201
|
Malmuthuge N, Guan LL. Understanding the gut microbiome of dairy calves: Opportunities to improve early-life gut health. J Dairy Sci 2017; 100:5996-6005. [PMID: 28501408 DOI: 10.3168/jds.2016-12239] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 04/04/2017] [Indexed: 12/17/2022]
Abstract
Early gut microbiota plays a vital role in the long-term health of the host. However, understanding of these microbiota is very limited in livestock species, especially in dairy calves. Neonatal calves are highly susceptible to enteric infections, one of the major causes of calf death, so approaches to improving gut health and overall calf health are needed. An increasing number of studies are exploring the microbial composition of the gut, the mucosal immune system, and early dietary interventions to improve the health of dairy calves, revealing possibilities for effectively reducing the susceptibility of calves to enteric infections while promoting growth. Still, comprehensive understanding of the effect of dietary interventions on gut microbiota-one of the key aspects of gut health-is lacking. Such knowledge may provide in-depth understanding of the mechanisms behind functional changes in response to dietary interventions. Understanding of host-microbial interactions with dietary interventions and the role of the gut microbiota during pathogenesis at the site of infection in early life is vital for designing effective tools and techniques to improve calf gut health.
Collapse
Affiliation(s)
- Nilusha Malmuthuge
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5 Canada
| | - Le Luo Guan
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, T6G 2P5 Canada.
| |
Collapse
|
202
|
Buss C, Entringer S, Moog NK, Toepfer P, Fair DA, Simhan HN, Heim CM, Wadhwa PD. Intergenerational Transmission of Maternal Childhood Maltreatment Exposure: Implications for Fetal Brain Development. J Am Acad Child Adolesc Psychiatry 2017; 56:373-382. [PMID: 28433086 PMCID: PMC5402756 DOI: 10.1016/j.jaac.2017.03.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Revised: 03/01/2017] [Accepted: 03/02/2017] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Growing evidence suggests the deleterious consequences of exposure to childhood maltreatment (CM) not only might endure over the exposed individual's lifespan but also might be transmitted across generations. The time windows, mechanisms, and targets of such intergenerational transmission are poorly understood. The prevailing paradigm posits that mother-to-child transmission of the effects of maternal CM likely occurs after her child's birth. The authors seek to extend this paradigm and advance a transdisciplinary framework that integrates the concepts of biological embedding of life experiences and fetal origins of health and disease risk. METHOD The authors posit that the period of embryonic and fetal life represents a particularly sensitive time for intergenerational transmission; that the developing brain represents a target of particular interest; and that stress-sensitive maternal-placental-fetal biological (endocrine, immune) pathways represent leading candidate mechanisms of interest. RESULTS The plausibility of this model is supported by theoretical considerations and empirical findings in humans and animals. The authors synthesize several research areas and identify important knowledge gaps that might warrant further study. CONCLUSION The scientific and public health relevance of this effort relates to achieving a better understanding of the "when," "what," and "how" of intergenerational transmission of CM, with implications for early identification of risk, prevention, and intervention.
Collapse
Affiliation(s)
- Claudia Buss
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH); the University of California-Irvine; and the University of California-Irvine Development, Health and Disease Research Program, Orange, CA.
| | | | | | | | | | | | | | | |
Collapse
|
203
|
Thomas S, Izard J, Walsh E, Batich K, Chongsathidkiet P, Clarke G, Sela DA, Muller AJ, Mullin JM, Albert K, Gilligan JP, DiGuilio K, Dilbarova R, Alexander W, Prendergast GC. The Host Microbiome Regulates and Maintains Human Health: A Primer and Perspective for Non-Microbiologists. Cancer Res 2017; 77:1783-1812. [PMID: 28292977 PMCID: PMC5392374 DOI: 10.1158/0008-5472.can-16-2929] [Citation(s) in RCA: 215] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 02/07/2023]
Abstract
Humans consider themselves discrete autonomous organisms, but recent research is rapidly strengthening the appreciation that associated microorganisms make essential contributions to human health and well being. Each person is inhabited and also surrounded by his/her own signature microbial cloud. A low diversity of microorganisms is associated with a plethora of diseases, including allergy, diabetes, obesity, arthritis, inflammatory bowel diseases, and even neuropsychiatric disorders. Thus, an interaction of microorganisms with the host immune system is required for a healthy body. Exposure to microorganisms from the moment we are born and appropriate microbiome assembly during childhood are essential for establishing an active immune system necessary to prevent disease later in life. Exposure to microorganisms educates the immune system, induces adaptive immunity, and initiates memory B and T cells that are essential to combat various pathogens. The correct microbial-based education of immune cells may be critical in preventing the development of autoimmune diseases and cancer. This review provides a broad overview of the importance of the host microbiome and accumulating knowledge of how it regulates and maintains a healthy human system. Cancer Res; 77(8); 1783-812. ©2017 AACR.
Collapse
Affiliation(s)
- Sunil Thomas
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania.
| | - Jacques Izard
- Department of Food Science and Technology, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Emily Walsh
- Department of Oral Medicine, Infection and Immunity, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Kristen Batich
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Surgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Pakawat Chongsathidkiet
- Department of Neurosurgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Surgery, Duke Brain Tumor Immunotherapy Program, Duke University Medical Center, Durham, North Carolina
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Gerard Clarke
- Department of Psychiatry and Neurobehavioural Science, APC Microbiome Institute University College Cork, Cork, Ireland
| | - David A Sela
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
- Department of Microbiology, University of Massachusetts, Amherst, Massachusetts
- Center for Microbiome Research, University of Massachusetts Medical School, Worcester, Massachusetts
| | | | - James M Mullin
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Korin Albert
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts
- Department of Food Science, University of Massachusetts, Amherst, Massachusetts
| | - John P Gilligan
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | | - Rima Dilbarova
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | - Walker Alexander
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania
| | | |
Collapse
|
204
|
The Central Nervous System and the Gut Microbiome. Cell 2017; 167:915-932. [PMID: 27814521 DOI: 10.1016/j.cell.2016.10.027] [Citation(s) in RCA: 889] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/14/2016] [Accepted: 10/16/2016] [Indexed: 12/11/2022]
Abstract
Neurodevelopment is a complex process governed by both intrinsic and extrinsic signals. While historically studied by researching the brain, inputs from the periphery impact many neurological conditions. Indeed, emerging data suggest communication between the gut and the brain in anxiety, depression, cognition, and autism spectrum disorder (ASD). The development of a healthy, functional brain depends on key pre- and post-natal events that integrate environmental cues, such as molecular signals from the gut. These cues largely originate from the microbiome, the consortium of symbiotic bacteria that reside within all animals. Research over the past few years reveals that the gut microbiome plays a role in basic neurogenerative processes such as the formation of the blood-brain barrier, myelination, neurogenesis, and microglia maturation and also modulates many aspects of animal behavior. Herein, we discuss the biological intersection of neurodevelopment and the microbiome and explore the hypothesis that gut bacteria are integral contributors to development and function of the nervous system and to the balance between mental health and disease.
Collapse
|
205
|
Dying for love: Perimenopausal degeneration of vaginal microbiome drives the chronic inflammation-malignant transformation of benign prostatic hyperplasia to prostatic adenocarcinoma. Med Hypotheses 2017; 101:44-47. [DOI: 10.1016/j.mehy.2017.02.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2016] [Revised: 02/11/2017] [Accepted: 02/13/2017] [Indexed: 12/14/2022]
|
206
|
Moya-Pérez A, Luczynski P, Renes IB, Wang S, Borre Y, Anthony Ryan C, Knol J, Stanton C, Dinan TG, Cryan JF. Intervention strategies for cesarean section-induced alterations in the microbiota-gut-brain axis. Nutr Rev 2017; 75:225-240. [PMID: 28379454 PMCID: PMC5410982 DOI: 10.1093/nutrit/nuw069] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Microbial colonization of the gastrointestinal tract is an essential process that modulates host physiology and immunity. Recently, researchers have begun to understand how and when these microorganisms colonize the gut and the early-life factors that impact their natural ecological establishment. The vertical transmission of maternal microbes to the offspring is a critical factor for host immune and metabolic development. Increasing evidence also points to a role in the wiring of the gut-brain axis. This process may be altered by various factors such as mode of delivery, gestational age at birth, the use of antibiotics in early life, infant feeding, and hygiene practices. In fact, these early exposures that impact the intestinal microbiota have been associated with the development of diseases such as obesity, type 1 diabetes, asthma, allergies, and even neurodevelopmental disorders. The present review summarizes the impact of cesarean birth on the gut microbiome and the health status of the developing infant and discusses possible preventative and restorative strategies to compensate for early-life microbial perturbations.
Collapse
Affiliation(s)
- Angela Moya-Pérez
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Pauline Luczynski
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Ingrid B. Renes
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Shugui Wang
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Yuliya Borre
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - C. Anthony Ryan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Jan Knol
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Catherine Stanton
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - Timothy G. Dinan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| | - John F. Cryan
- A. Moya-Pérez, P. Luczynski, Y. Borre, C.A. Ryan, C. Stanton, T.G. Dinan, and J.F. Cryan are with the APC Microbiome Institute; C.A. Ryan is with the Department of Paediatrics and Child Health; T.G. Dinan is with the Department of Psychiatry and Neurobehavioural Science; and J.F. Cryan is with the Department of Anatomy and Neuroscience; University College Cork, Cork, Ireland. I.B. Renes and J. Knol are with Nutricia Research, Utrecht, the Netherlands. S. Wang is with Nutricia Research, Singapore. J. Knol is with the Laboratory of Microbiology, Wageningen University, Wageningen, the Netherlands. C. Stanton is with the Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland
| |
Collapse
|
207
|
Smith CC, Srygley RB, Dietrich EI, Mueller UG. Partitioning the effects of mating and nuptial feeding on the microbiome in gift-giving insects. ENVIRONMENTAL MICROBIOLOGY REPORTS 2017; 9:104-112. [PMID: 27894162 DOI: 10.1111/1758-2229.12506] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 10/27/2016] [Indexed: 06/06/2023]
Abstract
Mating is a ubiquitous social interaction with the potential to influence the microbiome by facilitating transmission, modifying host physiology, and in species where males donate nuptial gifts to females, altering diet. We manipulated mating and nuptial gift consumption in two insects that differ in nuptial gift size, the Mormon cricket Anabrus simplex and the decorated cricket Gryllodes sigillatus, with the expectation that larger gifts are more likely to affect the gut microbiome. Surprisingly, mating, but not nuptial gift consumption, affected the structure of bacterial communities in the gut, and only in Mormon crickets. The change in structure was due to a precipitous drop in the abundance of lactic-acid bacteria in unmated females, a taxon known for their beneficial effects on nutrition and immunity. Mating did not affect phenoloxidase or lysozyme-like antibacterial activity in either species, suggesting that any physiological response to mating on host-microbe interactions is decoupled from systemic immunity. Protein supplementation also did not affect the gut microbiome in decorated crickets, suggesting that insensitivity of gut microbes to dietary protein could contribute to the lack of an effect of nuptial gift consumption. Our study provides experimental evidence that sexual interactions can affect the microbiome and suggests mating can promote beneficial gut bacteria.
Collapse
Affiliation(s)
- Chad C Smith
- Department of Integrative Biology, 1 University Station C0990, University of Texas at Austin, Austin, TX, 78712, USA
| | - Robert B Srygley
- Northern Plains Area Research Laboratory, USDA-Agricultural Research Service, 1500 N. Central Avenue, Sidney, MT, 59270, USA
| | - Emma I Dietrich
- Department of Integrative Biology, 1 University Station C0990, University of Texas at Austin, Austin, TX, 78712, USA
| | - Ulrich G Mueller
- Department of Integrative Biology, 1 University Station C0990, University of Texas at Austin, Austin, TX, 78712, USA
| |
Collapse
|
208
|
Foster JA, Rinaman L, Cryan JF. Stress & the gut-brain axis: Regulation by the microbiome. Neurobiol Stress 2017; 7:124-136. [PMID: 29276734 PMCID: PMC5736941 DOI: 10.1016/j.ynstr.2017.03.001] [Citation(s) in RCA: 628] [Impact Index Per Article: 89.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/16/2017] [Accepted: 03/02/2017] [Indexed: 12/11/2022] Open
Abstract
The importance of the gut–brain axis in regulating stress-related responses has long been appreciated. More recently, the microbiota has emerged as a key player in the control of this axis, especially during conditions of stress provoked by real or perceived homeostatic challenge. Diet is one of the most important modifying factors of the microbiota-gut-brain axis. The routes of communication between the microbiota and brain are slowly being unravelled, and include the vagus nerve, gut hormone signaling, the immune system, tryptophan metabolism, and microbial metabolites such as short chain fatty acids. The importance of the early life gut microbiota in shaping later health outcomes also is emerging. Results from preclinical studies indicate that alterations of the early microbial composition by way of antibiotic exposure, lack of breastfeeding, birth by Caesarean section, infection, stress exposure, and other environmental influences - coupled with the influence of host genetics - can result in long-term modulation of stress-related physiology and behaviour. The gut microbiota has been implicated in a variety of stress-related conditions including anxiety, depression and irritable bowel syndrome, although this is largely based on animal studies or correlative analysis in patient populations. Additional research in humans is sorely needed to reveal the relative impact and causal contribution of the microbiome to stress-related disorders. In this regard, the concept of psychobiotics is being developed and refined to encompass methods of targeting the microbiota in order to positively impact mental health outcomes. At the 2016 Neurobiology of Stress Workshop in Newport Beach, CA, a group of experts presented the symposium “The Microbiome: Development, Stress, and Disease”. This report summarizes and builds upon some of the key concepts in that symposium within the context of how microbiota might influence the neurobiology of stress.
Collapse
Affiliation(s)
- Jane A Foster
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | - Linda Rinaman
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States
| | - John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland.,Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
209
|
Abstract
Objective: To systematically review the updated information about the gut microbiota-brain axis. Data Sources: All articles about gut microbiota-brain axis published up to July 18, 2016, were identified through a literature search on PubMed, ScienceDirect, and Web of Science, with the keywords of “gut microbiota”, “gut-brain axis”, and “neuroscience”. Study Selection: All relevant articles on gut microbiota and gut-brain axis were included and carefully reviewed, with no limitation of study design. Results: It is well-recognized that gut microbiota affects the brain's physiological, behavioral, and cognitive functions although its precise mechanism has not yet been fully understood. Gut microbiota-brain axis may include gut microbiota and their metabolic products, enteric nervous system, sympathetic and parasympathetic branches within the autonomic nervous system, neural-immune system, neuroendocrine system, and central nervous system. Moreover, there may be five communication routes between gut microbiota and brain, including the gut-brain's neural network, neuroendocrine-hypothalamic-pituitary-adrenal axis, gut immune system, some neurotransmitters and neural regulators synthesized by gut bacteria, and barrier paths including intestinal mucosal barrier and blood-brain barrier. The microbiome is used to define the composition and functional characteristics of gut microbiota, and metagenomics is an appropriate technique to characterize gut microbiota. Conclusions: Gut microbiota-brain axis refers to a bidirectional information network between the gut microbiota and the brain, which may provide a new way to protect the brain in the near future.
Collapse
Affiliation(s)
- Hong-Xing Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yu-Ping Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053; Center of Epilepsy, Beijing Institute for Brain Disorders, Laboratory of Brain Disorders, Capital Medical University, Beijing 100069, China
| |
Collapse
|
210
|
Stress during pregnancy alters temporal and spatial dynamics of the maternal and offspring microbiome in a sex-specific manner. Sci Rep 2017; 7:44182. [PMID: 28266645 PMCID: PMC5339804 DOI: 10.1038/srep44182] [Citation(s) in RCA: 168] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 02/06/2017] [Indexed: 02/07/2023] Open
Abstract
The microbiome is a regulator of host immunity, metabolism, neurodevelopment, and behavior. During early life, bacterial communities within maternal gut and vaginal compartments can have an impact on directing these processes. Maternal stress experience during pregnancy may impact offspring development by altering the temporal and spatial dynamics of the maternal microbiome during pregnancy. To examine the hypothesis that maternal stress disrupts gut and vaginal microbial dynamics during critical prenatal and postnatal windows, we used high-resolution 16S rRNA marker gene sequencing to examine outcomes in our mouse model of early prenatal stress. Consistent with predictions, maternal fecal communities shift across pregnancy, a process that is disrupted by stress. Vaginal bacterial community structure and composition exhibit lasting disruption following stress exposure. Comparison of maternal and offspring microbiota revealed that similarities in bacterial community composition was predicted by a complex interaction between maternal body niche and offspring age and sex. Importantly, early prenatal stress influenced offspring bacterial community assembly in a temporal and sex-specific manner. Taken together, our results demonstrate that early prenatal stress may influence offspring development through converging modifications to gut microbial composition during pregnancy and transmission of dysbiotic vaginal microbiome at birth.
Collapse
|
211
|
Microbiota alteration is associated with the development of stress-induced despair behavior. Sci Rep 2017; 7:43859. [PMID: 28266612 PMCID: PMC5339726 DOI: 10.1038/srep43859] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 01/31/2017] [Indexed: 12/29/2022] Open
Abstract
Depressive disorders often run in families, which, in addition to the genetic component, may point to the microbiome as a causative agent. Here, we employed a combination of behavioral, molecular and computational techniques to test the role of the microbiota in mediating despair behavior. In chronically stressed mice displaying despair behavior, we found that the microbiota composition and the metabolic signature dramatically change. Specifically, we observed reduced Lactobacillus and increased circulating kynurenine levels as the most prominent changes in stressed mice. Restoring intestinal Lactobacillus levels was sufficient to improve the metabolic alterations and behavioral abnormalities. Mechanistically, we identified that Lactobacillus-derived reactive oxygen species may suppress host kynurenine metabolism, by inhibiting the expression of the metabolizing enzyme, IDO1, in the intestine. Moreover, maintaining elevated kynurenine levels during Lactobacillus supplementation diminished the treatment benefits. Collectively, our data provide a mechanistic scenario for how a microbiota player (Lactobacillus) may contribute to regulating metabolism and resilience during stress.
Collapse
|
212
|
The microbiome-immune-host defense barrier complex (microimmunosome) and developmental programming of noncommunicable diseases. Reprod Toxicol 2017; 68:49-58. [DOI: 10.1016/j.reprotox.2016.04.026] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 04/15/2016] [Accepted: 04/29/2016] [Indexed: 12/29/2022]
|
213
|
Vuong HE, Hsiao EY. Emerging Roles for the Gut Microbiome in Autism Spectrum Disorder. Biol Psychiatry 2017; 81:411-423. [PMID: 27773355 PMCID: PMC5285286 DOI: 10.1016/j.biopsych.2016.08.024] [Citation(s) in RCA: 345] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/28/2016] [Accepted: 08/18/2016] [Indexed: 01/16/2023]
Abstract
Autism spectrum disorder (ASD) is a serious neurodevelopmental disorder that affects one in 45 children in the United States, with a similarly striking prevalence in countries around the world. However, mechanisms underlying its etiology and manifestations remain poorly understood. Although ASD is diagnosed based on the presence and severity of impaired social communication and repetitive behavior, immune dysregulation and gastrointestinal issues are common comorbidities. The microbiome is an integral part of human physiology; recent studies show that changes in the gut microbiota can modulate gastrointestinal physiology, immune function, and even behavior. Links between particular bacteria from the indigenous gut microbiota and phenotypes relevant to ASD raise the important question of whether microbial dysbiosis plays a role in the development or presentation of ASD symptoms. Here we review reports of microbial dysbiosis in ASD. We further discuss potential effects of the microbiota on ASD-associated symptoms, drawing on signaling mechanisms for reciprocal interactions among the microbiota, immunity, gut function, and behavior. In addition, we discuss recent findings supporting a role for the microbiome as an interface between environmental and genetic risk factors that are associated with ASD. These studies highlight the integration of pathways across multiple body systems that together can impact brain and behavior and suggest that changes in the microbiome may contribute to symptoms of neurodevelopmental disease.
Collapse
Affiliation(s)
- Helen E. Vuong
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA,Correspondence to: ; 610 Charles E. Young Drive MSB 3825A; Los Angeles CA 90095; 310-825-0228
| |
Collapse
|
214
|
Rakers F, Rupprecht S, Dreiling M, Bergmeier C, Witte OW, Schwab M. Transfer of maternal psychosocial stress to the fetus. Neurosci Biobehav Rev 2017; 117:S0149-7634(16)30719-9. [PMID: 28237726 DOI: 10.1016/j.neubiorev.2017.02.019] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/14/2017] [Accepted: 02/20/2017] [Indexed: 12/18/2022]
Abstract
Psychosocial maternal stress experienced during different vulnerable periods throughout gestation is thought to increase the individual's risk to develop neuropsychiatric, cardiovascular and metabolic disease in later life. Cortisol has generally been identified as the major mediator of maternal stress transfer to the fetus. Its lipophilic nature allows a trans-placental passage and thus excessive maternal cortisol could persistently impair the development of the fetal hypothalamic-pituitary-adrenal axis (HPAA). However, cortisol alone cannot fully explain all effects of maternal stress especially during early to mid pregnancy before maturation of the fetal HPAA has even begun and expression of fetal glucocorticoid receptors is limited. This review focuses on mediators of maternal fetal stress transfer that in addition to cortisol have been proposed as transmitters of maternal stress: catecholamines, cytokines, serotonin/tryptophan, reactive-oxygen-species and the maternal microbiota. We propose that the effects of psychosocial maternal stress on fetal development and health and disease in later life are not a consequence of a single pathway but are mediated by multiple stress-transfer mechanisms acting together in a synergistic manner.
Collapse
Affiliation(s)
- Florian Rakers
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Sven Rupprecht
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Michelle Dreiling
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Christoph Bergmeier
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Matthias Schwab
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| |
Collapse
|
215
|
Mackos AR, Maltz R, Bailey MT. The role of the commensal microbiota in adaptive and maladaptive stressor-induced immunomodulation. Horm Behav 2017; 88:70-78. [PMID: 27760302 PMCID: PMC5303636 DOI: 10.1016/j.yhbeh.2016.10.006] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 10/13/2016] [Accepted: 10/14/2016] [Indexed: 02/06/2023]
Abstract
Over the past decade, it has become increasingly evident that there are extensive bidirectional interactions between the body and its microbiota. These interactions are evident during stressful periods, where it is recognized that commensal microbiota community structure is significantly changed. Many different stressors, ranging from early life stressors to stressors administered during adulthood, lead to significant, community-wide differences in the microbiota. The mechanisms through which this occurs are not yet known, but it is known that commensal microbes can recognize, and respond to, mammalian hormones and neurotransmitters, including those that are involved with the physiological response to stressful stimuli. In addition, the physiological stress response also changes many aspects of gastrointestinal physiology that can impact microbial community composition. Thus, there are many routes through which microbial community composition might be disrupted during stressful periods. The implications of these disruptions in commensal microbial communities for host health are still not well understood, but the commensal microbiota have been linked to stressor-induced immunopotentiation. The role of the microbiota in stressor-induced immunopotentiation can be adaptive, such as when these microbes stimulate innate defenses against bacterial infection. However, the commensal microbiota can also lead to maladaptive immune responses during stressor-exposure. This is evident in animal models of colonic inflammation where stressor exposure increases the inflammation through mechanisms involving the microbiota. It is likely that during stressor exposure, immune cell functioning is regulated by combined effects of both neurotransmitters/hormones and commensal microbes. Defining this regulation should be a focus of future studies.
Collapse
Affiliation(s)
- Amy R Mackos
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States.
| | - Ross Maltz
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States; Department of Gastroenterology, Nationwide Children's Hospital, Columbus, OH 43205, United States
| | - Michael T Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, United States; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43210, United States
| |
Collapse
|
216
|
Moussaoui N, Jacobs JP, Larauche M, Biraud M, Million M, Mayer E, Taché Y. Chronic Early-life Stress in Rat Pups Alters Basal Corticosterone, Intestinal Permeability, and Fecal Microbiota at Weaning: Influence of Sex. J Neurogastroenterol Motil 2017; 23:135-143. [PMID: 27829577 PMCID: PMC5216644 DOI: 10.5056/jnm16105] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
Background/Aims Wistar rat dams exposed to limited nesting stress (LNS) from post-natal days (PND) 2 to 10 display erratic maternal behavior, and their pups show delayed maturation of the hypothalamic-pituitary-adrenal axis and impaired epithelial barrier at PND10 and a visceral hypersensitivity at adulthood. Little is known about the impact of early life stress on the offspring before adulthood and the influence of sex. We investigated whether male and female rats previously exposed to LNS displays at weaning altered corticosterone, intestinal permeability, and microbiota. Methods Wistar rat dams and litters were maintained from PND2 to 10 with limited nesting/bedding materials and thereafter reverted to normal housing up to weaning (PND21). Control litters had normal housing. At weaning, we monitored body weight, corticosterone plasma levels (enzyme immunoassay), in vivo intestinal to colon permeability (fluorescein isothiocyanate-dextran 4 kDa) and fecal microbiota (DNA extraction and amplification of the V4 region of the 16S ribosomal RNA gene). Results At weaning, LNS pups had hypercorticosteronemia and enhanced intestinal permeability with females > males while body weights were similar. LNS decreased fecal microbial diversity and induced a distinct composition characterized by increased abundance of Gram positive cocci and reduction of fiber-degrading, butyrate-producing, and mucus-resident microbes. Conclusions These data indicate that chronic exposure to LNS during the first week post-natally has sustained effects monitored at weaning including hypercorticosteronemia, a leaky gut, and dysbiosis. These alterations may impact on the susceptibility to develop visceral hypersensitivity in adult rats and have relevance to the development of irritable bowel syndrome in childhood.
Collapse
Affiliation(s)
- Nabila Moussaoui
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Jonathan P Jacobs
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Muriel Larauche
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mandy Biraud
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Mulugeta Million
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Emeran Mayer
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Yvette Taché
- G. Oppenheimer Center for Neurobiology of Stress and Resilience, CURE: Digestive Diseases Research Center, Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine and Brain Research Institute, University of California, and VA Greater Los Angeles Healthcare System, Los Angeles, California, USA
| |
Collapse
|
217
|
Douros K, Moustaki M, Tsabouri S, Papadopoulou A, Papadopoulos M, Priftis KN. Prenatal Maternal Stress and the Risk of Asthma in Children. Front Pediatr 2017; 5:202. [PMID: 28979893 PMCID: PMC5611367 DOI: 10.3389/fped.2017.00202] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Accepted: 09/05/2017] [Indexed: 01/12/2023] Open
Abstract
Emerging evidence indicate that maternal prenatal stress (MPS) can result in a range of long-term adverse effects in the offspring. The underlying mechanism of MPS is not fully understood. However, its complexity is emphasized by the number of purportedly involved pathways namely, placental deregulated metabolism of maternal steroids, impaired maturation of fetal HPA axis, imbalanced efflux of commensal bacteria across the placenta, and skewed immune development toward Th2. Fetal programming probably exerts a pivotal role in the end result of the above pathways through the modulation of gene expression. In this review, we highlight the current knowledge from epidemiological and experimental studies regarding the effects of MPS on asthma development in the offspring.
Collapse
Affiliation(s)
- Konstantinos Douros
- 3rd Department of Pediatrics, "Attikon" University General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria Moustaki
- Cystic Fibrosis Department, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Sophia Tsabouri
- Department of Paediatrics, School of Medicine, University of Ioannina, Ioannina, Greece
| | - Anna Papadopoulou
- 3rd Department of Pediatrics, "Attikon" University General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Marios Papadopoulos
- 3rd Department of Pediatrics, "Attikon" University General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Kostas N Priftis
- 3rd Department of Pediatrics, "Attikon" University General Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
218
|
Kynurenine pathway metabolism and the microbiota-gut-brain axis. Neuropharmacology 2017; 112:399-412. [DOI: 10.1016/j.neuropharm.2016.07.002] [Citation(s) in RCA: 311] [Impact Index Per Article: 44.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 06/30/2016] [Accepted: 07/04/2016] [Indexed: 02/07/2023]
|
219
|
Diaz Heijtz R. Fetal, neonatal, and infant microbiome: Perturbations and subsequent effects on brain development and behavior. Semin Fetal Neonatal Med 2016; 21:410-417. [PMID: 27255860 DOI: 10.1016/j.siny.2016.04.012] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The human gastrointestinal tract harbors a diverse and complex community of microbes, termed gut microbiota, that normally assemble during the first postnatal years of life. This evolution-driven process has been shown to contribute to the developmental programming of epithelial barrier function, gut homeostasis, and angiogenesis, as well as the development and function of the immune system. Research over the last few years has revealed that the actions of the gut microbiota have much wider effects on host physiology and development than originally believed, including the modulation of brain development and behavior. This article briefly reviews recent findings on the impact of the gut microbiota on brain development, and how disturbances in the assembly and maturation of the gut microbiota may impact development of motor, social, and cognitive functions. The potential link between microbiota and metabolic requirements of the developing brain is also considered.
Collapse
|
220
|
Witkin SS, Linhares IM. Why do lactobacilli dominate the human vaginal microbiota? BJOG 2016; 124:606-611. [DOI: 10.1111/1471-0528.14390] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2016] [Indexed: 01/14/2023]
Affiliation(s)
- SS Witkin
- Division of Immunology and Infectious Diseases; Department of Obstetrics and Gynecology; Weill Cornell Medicine; New York NY USA
| | - IM Linhares
- Department of Gynecology and Obstetrics; University of Sao Paulo Medical School; Sao Paulo Brazil
| |
Collapse
|
221
|
Degroote S, Hunting DJ, Baccarelli AA, Takser L. Maternal gut and fetal brain connection: Increased anxiety and reduced social interactions in Wistar rat offspring following peri-conceptional antibiotic exposure. Prog Neuropsychopharmacol Biol Psychiatry 2016; 71:76-82. [PMID: 27346743 PMCID: PMC6584952 DOI: 10.1016/j.pnpbp.2016.06.010] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/19/2016] [Accepted: 06/19/2016] [Indexed: 12/25/2022]
Abstract
BACKGROUND A growing body of evidence indicates that gut microbiota characteristics may be closely related to mental dysfunctions. However, no studies have investigated fetal brain development in relation to the maternal gut microbiota, despite the extensive use of antibiotics in obstetric practice. OBJECTIVE To determine how periconceptional exposure to SuccinylSulfaThiazole (SST), a non-absorbable antibiotic, can affect behavior in rat offspring. This antibiotic drug has previously been shown to substantially perturb the gut microbiota in rats following a 28-day exposure. METHODS Female Wistar rats were divided in two groups: control, or exposed during one month before breeding until gestational day 15 to a diet containing 1% SST. We administered behavioral tests to offspring, i.e., open field (post-natal day 20), social interactions (P25), marble burying (P30), elevated plus maze (P35), and prepulse inhibition of the acoustic startle reflex (sensory gating) (P45). RESULTS Both male and female offspring exposed peri-conceptionally to SST showed reduced social interactions, with a decrease of about half in time spent in social interactions compared to controls, reduced exploration of the open arm by 20% in the elevated plus maze test indicating increased anxiety and altered sensorimotor gating, with a 1.5-2-fold decrease in startle inhibition. CONCLUSION Maternal periconceptional exposure to SST provokes alterations in offspring behavior in the absence of maternal infection. Because we administered SST, a non-absorbable antibiotic, only to the dam, we conclude that these neurobehavioral alterations in the offspring are related to maternal gut microbiota alterations.
Collapse
Affiliation(s)
- Stéphanie Degroote
- Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada.
| | - Darel J. Hunting
- Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada
| | - Andrea A. Baccarelli
- Harvard School of Public Health, 677 Huntington Ave, Boston, MA 02115, United States
| | - Larissa Takser
- Faculté de Médecine et Sciences de la Santé, Université de Sherbrooke, 3001 12e Avenue Nord, Sherbrooke, QC J1H 5H3, Canada.
| |
Collapse
|
222
|
What’s bugging your teen?—The microbiota and adolescent mental health. Neurosci Biobehav Rev 2016; 70:300-312. [DOI: 10.1016/j.neubiorev.2016.06.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Revised: 06/04/2016] [Accepted: 06/06/2016] [Indexed: 02/08/2023]
|
223
|
|
224
|
Clarke G, Cryan JF. Preface: The Gut Microbiome and Behavior under the microscope: Where to focus? INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 131:xv-xxiii. [PMID: 27793229 DOI: 10.1016/s0074-7742(16)30165-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- G Clarke
- Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland.
| | - J F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
225
|
Principi N, Esposito S. Gut microbiota and central nervous system development. J Infect 2016; 73:536-546. [PMID: 27725185 DOI: 10.1016/j.jinf.2016.09.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 09/26/2016] [Accepted: 09/29/2016] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Gut dysbiosis has been associated with several clinically relevant conditions, including alterations of central nervous system (CNS) structure and function development. This review discussed aspects of the relationship between gut microbiota and the CNS during development. METHODS PubMed was used to search for all of the studies published over the last 15 years using the key word "microbiota" and "gut" or "intestinal" and "nervous system". More than 350 articles were found, and only those published in English and providing data on aspects related to neurologic diseases were included in the evaluation. RESULTS The data indicate that the gut microbiota influences CNS development and function and that gut dysbiosis is associated with significant neurological problems. However, most of these data have been collected in experimental animals and cannot be transferred to humans. Moreover, it is not definitively established whether neurologic diseases depend on a generic modification of the gut microbiota or whether a single bacterial phylum or species plays a specific role for any single condition. Furthermore, limited information exists regarding protective bacteria. CONCLUSIONS Both probiotics and prebiotics can have different impacts on CNS according to the microbial species or oligosaccharides that are administered. In humans, particularly in children, several factors may be important in conditioning gut microbiota modifications; unfortunately, most of these factors act simultaneously. More efforts are required to fully define both the array of complex behaviors that are influenced by the gut microbiota at the CNS level and the mechanisms involved.
Collapse
Affiliation(s)
- Nicola Principi
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Susanna Esposito
- Pediatric Highly Intensive Care Unit, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| |
Collapse
|
226
|
Mackos AR, Varaljay VA, Maltz R, Gur TL, Bailey MT. Role of the Intestinal Microbiota in Host Responses to Stressor Exposure. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2016; 131:1-19. [PMID: 27793214 DOI: 10.1016/bs.irn.2016.08.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Humans have coevolved over time to not only tolerate but also rely on trillions of microbes that aid in the development of our immune system, provide nutrients, break down potentially noxious substances, and act as a barrier against potentially pathogenic organisms. These microbes, collectively known as the microbiota, live in relatively stable communities on mucosal surfaces such as the respiratory tract and gastrointestinal tract. Changes to the microbiota are often transient, due to changes in diet, antibiotic exposure, and psychological stressor exposure. This chapter will discuss how psychological stressors can shape the intestinal microbial community and how these perturbations can contribute to stressor-induced changes in immune function, neurodevelopment, and behavioral deficits.
Collapse
Affiliation(s)
- A R Mackos
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - V A Varaljay
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States
| | - R Maltz
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; Nationwide Children's Hospital, Columbus, OH, United States
| | - T L Gur
- Wexner Medical Center at The Ohio State University, Columbus, OH, United States; The Institute for Behavioral Medicine Research (IBMR) at The Ohio State University, Columbus, OH, United States
| | - M T Bailey
- Center for Microbial Pathogenesis, The Research Institute at Nationwide Children's Hospital, Columbus, OH, United States; The Institute for Behavioral Medicine Research (IBMR) at The Ohio State University, Columbus, OH, United States; The Ohio State University College of Medicine, Columbus, OH, United States.
| |
Collapse
|
227
|
Callaghan BL, Cowan CSM, Richardson R. Treating Generational Stress. Psychol Sci 2016; 27:1171-80. [DOI: 10.1177/0956797616653103] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Accepted: 05/12/2016] [Indexed: 12/26/2022] Open
Affiliation(s)
- Bridget L. Callaghan
- School of Psychology, University of New South Wales
- Department of Psychology, Columbia University
- Department of Psychiatry, University of Melbourne
| | | | | |
Collapse
|
228
|
O'Callaghan TF, Ross RP, Stanton C, Clarke G. The gut microbiome as a virtual endocrine organ with implications for farm and domestic animal endocrinology. Domest Anim Endocrinol 2016; 56 Suppl:S44-55. [PMID: 27345323 DOI: 10.1016/j.domaniend.2016.05.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 05/06/2016] [Accepted: 05/11/2016] [Indexed: 12/21/2022]
Abstract
The gut microbiome exerts a marked influence on host physiology, and manipulation of its composition has repeatedly been shown to influence host metabolism and body composition. This virtual endocrine organ also has a role in the regulation of the plasma concentrations of tryptophan, an essential amino acid and precursor to serotonin, a key neurotransmitter within both the enteric and central nervous systems. Control over the hypothalamic-pituitary-adrenal axis also appears to be under the influence of the gut microbiota. This is clear from studies in microbiota-deficient germ-free animals with exaggerated responses to psychological stress that can be normalized by monocolonization with certain bacterial species including Bifidobacterium infantis. Therapeutic targeting of the gut microbiota may thus be useful in treating or preventing stress-related microbiome-gut-brain axis disorders and metabolic diseases, much the same way as redirections of metabolopathies can be achieved through more traditional endocrine hormone-based interventions. Moreover, the implications of these findings need to be considered in the context of farm and domestic animal physiology, behavior, and food safety.
Collapse
Affiliation(s)
- T F O'Callaghan
- Department of Biosciences, Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Microbiology, University College Cork, Cork, Ireland
| | - R P Ross
- APC Microbiome Institute, University College Cork, Cork, Ireland; College of Science Engineering and Food Science, University College Cork, Cork, Ireland
| | - C Stanton
- Department of Biosciences, Teagasc Moorepark Food Research Centre, Fermoy, Cork, Ireland; APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland
| | - G Clarke
- APC Microbiome Institute, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioral Science, University College Cork, Cork, Ireland.
| |
Collapse
|
229
|
Immune recognition and response to the intestinal microbiome in type 1 diabetes. J Autoimmun 2016; 71:10-8. [DOI: 10.1016/j.jaut.2016.02.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 02/03/2016] [Accepted: 02/04/2016] [Indexed: 12/19/2022]
|
230
|
Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, Wesselingh S. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol Psychiatry 2016; 21:738-48. [PMID: 27090305 PMCID: PMC4879184 DOI: 10.1038/mp.2016.50] [Citation(s) in RCA: 608] [Impact Index Per Article: 76.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 02/22/2016] [Accepted: 02/25/2016] [Indexed: 02/06/2023]
Abstract
The human body hosts an enormous abundance and diversity of microbes, which perform a range of essential and beneficial functions. Our appreciation of the importance of these microbial communities to many aspects of human physiology has grown dramatically in recent years. We know, for example, that animals raised in a germ-free environment exhibit substantially altered immune and metabolic function, while the disruption of commensal microbiota in humans is associated with the development of a growing number of diseases. Evidence is now emerging that, through interactions with the gut-brain axis, the bidirectional communication system between the central nervous system and the gastrointestinal tract, the gut microbiome can also influence neural development, cognition and behaviour, with recent evidence that changes in behaviour alter gut microbiota composition, while modifications of the microbiome can induce depressive-like behaviours. Although an association between enteropathy and certain psychiatric conditions has long been recognized, it now appears that gut microbes represent direct mediators of psychopathology. Here, we examine roles of gut microbiome in shaping brain development and neurological function, and the mechanisms by which it can contribute to mental illness. Further, we discuss how the insight provided by this new and exciting field of research can inform care and provide a basis for the design of novel, microbiota-targeted, therapies.
Collapse
Affiliation(s)
- G B Rogers
- South Australian Health and Medical Research Institute, Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - D J Keating
- South Australian Health and Medical Research Institute, Centre for Neuroscience and Department of Human Physiology, Flinders University, Adelaide, SA, Australia
| | - R L Young
- South Australian Health and Medical Research Institute, Department of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - M-L Wong
- South Australian Health and Medical Research Institute, Mind and Brain Theme, and Flinders University, Adelaide, SA, Australia
| | - J Licinio
- South Australian Health and Medical Research Institute, Mind and Brain Theme, and Flinders University, Adelaide, SA, Australia
| | - S Wesselingh
- South Australian Health and Medical Research Institute, Infection and Immunity Theme, School of Medicine, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
231
|
Brennan-Olsen SL, Pasco JA, Williams LJ, Hyde NK, Jacka FN. The Microbiome: A Biological Mechanism Underpinning the Social Gradient of Musculoskeletal Conditions? J Bone Miner Res 2016; 31:1315. [PMID: 27018041 DOI: 10.1002/jbmr.2844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 03/24/2016] [Indexed: 11/08/2022]
Affiliation(s)
- Sharon L Brennan-Olsen
- Deakin University, School of Medicine, Geelong, Australia.,Institute for Health and Ageing, Australian Catholic University, Melbourne, Australia.,Australian Institute for Musculoskeletal Science, Melbourne, Australia
| | - Julie A Pasco
- Deakin University, School of Medicine, Geelong, Australia.,The University of Melbourne, Melbourne, Australia
| | | | - Natalie K Hyde
- Deakin University, School of Medicine, Geelong, Australia
| | - Felice N Jacka
- Deakin University, School of Medicine, Geelong, Australia
| |
Collapse
|
232
|
Behavioural traits propagate across generations via segregated iterative-somatic and gametic epigenetic mechanisms. Nat Commun 2016; 7:11492. [PMID: 27173585 PMCID: PMC4869176 DOI: 10.1038/ncomms11492] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 04/01/2016] [Indexed: 12/12/2022] Open
Abstract
Parental behavioural traits can be transmitted by non-genetic mechanisms to the offspring. Although trait transmission via sperm has been extensively researched, epidemiological studies indicate the exclusive/prominent maternal transmission of many non-genetic traits. Since maternal conditions impact the offspring during gametogenesis and through fetal/early-postnatal life, the resultant phenotype is likely the aggregate of consecutive germline and somatic effects; a concept that has not been previously studied. Here, we dissected a complex maternally transmitted phenotype, reminiscent of comorbid generalized anxiety/depression, to elementary behaviours/domains and their transmission mechanisms in mice. We show that four anxiety/stress-reactive traits are transmitted via independent iterative-somatic and gametic epigenetic mechanisms across multiple generations. Somatic/gametic transmission alters DNA methylation at enhancers within synaptic genes whose functions can be linked to the behavioural traits. Traits have generation-dependent penetrance and sex specificity resulting in pleiotropy. A transmission-pathway-based concept can refine current inheritance models of psychiatric diseases and facilitate the development of better animal models and new therapeutic approaches. Physiological effects of psychological stress and infection in mothers can increase the incidence of anxiety and psychiatric diseases in offsprings and in subsequent generation. Here, Miklos Toth and colleagues show that intergenerational inheritance of neurological traits is propagated across multiple generations independently by parallel non-genetic mechanisms involving independent segregation of epigenetic specific loci.
Collapse
|
233
|
|
234
|
Murphy K, Mitchell CM. The Interplay of Host Immunity, Environment and the Risk of Bacterial Vaginosis and Associated Reproductive Health Outcomes. J Infect Dis 2016; 214 Suppl 1:S29-35. [PMID: 27056955 DOI: 10.1093/infdis/jiw140] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Bacterial vaginosis (BV) is one of the most common causes of vaginal symptoms in US women, but its causal mechanism has not yet been defined. BV is more prevalent in women who are immunosuppressed, and several risk factors for the development of BV are associated with lower quantities of immune mediators in vaginal fluid. In contrast, the poor reproductive health outcomes associated with BV, such as preterm birth and human immunodeficiency virus type 1 acquisition, are associated with increased levels of proinflammatory immune mediators in the genital tract. In this article, we discuss how variations in the host immune profile and environmental effects on host immunity may influence the risk of BV, as well as the risk of complications associated with BV.
Collapse
Affiliation(s)
- Kerry Murphy
- Division of Infectious Diseases, Albert Einstein College of Medicine, Bronx, New York
| | - Caroline M Mitchell
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston
| |
Collapse
|
235
|
Cryan JF. Stress and the Microbiota-Gut-Brain Axis: An Evolving Concept in Psychiatry. CANADIAN JOURNAL OF PSYCHIATRY. REVUE CANADIENNE DE PSYCHIATRIE 2016; 61:201-3. [PMID: 27254411 PMCID: PMC4794959 DOI: 10.1177/0706743716635538] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- John F Cryan
- APC Microbiome Institute, University College Cork, Cork, Ireland Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| |
Collapse
|
236
|
Brain-gut-microbiota axis: challenges for translation in psychiatry. Ann Epidemiol 2016; 26:366-72. [PMID: 27005587 DOI: 10.1016/j.annepidem.2016.02.008] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 02/01/2016] [Accepted: 02/26/2016] [Indexed: 12/14/2022]
Abstract
PURPOSE The accruing data linking the gut microbiome to the development and function of the central nervous system has been proposed as a paradigm shift in neuroscience. The gut microbiota can communicate with the brain via neuroimmune, neuroendocrine, and neural pathways comprising the brain-gut-microbiota axis. Dysfunctional neuroimmune pathways are implicated in stress-related psychiatric disorders. METHODS Using depression as our primary example, we review both the preclinical and clinical evidence supporting the possible role played by the gut microbiota in stress-related psychiatric disorders. We consider how this can inform future treatment strategies and outline the challenges and necessary studies for moving the field forward. RESULTS The role played by the gut microbiota has not been fully elucidated in psychiatric populations. Although tempting to speculate that psychiatric patients may benefit from therapeutic modulation of the brain-gut-microbiota axis, the translational applications of the results obtained in rodent studies have yet to be demonstrated. CONCLUSIONS Evidence of altered gut microbiota composition and function in psychiatric patients is limited and cannot be regarded as proven. Moreover the efficacy of targeting the gut microbiota has not yet been established, and needs further investigation.
Collapse
|
237
|
Abstract
PURPOSE OF REVIEW There is an increasing realization that the microorganisms which reside within our gut form part of a complex multidirectional communication network with the brain known as the microbiome-gut-brain axis. In this review, we focus on recent findings which support a role for this axis in modulating neurodevelopment and behavior. RECENT FINDINGS A growing body of research is uncovering that under homeostatic conditions and in response to internal and external stressors, the bacterial commensals of our gut can signal to the brain through a variety of mechanisms to influence processes such neurotransmission, neurogenesis, microglia activation, and modulate behavior. Moreover, the mechanisms underlying the ability of stress to modulate the microbiota and also for microbiota to change the set point for stress sensitivity are being unraveled. Dysregulation of the gut microbiota composition has been identified in a number of psychiatric disorders, including depression. This has led to the concept of bacteria that have a beneficial effect upon behavior and mood (psychobiotics) being proposed for potential therapeutic interventions. SUMMARY Understanding the mechanisms by which the bacterial commensals of our gut are involved in brain function may lead to the development of novel microbiome-based therapies for these mood and behavioral disorders.
Collapse
|
238
|
McCarthy MM. Multifaceted origins of sex differences in the brain. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150106. [PMID: 26833829 DOI: 10.1098/rstb.2015.0106] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2015] [Indexed: 12/18/2022] Open
Abstract
Studies of sex differences in the brain range from reductionistic cell and molecular analyses in animal models to functional imaging in awake human subjects, with many other levels in between. Interpretations and conclusions about the importance of particular differences often vary with differing levels of analyses and can lead to discord and dissent. In the past two decades, the range of neurobiological, psychological and psychiatric endpoints found to differ between males and females has expanded beyond reproduction into every aspect of the healthy and diseased brain, and thereby demands our attention. A greater understanding of all aspects of neural functioning will only be achieved by incorporating sex as a biological variable. The goal of this review is to highlight the current state of the art of the discipline of sex differences research with an emphasis on the brain and to contextualize the articles appearing in the accompanying special issue.
Collapse
Affiliation(s)
- Margaret M McCarthy
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
239
|
Jašarević E, Morrison KE, Bale TL. Sex differences in the gut microbiome-brain axis across the lifespan. Philos Trans R Soc Lond B Biol Sci 2016; 371:20150122. [PMID: 26833840 DOI: 10.1098/rstb.2015.0122] [Citation(s) in RCA: 186] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2015] [Indexed: 02/06/2023] Open
Abstract
In recent years, the bidirectional communication between the gut microbiome and the brain has emerged as a factor that influences immunity, metabolism, neurodevelopment and behaviour. Cross-talk between the gut and brain begins early in life immediately following the transition from a sterile in utero environment to one that is exposed to a changing and complex microbial milieu over a lifetime. Once established, communication between the gut and brain integrates information from the autonomic and enteric nervous systems, neuroendocrine and neuroimmune signals, and peripheral immune and metabolic signals. Importantly, the composition and functional potential of the gut microbiome undergoes many transitions that parallel dynamic periods of brain development and maturation for which distinct sex differences have been identified. Here, we discuss the sexually dimorphic development, maturation and maintenance of the gut microbiome-brain axis, and the sex differences therein important in disease risk and resilience throughout the lifespan.
Collapse
Affiliation(s)
- Eldin Jašarević
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathleen E Morrison
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tracy L Bale
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
240
|
O’ Mahony SM, Stilling RM, Dinan TG, Cryan JF. The microbiome and childhood diseases: Focus on brain-gut axis. ACTA ACUST UNITED AC 2015; 105:296-313. [DOI: 10.1002/bdrc.21118] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Siobhain M. O’ Mahony
- Department of Anatomy and Neuroscience, University College Cork; Cork Ireland
- Laboratory of Neurogastroenterology; APC Microbiome Institute; Cork Ireland
| | - Roman M. Stilling
- Laboratory of Neurogastroenterology; APC Microbiome Institute; Cork Ireland
| | - Timothy G. Dinan
- Laboratory of Neurogastroenterology; APC Microbiome Institute; Cork Ireland
- Department of Psychiatry and Neurobehavioural Science; University College Cork; Cork Ireland
| | - John F. Cryan
- Department of Anatomy and Neuroscience, University College Cork; Cork Ireland
- Laboratory of Neurogastroenterology; APC Microbiome Institute; Cork Ireland
| |
Collapse
|
241
|
Banks WA, Gray AM, Erickson MA, Salameh TS, Damodarasamy M, Sheibani N, Meabon JS, Wing EE, Morofuji Y, Cook DG, Reed MJ. Lipopolysaccharide-induced blood-brain barrier disruption: roles of cyclooxygenase, oxidative stress, neuroinflammation, and elements of the neurovascular unit. J Neuroinflammation 2015; 12:223. [PMID: 26608623 PMCID: PMC4660627 DOI: 10.1186/s12974-015-0434-1] [Citation(s) in RCA: 404] [Impact Index Per Article: 44.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Accepted: 11/14/2015] [Indexed: 01/31/2023] Open
Abstract
Background Disruption of the blood-brain barrier (BBB) occurs in many diseases and is often mediated by inflammatory and neuroimmune mechanisms. Inflammation is well established as a cause of BBB disruption, but many mechanistic questions remain. Methods We used lipopolysaccharide (LPS) to induce inflammation and BBB disruption in mice. BBB disruption was measured using 14C-sucrose and radioactively labeled albumin. Brain cytokine responses were measured using multiplex technology and dependence on cyclooxygenase (COX) and oxidative stress determined by treatments with indomethacin and N-acetylcysteine. Astrocyte and microglia/macrophage responses were measured using brain immunohistochemistry. In vitro studies used Transwell cultures of primary brain endothelial cells co- or tri-cultured with astrocytes and pericytes to measure effects of LPS on transendothelial electrical resistance (TEER), cellular distribution of tight junction proteins, and permeability to 14C-sucrose and radioactive albumin. Results In comparison to LPS-induced weight loss, the BBB was relatively resistant to LPS-induced disruption. Disruption occurred only with the highest dose of LPS and was most evident in the frontal cortex, thalamus, pons-medulla, and cerebellum with no disruption in the hypothalamus. The in vitro and in vivo patterns of LPS-induced disruption as measured with 14C-sucrose, radioactive albumin, and TEER suggested involvement of both paracellular and transcytotic pathways. Disruption as measured with albumin and 14C-sucrose, but not TEER, was blocked by indomethacin. N-acetylcysteine did not affect disruption. In vivo, the measures of neuroinflammation induced by LPS were mainly not reversed by indomethacin. In vitro, the effects on LPS and indomethacin were not altered when brain endothelial cells (BECs) were cultured with astrocytes or pericytes. Conclusions The BBB is relatively resistant to LPS-induced disruption with some brain regions more vulnerable than others. LPS-induced disruption appears is to be dependent on COX but not on oxidative stress. Based on in vivo and in vitro measures of neuroinflammation, it appears that astrocytes, microglia/macrophages, and pericytes play little role in the LPS-mediated disruption of the BBB.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Alicia M Gray
- University of Washington School of Medicine, Seattle, WA, USA.
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Therese S Salameh
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Mamatha Damodarasamy
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| | - Nader Sheibani
- Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - James S Meabon
- Mental Health Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Emily E Wing
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - Yoichi Morofuji
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA. .,Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA. .,Department of Neurosurgery, University of Nagasaki, Nagasaki, Japan.
| | - David G Cook
- Geriatric Research Education and Clinical Center-VA Puget Sound Health Care System, Seattle, WA, USA.
| | - May J Reed
- Division of Gerontology and Geriatric Medicine, Department of Internal Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
242
|
Overall KL. Behavior, biological markers, interspecific outcome assessments, and One Medicine. J Vet Behav 2015. [DOI: 10.1016/j.jveb.2015.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
243
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015; 9:392. [PMID: 26528128 PMCID: PMC4604320 DOI: 10.3389/fncel.2015.00392] [Citation(s) in RCA: 649] [Impact Index Per Article: 72.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 09/21/2015] [Indexed: 12/12/2022] Open
Abstract
The emerging links between our gut microbiome and the central nervous system (CNS) are regarded as a paradigm shift in neuroscience with possible implications for not only understanding the pathophysiology of stress-related psychiatric disorders, but also their treatment. Thus the gut microbiome and its influence on host barrier function is positioned to be a critical node within the brain-gut axis. Mounting preclinical evidence broadly suggests that the gut microbiota can modulate brain development, function and behavior by immune, endocrine and neural pathways of the brain-gut-microbiota axis. Detailed mechanistic insights explaining these specific interactions are currently underdeveloped. However, the concept that a "leaky gut" may facilitate communication between the microbiota and these key signaling pathways has gained traction. Deficits in intestinal permeability may underpin the chronic low-grade inflammation observed in disorders such as depression and the gut microbiome plays a critical role in regulating intestinal permeability. In this review we will discuss the possible role played by the gut microbiota in maintaining intestinal barrier function and the CNS consequences when it becomes disrupted. We will draw on both clinical and preclinical evidence to support this concept as well as the key features of the gut microbiota which are necessary for normal intestinal barrier function.
Collapse
Affiliation(s)
- John R Kelly
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Paul J Kennedy
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland
| | - John F Cryan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Anatomy and Neuroscience, University College Cork Cork, Ireland
| | - Timothy G Dinan
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Gerard Clarke
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Psychiatry and Neurobehavioural Science, University College Cork Cork, Ireland
| | - Niall P Hyland
- Laboratory of Neurogastroenterology, APC Microbiome Institute, University College Cork Cork, Ireland ; Department of Pharmacology and Therapeutics, University College Cork Cork, Ireland
| |
Collapse
|
244
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
245
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
246
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
247
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
248
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
249
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|
250
|
Kelly JR, Kennedy PJ, Cryan JF, Dinan TG, Clarke G, Hyland NP. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci 2015. [DOI: 10.3389/fncel.2015.00392 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023] Open
|