reliable neuroimaging techniques or behavioral indices. The future directions should therefore, focus on the exploration of newer investigational techniques to identify these reliable early biomarkers and define the specific gut microbes that contribute to the development of Parkinson's disease. This ultimately should pave the way to safer and novel therapeutic approaches that avoid the complications of the drugs delivered today to the brain of Parkinson's disease patients.
Collapse
Affiliation(s)
- Arun T Nair
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research, Mysuru), Ootacamund, Tamilnadu,
India
| | - Vadivelan Ramachandran
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research, Mysuru), Ootacamund, Tamilnadu,
India
- Correspondence: Vadivelan Ramachandran, PhD, Department of Pharmacology, JSS College of Pharmacy ((JSS Academy of Higher Education and Research, Mysuru), Ootacamund, Tamilnadu 643001, India Tel: +91-9047539532, Fax: +91-423-2442937,
| | - Nanjan M Joghee
- JSS College of Pharmacy (JSS Academy of Higher Education and Research, Mysuru), Ootacamund, Tamilnadu,
India
| | - Shanish Antony
- Department of Pharmacology, Government Medical College, Kottayam, Kerala,
India
| | - Gopalakrishnan Ramalingam
- Department of Pharmacology, JSS College of Pharmacy (JSS Academy of Higher Education and Research, Mysuru), Ootacamund, Tamilnadu,
India
| |
Collapse
|
145
|
Wiley NC, Dinan TG, Ross RP, Stanton C, Clarke G, Cryan JF. The microbiota-gut-brain axis as a key regulator of neural function and the stress response: Implications for human and animal health. J Anim Sci 2018; 95:3225-3246. [PMID: 28727115 DOI: 10.2527/jas.2016.1256] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The brain-gut-microbiota axis comprises an extensive communication network between the brain, the gut, and the microbiota residing there. Development of a diverse gut microbiota is vital for multiple features of behavior and physiology, as well as many fundamental aspects of brain structure and function. Appropriate early-life assembly of the gut microbiota is also believed to play a role in subsequent emotional and cognitive development. If the composition, diversity, or assembly of the gut microbiota is impaired, this impairment can have a negative impact on host health and lead to disorders such as obesity, diabetes, inflammatory diseases, and even potentially neuropsychiatric illnesses, including anxiety and depression. Therefore, much research effort in recent years has focused on understanding the potential of targeting the intestinal microbiota to prevent and treat such disorders. This review aims to explore the influence of the gut microbiota on host neural function and behavior, particularly those of relevance to stress-related disorders. The involvement of microbiota in diverse neural functions such as myelination, microglia function, neuronal morphology, and blood-brain barrier integrity across the life span, from early life to adolescence to old age, will also be discussed. Nurturing an optimal gut microbiome may also prove beneficial in animal science as a means to manage stressful situations and to increase productivity of farm animals. The implications of these observations are manifold, and researchers are hopeful that this promising body of preclinical work can be successfully translated to the clinic and beyond.
Collapse
|
146
|
Fuentes IM, Pierce AN, Di Silvestro ER, Maloney MO, Christianson JA. Differential Influence of Early Life and Adult Stress on Urogenital Sensitivity and Function in Male Mice. Front Syst Neurosci 2018; 11:97. [PMID: 29379420 PMCID: PMC5771376 DOI: 10.3389/fnsys.2017.00097] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 12/19/2017] [Indexed: 12/21/2022] Open
Abstract
Experiences of adverse childhood events have been associated with improper output of the hypothalamic-pituitary-adrenal (HPA) axis in adulthood, as well as development of comorbid functional pain disorders. Symptoms of chronic prostatitis/chronic pelvic pain syndrome frequently overlap with those of interstitial cystitis/painful bladder syndrome and symptom severity is often triggered by stress. The objective of this study was to investigate the influence early life stress and acute adult stress on (1) perigenital sensitivity, (2) micturition, (3) anhedonia, and (4) HPA axis regulation and output in male C56Bl/6 mice. Neonatal maternal separation (NMS) was performed for 3 h a day from postnatal day 1 to 21 and naïve pups remained unhandled during this time. As adults, male mice were tested for referred prostate sensitivity and micturition patterning prior to and 1 and 8 days after exposure to 1 h of water avoidance stress (WAS). Following testing, prostate and bladder tissues were used for mast cell and Western blot analysis and RT-PCR was performed on mRNA from hypothalamus, amygdala, and hippocampus. Serum corticosterone (CORT) was also measured by enzyme-linked immunosorbent assay (ELISA). A significant increase in perigenital sensitivity and micturition frequency was observed in NMS mice and these measures were exacerbated by WAS exposure. Exposure to NMS significantly increased mast cell degranulation in both the bladder and prostate. Mast cell degranulation was also increased in naïve prostate tissue following WAS exposure. Cytokine mRNA levels were influenced by both NMS and WAS exposure, though WAS had a larger impact on central gene expression. Protein levels of CRF1 were differentially regulated by NMS and WAS in the bladder and prostate and serum CORT levels were significantly diminished following stress exposure. Taken together, these data suggest that NMS results in neurogenic inflammation and hypersensitivity within the urogenital organs, coupled with diminished gene expression and output from the HPA axis. Future studies of NMS in male mice may provide a useful tool as a preclinical model of male chronic urological pain syndromes for investigating potential pharmacological and interventional therapies.
Collapse
Affiliation(s)
- Isabella M Fuentes
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Angela N Pierce
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Elizabeth R Di Silvestro
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Molly O Maloney
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| | - Julie A Christianson
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
147
|
Experimental Models of Irritable Bowel Syndrome and the Role of the Enteric Neurotransmission. J Clin Med 2018; 7:jcm7010004. [PMID: 29301333 PMCID: PMC5791012 DOI: 10.3390/jcm7010004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 12/14/2017] [Accepted: 12/18/2017] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is one of the most common gastrointestinal diseases in humans. It is characterized by visceral pain and/or discomfort, hypersensitivity and abnormal motor responses along with change in gut habits. Although the etio-pathogenesis of IBS is only partially understood, a main role has been attributed to psychosocial stress of different origin. Animal models such as neonatal maternal separation, water avoidance stress and wrap restraint stress have been developed as psychosocial stressors in the attempt to reproduce the IBS symptomatology and identify the cellular mechanisms responsible for the disease. The study of these models has led to the production of drugs potentially useful for IBS treatment. This review intends to give an overview on the results obtained with the animal models; to emphasize the role of the enteric nervous system in IBS appearance and evolution and as a possible target of drug therapies.
Collapse
|
148
|
Grochowska M, Wojnar M, Radkowski M. The gut microbiota in neuropsychiatric disorders. Acta Neurobiol Exp (Wars) 2018. [DOI: 10.21307/ane-2018-008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
149
|
Burke NN, Fan CY, Trang T. Microglia in health and pain: impact of noxious early life events. Exp Physiol 2018; 101:1003-21. [PMID: 27474262 DOI: 10.1113/ep085714] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 05/27/2016] [Indexed: 01/08/2023]
Abstract
NEW FINDINGS What is the topic of this review? This review discusses the origins and development of microglia, and how stress, pain or inflammation in early life disturbs microglial function during critical developmental periods, leading to altered pain sensitivity and/or increased risk of chronic pain in later life. What advances does it highlight? We highlight recent advances in understanding how disrupted microglial function impacts the developing nervous system and the consequences for pain processing and susceptibility for development of chronic pain in later life. The discovery of microglia is accredited to Pío del Río-Hortega, who recognized this 'third element' of CNS cells as being morphologically distinct from neurons and astrocytes. For decades after this finding, microglia were altogether ignored or relegated as simply being support cells. Emerging from virtual obscurity, microglia have now gained notoriety as immune cells that assume a leading role in the development, maintenance and protection of a healthy CNS. Pioneering studies have recently shed light on the origins of microglia, their role in the developing nervous system and the complex roles they play beyond the immune response. These studies reveal that altered microglial function can have a profoundly negative impact on the developing brain and may be a determinant in a range of neurodevelopmental disorders and neurodegenerative diseases. The realization that aberrant microglial function also critically underlies chronic pain, a debilitating disorder that afflicts over 1.5 billion people worldwide, was a major conceptual leap forward in the pain field. Adding to this advance is emerging evidence that early life noxious experiences can have a long-lasting impact on central pain processing and adult pain sensitivity. With microglia now coming of age, in this review we examine the association between adverse early life events, such as stress, injury or inflammation, and the influence of sex differences, on the role of microglia in pain physiology in adulthood.
Collapse
Affiliation(s)
- Nikita N Burke
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Churmy Y Fan
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Tuan Trang
- Department of Comparative Biology and Experimental Medicine, Department of Physiology and Pharmacology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
150
|
Császár-Nagy N, Bókkon I. Mother-newborn separation at birth in hospitals: A possible risk for neurodevelopmental disorders? Neurosci Biobehav Rev 2018; 84:337-351. [DOI: 10.1016/j.neubiorev.2017.08.013] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/23/2017] [Accepted: 08/20/2017] [Indexed: 12/11/2022]
|