Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Ageing Res Rev ; 84: 101812, 2023 02.
Article En | MEDLINE | ID: mdl-36455790

The microbiota-gut-brain axis or simple gut-brain axis (GBA) is a complex and interactive bidirectional communication network linking the gut to the brain. Alterations in the composition of the gut microbiome have been linked to GBA dysfunction, central nervous system (CNS) inflammation, and dopaminergic degeneration, as those occurring in Parkinson's disease (PD). Besides inflammation, the activation of brain microglia is known to play a central role in the damage of dopaminergic neurons. Inflammation is attributed to the toxic effect of aggregated α-synuclein, in the brain of PD patients. It has been suggested that the α-synuclein misfolding might begin in the gut and spread "prion-like", via the vagus nerve into the lower brainstem and ultimately to the midbrain, known as the Braak hypothesis. In this review, we discuss how the microbiota-gut-brain axis and environmental influences interact with the immune system to promote a pro-inflammatory state that is involved in the initiation and progression of misfolded α-synuclein proteins and the beginning of the early non-motor symptoms of PD. Furthermore, we describe a speculative bidirectional model that explains how the enteric glia is involved in the initiation and spreading of inflammation, epithelial barrier disruption, and α-synuclein misfolding, finally reaching the central nervous system and contributing to neuroinflammatory processes involved with the initial non-motor symptoms of PD.


Brain-Gut Axis , Enteric Nervous System , Parkinson Disease , Humans , alpha-Synuclein/metabolism , Brain/metabolism , Brain-Gut Axis/physiology , Inflammation/metabolism , Neuroglia/metabolism , Neuroglia/pathology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Enteric Nervous System/microbiology , Enteric Nervous System/pathology
2.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Article En | MEDLINE | ID: mdl-34717799

The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via ß2-adrenergic receptors (ß2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.


Enteric Nervous System/microbiology , Enteric Nervous System/parasitology , Infections/microbiology , Infections/parasitology , Neurons/pathology , Neuroprotection , Organ Specificity , Yersinia pseudotuberculosis/physiology , Animals , Eosinophils/metabolism , Hematopoietic Stem Cells/metabolism , Immunity , Infections/immunology , Interleukin-13/metabolism , Interleukin-4/metabolism , Macrophages/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Strongyloides/physiology , Strongyloidiasis/genetics , Strongyloidiasis/immunology , Strongyloidiasis/parasitology , Transcriptome/genetics , Yersinia pseudotuberculosis Infections/genetics , Yersinia pseudotuberculosis Infections/immunology , Yersinia pseudotuberculosis Infections/microbiology
3.
Neuropharmacology ; 197: 108721, 2021 10 01.
Article En | MEDLINE | ID: mdl-34274348

For the last 20 years, researchers have focused their intention on the impact of gut microbiota in healthy and pathological conditions. This year (2021), more than 25,000 articles can be retrieved from PubMed with the keywords "gut microbiota and physiology", showing the constant progress and impact of gut microbes in scientific life. As a result, numerous therapeutic perspectives have been proposed to modulate the gut microbiota composition and/or bioactive factors released from microbes to restore our body functions. Currently, the gut is considered a primary site for the development of pathologies that modify brain functions such as neurodegenerative (Parkinson's, Alzheimer's, etc.) and metabolic (type 2 diabetes, obesity, etc.) disorders. Deciphering the mode of interaction between microbiota and the brain is a real original option to prevent (and maybe treat in the future) the establishment of gut-brain pathologies. The objective of this review is to describe recent scientific elements that explore the communication between gut microbiota and the brain by focusing our interest on the enteric nervous system (ENS) as an intermediate partner. The ENS, which is known as the "second brain", could be under the direct or indirect influence of the gut microbiota and its released factors (short-chain fatty acids, neurotransmitters, gaseous factors, etc.). Thus, in addition to their actions on tissue (adipose tissue, liver, brain, etc.), microbes can have an impact on local ENS activity. This potential modification of ENS function has global repercussions in the whole body via the gut-brain axis and represents a new therapeutic strategy. This article is part of the special Issue on 'Cross Talk between Periphery and the Brain'.


Brain-Gut Axis , Enteric Nervous System/physiopathology , Gastrointestinal Microbiome , Neurodegenerative Diseases/microbiology , Neurodegenerative Diseases/physiopathology , Animals , Enteric Nervous System/microbiology , Humans , Neurodegenerative Diseases/psychology
4.
J Clin Invest ; 131(13)2021 07 01.
Article En | MEDLINE | ID: mdl-34196310

The gut-brain axis (GBA) refers to the complex interactions between the gut microbiota and the nervous, immune, and endocrine systems, together linking brain and gut functions. Perturbations of the GBA have been reported in people with multiple sclerosis (pwMS), suggesting a possible role in disease pathogenesis and making it a potential therapeutic target. While research in the area is still in its infancy, a number of studies revealed that pwMS are more likely to exhibit altered microbiota, altered levels of short chain fatty acids and secondary bile products, and increased intestinal permeability. However, specific microbes and metabolites identified across studies and cohorts vary greatly. Small clinical and preclinical trials in pwMS and mouse models, in which microbial composition was manipulated through the use of antibiotics, fecal microbiota transplantation, and probiotic supplements, have provided promising outcomes in preventing CNS inflammation. However, results are not always consistent, and large-scale randomized controlled trials are lacking. Herein, we give an overview of how the GBA could contribute to MS pathogenesis, examine the different approaches tested to modulate the GBA, and discuss how they may impact neuroinflammation and demyelination in the CNS.


Gastrointestinal Microbiome , Multiple Sclerosis/therapy , Animals , Autoimmunity , Disease Models, Animal , Dysbiosis/immunology , Dysbiosis/physiopathology , Endocrine System/immunology , Endocrine System/physiopathology , Enteric Nervous System/immunology , Enteric Nervous System/microbiology , Enteric Nervous System/physiopathology , Fecal Microbiota Transplantation , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Gastrointestinal Microbiome/physiology , Humans , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Intestinal Mucosa/physiopathology , Models, Neurological , Multiple Sclerosis/etiology , Multiple Sclerosis/microbiology , Neuroimmunomodulation , Probiotics/therapeutic use
5.
Trends Microbiol ; 29(8): 686-699, 2021 08.
Article En | MEDLINE | ID: mdl-33309188

The gastrointestinal tract harbors an intrinsic neuronal network, the enteric nervous system (ENS). The ENS controls motility, fluid homeostasis, and blood flow, but also interacts with other components of the intestine such as epithelial and immune cells. Recent studies indicate that gut microbiota diversification, which occurs alongside postnatal ENS maturation, could be critical for the development and function of the ENS. Here we discuss the possibility that this functional relationship starts in utero, whereby the maternal microbiota would prime the developing ENS and shape its physiology. We review ENS/microbiota interactions and their modulation in physiological and pathophysiological contexts. While microbial modulation of the ENS physiology is now well established, further studies are required to understand the contribution of the gut microbiota to the development and pathology of the ENS and to reveal the precise mechanisms underlying microbiota-to-ENS communications.


Enteric Nervous System/physiology , Gastrointestinal Microbiome/genetics , Gene Expression Regulation, Bacterial , Homeostasis , Enteric Nervous System/immunology , Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Humans , Intestines/microbiology , Neurons/physiology
6.
Physiol Rep ; 8(21): e14611, 2020 11.
Article En | MEDLINE | ID: mdl-33185323

BACKGROUND: Intestinal bacteria have been increasingly shown to be involved in early postnatal development. Previous work has shown that intestinal bacteria are necessary for the structural development and intrinsic function of the enteric nervous system in early postnatal life. Furthermore, colonization with a limited number of bacteria appears to be sufficient for the formation of a normal enteric nervous system. We tested the hypothesis that common bacterial components could influence the programming of developing enteric neurons. METHODS: The developmental programming of enteric neurons was studied by isolating enteric neural crest-derived cells from the fetal gut of C57Bl/6 mice at embryonic day 15.5. After the establishment of the cell line, cultured enteric neuronal precursors were exposed to increasing concentrations of a panel of bacterial components including lipopolysaccharide, flagellin, and components of peptidoglycan. KEY RESULT: Exposure to bacterial components consistently affected proportions of enteric neuronal precursors that developed into nitrergic neurons. Furthermore, flagellin and D-gamma-Glu-mDAP were found to promote the development of serotonergic neurons. Proportions of dopaminergic neurons remained unchanged. Proliferation of neuronal precursor cells was significantly increased upon exposure to lipopolysaccharide and flagellin, while no significant changes were observed in the proportion of apoptotic neuronal precursors compared to baseline with exposure to any bacterial component. CONCLUSIONS AND INTERFACES: These findings suggest that bacterial components may influence the development of enteric neurons.


Bacteria/metabolism , Enteric Nervous System/cytology , Enteric Nervous System/microbiology , Nerve Tissue Proteins/metabolism , Neurons/cytology , Neurons/microbiology , Animals , Apoptosis , Cell Differentiation/physiology , Cells, Cultured , Enteric Nervous System/metabolism , Female , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/genetics , Neurons/metabolism , Pregnancy
7.
Am J Physiol Gastrointest Liver Physiol ; 319(5): G541-G548, 2020 11 01.
Article En | MEDLINE | ID: mdl-32902314

Recent studies on humans and their key experimental model, the mouse, have begun to uncover the importance of gastrointestinal (GI) microbiota and enteric nervous system (ENS) interactions during developmental windows spanning from conception to adolescence. Disruptions in GI microbiota and ENS during these windows by environmental factors, particularly antibiotic exposure, have been linked to increased susceptibility of the host to several diseases. Mouse models have provided new insights to potential signaling factors between the microbiota and ENS. We review very recent work on maturation of GI microbiota and ENS during three key developmental windows: embryogenesis, early postnatal, and postweaning periods. We discuss advances in understanding of interactions between the two systems and highlight research avenues for future studies.


Enteric Nervous System/growth & development , Enteric Nervous System/physiology , Gastrointestinal Microbiome/physiology , Animals , Enteric Nervous System/microbiology , Environment , Humans , Mice , Microbiota
8.
Infect Immun ; 88(9)2020 08 19.
Article En | MEDLINE | ID: mdl-32341116

The orchestration of host immune responses to enteric bacterial pathogens is a complex process involving the integration of numerous signals, including from the nervous system. Despite the recent progress in understanding the contribution of neuroimmune interactions in the regulation of inflammation, the mechanisms and effects of this communication during enteric bacterial infection are only beginning to be characterized. As part of this neuroimmune communication, neurons specialized to detect painful or otherwise noxious stimuli can respond to bacterial pathogens. Highlighting the complexity of these systems, the immunological consequences of sensory neuron activation can be either host adaptive or maladaptive, depending on the pathogen and organ system. These are but one of many types of neuroimmune circuits, with the vagus nerve and sympathetic innervation of numerous organs now known to modulate immune cell function and therefore dictate immunological outcomes during health and disease. Here, we review the evidence for neuroimmune communication in response to bacterial pathogens, and then discuss the consequences to host morbidity and mortality during infection of the gastrointestinal tract.


Enteric Nervous System/immunology , Enterobacteriaceae Infections/immunology , Gastrointestinal Microbiome/immunology , Gastrointestinal Tract/immunology , Neuroimmunomodulation/genetics , Sensory Receptor Cells/immunology , Animals , Calcitonin Gene-Related Peptide/genetics , Calcitonin Gene-Related Peptide/immunology , Citrobacter/growth & development , Citrobacter/immunology , Enteric Nervous System/microbiology , Enterobacteriaceae Infections/genetics , Enterobacteriaceae Infections/microbiology , Enterobacteriaceae Infections/pathology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/microbiology , Gene Expression Regulation/immunology , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Pathogen-Associated Molecular Pattern Molecules/immunology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Sensory Receptor Cells/microbiology , TRPA1 Cation Channel/genetics , TRPA1 Cation Channel/immunology , TRPV Cation Channels/genetics , TRPV Cation Channels/immunology , Toll-Like Receptors/genetics , Toll-Like Receptors/immunology
9.
Front Immunol ; 11: 189, 2020.
Article En | MEDLINE | ID: mdl-32256485

Background: Chorioamnionitis, inflammation of the fetal membranes during pregnancy, is often caused by intra-amniotic (IA) infection with single or multiple microbes. Chorioamnionitis can be either acute or chronic and is associated with adverse postnatal outcomes of the intestine, including necrotizing enterocolitis (NEC). Neonates with NEC have structural and functional damage to the intestinal mucosa and the enteric nervous system (ENS), with loss of enteric neurons and glial cells. Yet, the impact of acute, chronic, or repetitive antenatal inflammatory stimuli on the development of the intestinal mucosa and ENS has not been studied. The aim of this study was therefore to investigate the effect of acute, chronic, and repetitive microbial exposure on the intestinal mucosa, submucosa and ENS in premature lambs. Materials and Methods: A sheep model of pregnancy was used in which the ileal mucosa, submucosa, and ENS were assessed following IA exposure to lipopolysaccharide (LPS) for 2 or 7 days (acute), Ureaplasma parvum (UP) for 42 days (chronic), or repetitive microbial exposure (42 days UP with 2 or 7 days LPS). Results: IA LPS exposure for 7 days or IA UP exposure for 42 days caused intestinal injury and inflammation in the mucosal and submucosal layers of the gut. Repetitive microbial exposure did not further aggravate injury of the terminal ileum. Chronic IA UP exposure caused significant structural ENS alterations characterized by loss of PGP9.5 and S100ß immunoreactivity, whereas these changes were not found after re-exposure of chronic UP-exposed fetuses to LPS for 2 or 7 days. Conclusion: The in utero loss of PGP9.5 and S100ß immunoreactivity following chronic UP exposure corresponds with intestinal changes in neonates with NEC and may therefore form a novel mechanistic explanation for the association of chorioamnionitis and NEC.


Chorioamnionitis/veterinary , Enteric Nervous System/injuries , Enteric Nervous System/microbiology , Enterocolitis, Necrotizing/veterinary , Fetus/microbiology , Sheep/embryology , Ureaplasma Infections/complications , Ureaplasma Infections/veterinary , Ureaplasma , Animals , Animals, Newborn , Chorioamnionitis/chemically induced , Chorioamnionitis/microbiology , Chronic Disease/veterinary , Disease Models, Animal , Enteric Nervous System/drug effects , Enterocolitis, Necrotizing/chemically induced , Enterocolitis, Necrotizing/microbiology , Female , Intestinal Mucosa/drug effects , Intestinal Mucosa/microbiology , Lipopolysaccharides/pharmacology , Pregnancy , Premature Birth/veterinary , S100 Calcium Binding Protein beta Subunit/metabolism , Sheep/microbiology , Ubiquitin Thiolesterase/metabolism , Ureaplasma Infections/microbiology
10.
Front Immunol ; 11: 602070, 2020.
Article En | MEDLINE | ID: mdl-33552060

Galanin (GAL) is a broad-spectrum peptide that was first identified 37 years ago. GAL, which acts through three specific receptor subtypes, is one of the most important molecules on an ever-growing list of neurotransmitters. Recent studies indicate that this peptide is commonly present in the gastrointestinal (GI) tract and GAL distribution can be seen in the enteric nervous system (ENS). The function of the GAL in the gastrointestinal tract is, inter alia, to regulate motility and secretion. It should be noted that the distribution of neuropeptides is largely dependent on the research model, as well as the part of the gastrointestinal tract under study. During the development of digestive disorders, fluctuations in GAL levels were observed. The occurrence of GAL largely depends on the stage of the disease, e.g., in porcine experimental colitis GAL secretion is caused by infection with Brachyspira hyodysenteriae. Many authors have suggested that increased GAL presence is related to the involvement of GAL in organ renewal. Additionally, it is tempting to speculate that GAL may be used in the treatment of gastroenteritis. This review aims to present the function of GAL in the mammalian gastrointestinal tract under physiological conditions. In addition, since GAL is undoubtedly involved in the regulation of inflammatory processes, and the aim of this publication is to provide up-to-date knowledge of the distribution of GAL in experimental models of gastrointestinal inflammation, which may help to accurately determine the role of this peptide in inflammatory diseases and its potential future use in the treatment of gastrointestinal disorders.


Enteric Nervous System/immunology , Galanin/immunology , Gastrointestinal Tract/immunology , Animals , Brachyspira hyodysenteriae/immunology , Colitis/immunology , Colitis/microbiology , Colitis/pathology , Enteric Nervous System/microbiology , Enteric Nervous System/pathology , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/microbiology , Gram-Negative Bacterial Infections/pathology , Humans , Inflammation/immunology , Inflammation/microbiology , Inflammation/pathology , Swine
11.
Article En | MEDLINE | ID: mdl-30858329

The small intestine is the longest organ in the human body, spanning a length of ∼5 m and compartmentalized into three distinct regions with specific roles in maintenance of comprehensive homeostasis. Along its length exists as a unique and independent system-called the enteric nervous system (ENS)-which coordinates the multitude of functions continuously around the clock. Yet, with so many vital roles played, the functions, relationships, and roles of the small intestine and ENS remain largely elusive. This fundamental hole in the physiology of the small intestine and ENS introduces a substantial number of challenges when attempting to create bioelectronic approaches for treatment of various disorders originating in the small intestine. Here, we review existing therapeutic options for modulating the small intestine, discuss fundamental gaps that must be addressed, and highlight novel methods and approaches to consider for development of bioelectronic approaches aiming to modulate the small intestine.


Enteric Nervous System/physiology , Gastrointestinal Microbiome/physiology , Intestine, Small/physiology , Animals , Enteric Nervous System/microbiology , Homeostasis , Humans , Intestine, Small/microbiology
12.
Physiol Rev ; 99(4): 1877-2013, 2019 10 01.
Article En | MEDLINE | ID: mdl-31460832

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.


Bacteria/metabolism , Brain Diseases/microbiology , Brain/microbiology , Gastrointestinal Microbiome , Intestines/microbiology , Age Factors , Aging , Animals , Bacteria/immunology , Bacteria/pathogenicity , Behavior , Brain/immunology , Brain/metabolism , Brain/physiopathology , Brain Diseases/metabolism , Brain Diseases/physiopathology , Brain Diseases/psychology , Dysbiosis , Enteric Nervous System/metabolism , Enteric Nervous System/microbiology , Enteric Nervous System/physiopathology , Host-Pathogen Interactions , Humans , Intestines/immunology , Neuroimmunomodulation , Neuronal Plasticity , Risk Factors
13.
J Neuroendocrinol ; 31(5): e12684, 2019 05.
Article En | MEDLINE | ID: mdl-30614568

The gut microbiota has emerged as an environmental factor that modulates the development of the central nervous system (CNS) and the enteric nervous system (ENS). Before obtaining its own microbiota, eutherian foetuses are exposed to products and metabolites from the maternal microbiota. At birth, the infants are colonised by microorganisms. The microbial composition in early life is strongly influenced by the mode of delivery, the feeding method, the use of antibiotics and the maternal microbial composition. Microbial products and microbially produced metabolites act as signalling molecules that have direct or indirect effects on the CNS and the ENS. An increasing number of studies show that the gut microbiota can modulate important processes during development, including neurogenesis, myelination, glial cell function, synaptic pruning and blood-brain barrier permeability. Furthermore, numerous studies indicate that there is a developmental window early in life during which the gut microbial composition is crucial and perturbation of the gut microbiota during this period causes long-lasting effects on the development of the CNS and the ENS. However, other functions are readily modulated in adult animals, including microglia activation and neuroinflammation. Several neurobehavioural, neurodegenerative, mental and metabolic disorders, including Parkinson disease, autism spectrum disorder, schizophrenia, Alzheimer's disease, depression and obesity, have been linked to the gut microbiota. This review focuses on the role of the microorganisms in the development and function of the CNS and the ENS, as well as their potential role in pathogenesis.


Central Nervous System Diseases/microbiology , Central Nervous System/microbiology , Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Animals , Central Nervous System/growth & development , Central Nervous System/physiopathology , Enteric Nervous System/growth & development , Enteric Nervous System/physiopathology , Humans
14.
J Parkinsons Dis ; 8(s1): S31-S39, 2018.
Article En | MEDLINE | ID: mdl-30584161

In the last two decades it has become clear that Parkinson's disease (PD) is associated with a plethora of gastrointestinal symptoms originating from functional and structural changes in the gut and its associated neural structures. This is of particular interest not only because such symptoms have a major impact on the quality of life of PD patients, but also since accumulating evidence suggests that in at least a subgroup of patients, these disturbances precede the motor symptoms and diagnosis of PD by years and may thus give important insights into the origin and pathogenesis of the disease. In this mini-review we attempt to concisely summarize the current knowledge after two decades of research on the gut-brain axis in PD. We focus on alpha-synuclein pathology, biomarkers, and the gut microbiota and envision the development and impact of these research areas for the two decades to come.


Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Parkinson Disease/microbiology , Biomarkers , Brain/metabolism , Enteric Nervous System/metabolism , Humans , Parkinson Disease/metabolism , alpha-Synuclein/metabolism
15.
Proc Natl Acad Sci U S A ; 115(25): 6458-6463, 2018 06 19.
Article En | MEDLINE | ID: mdl-29866843

The enteric nervous system (ENS) is crucial for essential gastrointestinal physiologic functions such as motility, fluid secretion, and blood flow. The gut is colonized by trillions of bacteria that regulate host production of several signaling molecules including serotonin (5-HT) and other hormones and neurotransmitters. Approximately 90% of 5-HT originates from the intestine, and activation of the 5-HT4 receptor in the ENS has been linked to adult neurogenesis and neuroprotection. Here, we tested the hypothesis that the gut microbiota could induce maturation of the adult ENS through release of 5-HT and activation of 5-HT4 receptors. Colonization of germ-free mice with a microbiota from conventionally raised mice modified the neuroanatomy of the ENS and increased intestinal transit rates, which was associated with neuronal and mucosal 5-HT production and the proliferation of enteric neuronal progenitors in the adult intestine. Pharmacological modulation of the 5-HT4 receptor, as well as depletion of endogenous 5-HT, identified a mechanistic link between the gut microbiota and maturation of the adult ENS through the release of 5-HT and activation of the 5-HT4 receptor. Taken together, these findings show that the microbiota modulates the anatomy of the adult ENS in a 5-HT-dependent fashion with concomitant changes in intestinal transit.


Enteric Nervous System/microbiology , Enteric Nervous System/physiology , Gastrointestinal Microbiome/physiology , Intestine, Small/microbiology , Serotonin/metabolism , Animals , Enteric Nervous System/metabolism , Female , Gastrointestinal Motility/physiology , Intestine, Small/metabolism , Mice , Mice, Inbred C57BL , Microbiota/physiology , Neurogenesis/physiology , Neurons/metabolism , Neurons/microbiology , Receptors, Serotonin, 5-HT4/metabolism
16.
Acta Neuropathol ; 136(3): 345-361, 2018 09.
Article En | MEDLINE | ID: mdl-29797112

Neurological diseases, such as Parkinson's disease, Alzheimer's disease, amyotrophic lateral sclerosis (ALS) and multiple sclerosis, are often associated with functional gastrointestinal disorders. These gastrointestinal disturbances may occur at all stages of the neurodegenerative diseases, to such an extent that they are now considered an integral part of their clinical picture. Several lines of evidence support the contention that, in central neurodegenerative diseases, changes in gut microbiota and enteric neuro-immune system alterations could contribute to gastrointesinal dysfunctions as well as initiation and upward spreading of the neurologic disorder. The present review has been intended to provide a comprehensive overview of the available knowledge on the role played by enteric microbiota, mucosal immune system and enteric nervous system, considered as an integrated network, in the pathophysiology of the main neurological diseases known to be associated with intestinal disturbances. In addition, based on current human and pre-clinical evidence, our intent was to critically discuss whether changes in the dynamic interplay between gut microbiota, intestinal epithelial barrier and enteric neuro-immune system are a consequence of the central neurodegeneration or might represent the starting point of the neurodegenerative process. Special attention has been paid also to discuss whether alterations of the enteric bacterial-neuro-immune network could represent a common path driving the onset of the main neurodegenerative diseases, even though each disease displays its own distinct clinical features.


Enteric Nervous System/microbiology , Gastrointestinal Microbiome/physiology , Intestinal Mucosa/microbiology , Neurodegenerative Diseases/microbiology , Animals , Enteric Nervous System/pathology , Humans , Intestinal Mucosa/pathology , Neurodegenerative Diseases/pathology
17.
Cell Mol Life Sci ; 75(7): 1145-1149, 2018 04.
Article En | MEDLINE | ID: mdl-29285574

Post-infectious irritable bowel syndrome is a well-defined pathological entity that develops in about one-third of subjects after an acute infection (bacterial, viral) or parasitic infestation. Only recently it has been documented that an high incidence of post-infectious irritable bowel syndrome occurs after Clostridium difficile infection. However, until now it is not known why in some patients recovered from this infection the gastrointestinal disturbances persist for months or years. Based on our in vitro studies on enteric glial cells exposed to the effects of C. difficile toxin B, we hypothesize that persistence of symptoms up to the development of irritable bowel syndrome might be due to a disturbance/impairment of the correct functions of the enteroglial intestinal network.


Clostridioides difficile/physiology , Clostridium Infections/microbiology , Enteric Nervous System/microbiology , Irritable Bowel Syndrome/microbiology , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Clostridioides difficile/metabolism , Host-Pathogen Interactions , Humans , Intestinal Mucosa/innervation , Intestinal Mucosa/microbiology , Models, Theoretical , Neuroglia/microbiology , Risk Factors
18.
J Diabetes Investig ; 9(2): 262-264, 2018 Mar.
Article En | MEDLINE | ID: mdl-29034596

Gut microbiota dysbiosis reduces expression of GLP-1 receptor (GLP-1R) and neuronal nitric oxide synthase (nNOS) in the enteric nervous system and hampers GLP-1-induced nitric oxide (NO) production through a pattern recognition receptor (PRR)-dependent mechanism, hence preventing activation of the gut-brain-periphery axis for control of insulin secretion and gastric emptying.


Dysbiosis/metabolism , Gastrointestinal Microbiome , Glucagon-Like Peptide 1/metabolism , Animals , Enteric Nervous System/metabolism , Enteric Nervous System/microbiology , Humans , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/metabolism
19.
PLoS One ; 12(7): e0181863, 2017.
Article En | MEDLINE | ID: mdl-28732069

BACKGROUND: We evaluated the effect of Saccharomyces boulardii CNCM I-745 on intestinal neuromuscular anomalies in an IBS-type mouse model of gastrointestinal motor dysfunctions elicited by Herpes Simplex Virus type 1 (HSV-1) exposure. METHODS: Mice were inoculated intranasally with HSV-1 (102 PFU) or vehicle at time 0 and 4 weeks later by the intragastric (IG) route (108 PFU). Six weeks after IG inoculum, mice were randomly allocated to receive oral gavage with either S. boulardii (107 CFU/day) or vehicle. After 4 weeks the following were determined: a) intestinal motility using fluorescein-isothiocyanate dextran distribution in the gut, fecal pellet expulsion, stool water content, and distal colonic transit of glass beads; b) integrity of the enteric nervous system (ENS) by immunohistochemistry on ileal whole-mount preparations and western blot of protein lysates from ileal longitudinal muscle and myenteric plexus; c) isometric muscle tension with electric field and pharmacological (carbachol) stimulation of ileal segments; and d) intestinal inflammation by levels of tumor necrosis factor α, interleukin(IL)-1ß, IL-10 and IL-4. RESULTS: S. boulardii CNCM I-745 improved HSV-1 induced intestinal dysmotility and alteration of intestinal transit observed ten weeks after IG inoculum of the virus. Also, the probiotic yeast ameliorated the structural alterations of the ENS induced by HSV-1 (i.e., reduced peripherin immunoreactivity and expression, increased glial S100ß protein immunoreactivity and neuronal nitric oxide synthase level, reduced substance P-positive fibers). Moreover, S. boulardii CNCM I-745 diminished the production of HSV-1 associated pro-inflammatory cytokines in the myenteric plexus and increased levels of anti-inflammatory interleukins. CONCLUSIONS: S. boulardii CNCM I-745 ameliorated gastrointestinal neuromuscular anomalies in a mouse model of gut dysfunctions typically observed with irritable bowel syndrome.


Gastrointestinal Motility/drug effects , Irritable Bowel Syndrome/microbiology , Irritable Bowel Syndrome/therapy , Probiotics/pharmacology , Saccharomyces boulardii/growth & development , Animals , Colon/metabolism , Colon/microbiology , Colon/virology , Cytokines/metabolism , Diarrhea/metabolism , Diarrhea/microbiology , Diarrhea/virology , Disease Models, Animal , Enteric Nervous System/metabolism , Enteric Nervous System/microbiology , Enteric Nervous System/virology , Herpes Simplex/metabolism , Herpes Simplex/microbiology , Herpes Simplex/virology , Herpesvirus 1, Human/pathogenicity , Ileum/metabolism , Ileum/microbiology , Ileum/virology , Inflammation/metabolism , Inflammation/microbiology , Inflammation/virology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Interleukin-4/metabolism , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/virology , Male , Mice , Mice, Inbred C57BL , Muscles/metabolism , Muscles/microbiology , Muscles/virology , Myenteric Plexus/metabolism , Myenteric Plexus/microbiology , Myenteric Plexus/virology , Tumor Necrosis Factor-alpha/metabolism
20.
Cell Metab ; 25(5): 1075-1090.e5, 2017 May 02.
Article En | MEDLINE | ID: mdl-28467926

Glucagon-like peptide-1 (GLP-1)-based therapies control glycemia in type 2 diabetic (T2D) patients. However, in some patients the treatment must be discontinued, defining a state of GLP-1 resistance. In animal models we identified a specific set of ileum bacteria impairing the GLP-1-activated gut-brain axis for the control of insulin secretion and gastric emptying. Using prediction algorithms, we identified bacterial pathways related to amino acid metabolism and transport system modules associated to GLP-1 resistance. The conventionalization of germ-free mice demonstrated their role in enteric neuron biology and the gut-brain-periphery axis. Altogether, insulin secretion and gastric emptying require functional GLP-1 receptor and neuronal nitric oxide synthase in the enteric nervous system within a eubiotic gut microbiota environment. Our data open a novel route to improve GLP-1-based therapies.


Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Dysbiosis/metabolism , Enteric Nervous System/metabolism , Gastrointestinal Microbiome , Nitric Oxide/metabolism , Animals , Brain/pathology , Diabetes Mellitus, Type 2/microbiology , Diabetes Mellitus, Type 2/pathology , Dysbiosis/microbiology , Dysbiosis/pathology , Enteric Nervous System/microbiology , Enteric Nervous System/pathology , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/pathology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Mice , Mice, Inbred C57BL
...