Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-34717799

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/parasitologia , Infecções/microbiologia , Infecções/parasitologia , Neurônios/patologia , Neuroproteção , Especificidade de Órgãos , Yersinia pseudotuberculosis/fisiologia , Animais , Eosinófilos/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Imunidade , Infecções/imunologia , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Strongyloides/fisiologia , Estrongiloidíase/genética , Estrongiloidíase/imunologia , Estrongiloidíase/parasitologia , Transcriptoma/genética , Infecções por Yersinia pseudotuberculosis/genética , Infecções por Yersinia pseudotuberculosis/imunologia , Infecções por Yersinia pseudotuberculosis/microbiologia
2.
Physiol Rev ; 99(4): 1877-2013, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31460832

RESUMO

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.


Assuntos
Bactérias/metabolismo , Encefalopatias/microbiologia , Encéfalo/microbiologia , Microbioma Gastrointestinal , Intestinos/microbiologia , Fatores Etários , Envelhecimento , Animais , Bactérias/imunologia , Bactérias/patogenicidade , Comportamento , Encéfalo/imunologia , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Encefalopatias/metabolismo , Encefalopatias/fisiopatologia , Encefalopatias/psicologia , Disbiose , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/fisiopatologia , Interações Hospedeiro-Patógeno , Humanos , Intestinos/imunologia , Neuroimunomodulação , Plasticidade Neuronal , Fatores de Risco
3.
Proc Natl Acad Sci U S A ; 115(25): 6458-6463, 2018 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-29866843

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestino Delgado/microbiologia , Serotonina/metabolismo , Animais , Sistema Nervoso Entérico/metabolismo , Feminino , Motilidade Gastrointestinal/fisiologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Microbiota/fisiologia , Neurogênese/fisiologia , Neurônios/metabolismo , Neurônios/microbiologia , Receptores 5-HT4 de Serotonina/metabolismo
4.
Infect Immun ; 88(9)2020 08 19.
Artigo em Inglês | MEDLINE | ID: mdl-32341116

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/imunologia , Infecções por Enterobacteriaceae/imunologia , Microbioma Gastrointestinal/imunologia , Trato Gastrointestinal/imunologia , Neuroimunomodulação/genética , Células Receptoras Sensoriais/imunologia , Animais , Peptídeo Relacionado com Gene de Calcitonina/genética , Peptídeo Relacionado com Gene de Calcitonina/imunologia , Citrobacter/crescimento & desenvolvimento , Citrobacter/imunologia , Sistema Nervoso Entérico/microbiologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Trato Gastrointestinal/inervação , Trato Gastrointestinal/microbiologia , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Moléculas com Motivos Associados a Patógenos/imunologia , Moléculas com Motivos Associados a Patógenos/metabolismo , Células Receptoras Sensoriais/microbiologia , Canal de Cátion TRPA1/genética , Canal de Cátion TRPA1/imunologia , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/imunologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia
5.
Am J Physiol Gastrointest Liver Physiol ; 319(5): G541-G548, 2020 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-32902314

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/crescimento & desenvolvimento , Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/fisiologia , Animais , Sistema Nervoso Entérico/microbiologia , Meio Ambiente , Humanos , Camundongos , Microbiota
6.
Cell Mol Life Sci ; 75(7): 1145-1149, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29285574

RESUMO

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.


Assuntos
Clostridioides difficile/fisiologia , Infecções por Clostridium/microbiologia , Sistema Nervoso Entérico/microbiologia , Síndrome do Intestino Irritável/microbiologia , Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/metabolismo , Interações Hospedeiro-Patógeno , Humanos , Mucosa Intestinal/inervação , Mucosa Intestinal/microbiologia , Modelos Teóricos , Neuroglia/microbiologia , Fatores de Risco
7.
Acta Neuropathol ; 136(3): 345-361, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29797112

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/microbiologia , Microbioma Gastrointestinal/fisiologia , Mucosa Intestinal/microbiologia , Doenças Neurodegenerativas/microbiologia , Animais , Sistema Nervoso Entérico/patologia , Humanos , Mucosa Intestinal/patologia , Doenças Neurodegenerativas/patologia
8.
Gastroenterology ; 151(5): 836-844, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27521479

RESUMO

The gastrointestinal (GI) tract is essential for the absorption of nutrients, induction of mucosal and systemic immune responses, and maintenance of a healthy gut microbiota. Key aspects of gastrointestinal physiology are controlled by the enteric nervous system (ENS), which is composed of neurons and glial cells. The ENS is exposed to and interacts with the outer (microbiota, metabolites, and nutrients) and inner (immune cells and stromal cells) microenvironment of the gut. Although the cellular blueprint of the ENS is mostly in place by birth, the functional maturation of intestinal neural networks is completed within the microenvironment of the postnatal gut, under the influence of gut microbiota and the mucosal immune system. Recent studies have shown the importance of molecular interactions among microbiota, enteric neurons, and immune cells for GI homeostasis. In addition to its role in GI physiology, the ENS has been associated with the pathogenesis of neurodegenerative disorders, such as Parkinson's disease, raising the possibility that microbiota-ENS interactions could offer a viable strategy for influencing the course of brain diseases. Here, we discuss recent advances on the role of microbiota and the immune system on the development and homeostasis of the ENS, a key relay station along the gut-brain axis.


Assuntos
Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/microbiologia , Microbioma Gastrointestinal , Sistema Nervoso Entérico/embriologia , Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Homeostase/fisiologia , Humanos , Mucosa Intestinal/embriologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiologia , Doença de Parkinson/etiologia
9.
Curr Opin Pediatr ; 27(5): 619-24, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26208234

RESUMO

PURPOSE OF REVIEW: Functional gastrointestinal disorders (FGIDs) are some of the most common and challenging disorders in pediatrics. Recurrent abdominal pain is the central feature of pain-associated FGIDs such as irritable bowel syndrome. A thorough understanding of current pathophysiological concepts is essential to successful management. RECENT FINDINGS: The brain-gut axis, role of microbiota and the biopsychosocial model are emerging concepts in FGIDs. The biopsychosocial model focuses on the interplay between genes, environment, and physical and psychosocial factors. Interactions between microbiota and the central, enteric and autonomic nervous systems form the link between gut functions and conscious perceptions. Irritable bowel syndrome is the most extensively studied and prototypical pain-associated FGIDs. An aberrant processing of pain or physiologic signals originating from the gut causes a state of visceral hypersensitivity - a central mechanism of functional pain. Psychosocial and autonomic influences also play large roles. Therapy is tailored to the individual patient and comorbid symptoms. SUMMARY: This review highlights the complex mechanisms and the aberrant brain-gut neural connections forming the basis of FGIDs. Successful management of FGIDs requires knowledge of the underlying pathophysiology coupled with a multidisciplinary treatment approach. Management should focus on cognitive behavioral therapy, dietary factors along with gastrointestinal motility and psychotropic drug therapy.


Assuntos
Dor Abdominal/fisiopatologia , Terapia Cognitivo-Comportamental/tendências , Sistema Nervoso Entérico/fisiopatologia , Gastroenteropatias/fisiopatologia , Trato Gastrointestinal/inervação , Psicotrópicos/uso terapêutico , Dor Abdominal/etiologia , Dor Abdominal/microbiologia , Sistema Nervoso Entérico/microbiologia , Comportamento Alimentar , Gastroenteropatias/microbiologia , Motilidade Gastrointestinal , Trato Gastrointestinal/microbiologia , Humanos
10.
Dig Dis Sci ; 60(5): 1195-205, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25424202

RESUMO

BACKGROUND: Acute gastroenteritis can precipitate irritable bowel syndrome (IBS) in humans. Cytolethal distending toxin is common to all pathogens causing gastroenteritis. Its active subunit, CdtB, is associated with post-infectious bowel changes in a rat model of Campylobacter jejuni infection, including small intestinal bacterial overgrowth (SIBO). AIM: To evaluate the role of host antibodies to CdtB in contributing to post-infectious functional sequelae in this rat model. METHODS: Ileal tissues from non-IBS human subjects, C. jejuni-infected and control rats were immunostained with antibodies to CdtB, c-Kit, S-100, PGP 9.5 and vinculin. Cytosolic and membrane proteins from mouse enteric neuronal cell lysates were immunoprecipitated with anti-CdtB and analyzed by mass spectrometry. ELISAs were performed on rat cardiac serum using CdtB or vinculin as antigens. RESULTS: Anti-CdtB antibodies bound to a cytosolic protein in interstitial cells of Cajal (ICC) and myenteric ganglia in C. jejuni-infected and naïve rats and human subjects. Mass spectrometry identified vinculin, confirmed by co-localization and ELISAs. Anti-CdtB antibodies were higher in C. jejuni-infected rats (1.27 ± 0.15) than controls (1.76 ± 0.12) (P < 0.05), and rats that developed SIBO (2.01 ± 0.18) vs. rats that did not (1.44 ± 0.11) (P = 0.019). Vinculin expression levels were reduced in C. jejuni-infected rats (0.058 ± 0.053) versus controls (0.087 ± 0.023) (P = 0.0001), with greater reductions in rats with two C. jejuni infections (P = 0.0001) and rats that developed SIBO (P = 0.001). CONCLUSIONS: Host anti-CdtB antibodies cross-react with vinculin in ICC and myenteric ganglia, required for normal gut motility. Circulating antibody levels and loss of vinculin expression correlate with number of C. jejuni exposures and SIBO, suggesting that effects on vinculin are important in the effects of C. jejuni infection on the host gut.


Assuntos
Anticorpos Antibacterianos/imunologia , Autoimunidade , Toxinas Bacterianas/imunologia , Infecções por Campylobacter/imunologia , Campylobacter jejuni/imunologia , Enterite/imunologia , Intestino Delgado/imunologia , Vinculina/imunologia , Animais , Infecções por Campylobacter/microbiologia , Infecções por Campylobacter/fisiopatologia , Campylobacter jejuni/patogenicidade , Reações Cruzadas , Modelos Animais de Doenças , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/microbiologia , Enterite/microbiologia , Enterite/fisiopatologia , Gânglios/imunologia , Gânglios/microbiologia , Humanos , Células Intersticiais de Cajal/imunologia , Células Intersticiais de Cajal/microbiologia , Intestino Delgado/inervação , Intestino Delgado/microbiologia , Intestino Delgado/fisiopatologia , Camundongos , Fenótipo , Ratos
11.
Cell Mol Life Sci ; 70(1): 55-69, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22638926

RESUMO

Recent advances in research have greatly increased our understanding of the importance of the gut microbiota. Bacterial colonization of the intestine is critical to the normal development of many aspects of physiology such as the immune and endocrine systems. It is emerging that the influence of the gut microbiota also extends to modulation of host neural development. Furthermore, the overall balance in composition of the microbiota, together with the influence of pivotal species that induce specific responses, can modulate adult neural function, peripherally and centrally. Effects of commensal gut bacteria in adult animals include protection from the central effects of infection and inflammation as well as modulation of normal behavioral responses. There is now robust evidence that gut bacteria influence the enteric nervous system, an effect that may contribute to afferent signaling to the brain. The vagus nerve has also emerged as an important means of communicating signals from gut bacteria to the CNS. Further understanding of the mechanisms underlying microbiome-gut-brain communication will provide us with new insight into the symbiotic relationship between gut microbiota and their mammalian hosts and help us identify the potential for microbial-based therapeutic strategies to aid in the treatment of mood disorders.


Assuntos
Sistema Nervoso Central/microbiologia , Sistema Nervoso Entérico/microbiologia , Intestinos/microbiologia , Modelos Biológicos , Animais , Encéfalo/metabolismo , Encéfalo/fisiologia , Sistema Nervoso Central/crescimento & desenvolvimento , Vida Livre de Germes , Humanos , Inflamação/microbiologia , Metagenoma , Camundongos , Neuropeptídeos/metabolismo , Neuropeptídeos/fisiologia , Transdução de Sinais , Estresse Fisiológico , Nervo Vago/metabolismo , Nervo Vago/fisiologia
12.
Gastroenterology ; 143(4): 1006-16.e4, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22732731

RESUMO

BACKGROUND & AIMS: Altered gastrointestinal motility is associated with significant morbidity and health care costs. Toll-like receptors (TLR) regulate intestinal homeostasis. We examined the roles of TLR4 signaling in survival of enteric neurons and gastrointestinal motility. METHODS: We assessed changes in intestinal motility by assessing stool frequency, bead expulsion, and isometric muscle recordings of colonic longitudinal muscle strips from mice that do not express TLR4 (Tlr4(Lps-d) or TLR4(-/-)) or Myd88 (Myd88(-/-)), in wild-type germ-free mice or wild-type mice depleted of the microbiota, and in mice with neural crest-specific deletion of Myd88 (Wnt1Cre(+/-)/Myd88(fl/fl)). We studied the effects of the TLR4 agonist lipopolysaccharide (LPS) on survival of cultured, immortalized fetal enteric neurons and enteric neuronal cells isolated from wild-type and Tlr4(Lps-d) mice at embryonic day 13.5. RESULTS: There was a significant delay in gastrointestinal motility and reduced numbers of nitrergic neurons in TLR4(Lps-d), TLR4(-/-), and Myd88(-/-) mice compared with wild-type mice. A similar phenotype was observed in germ-free mice, mice depleted of intestinal microbiota, and Wnt1Cre(+/-)/Myd88(fl/fl) mice. Incubation of enteric neuronal cells with LPS led to activation of the transcription factor nuclear factor (NF)-κB and increased cell survival. CONCLUSIONS: Interactions between enteric neurons and microbes increases neuron survival and gastrointestinal motility in mice. LPS activation of TLR4 and NF-κB appears to promote survival of enteric neurons. Factors that regulate TLR4 signaling in neurons might be developed to alter gastrointestinal motility.


Assuntos
Sistema Nervoso Entérico/metabolismo , Motilidade Gastrointestinal , Metagenoma , Fator 88 de Diferenciação Mieloide/metabolismo , Neurônios Nitrérgicos/metabolismo , Receptor 4 Toll-Like/metabolismo , Análise de Variância , Animais , Antibacterianos/farmacologia , Apoptose , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Neurônios Colinérgicos/fisiologia , Colo/fisiologia , Defecação , Ingestão de Alimentos , Endotoxinas/sangue , Sistema Nervoso Entérico/microbiologia , Fezes/microbiologia , Feminino , Motilidade Gastrointestinal/efeitos dos fármacos , Lipopolissacarídeos/sangue , Lipopolissacarídeos/farmacologia , Masculino , Camundongos , Camundongos Knockout , Modelos Animais , Contração Muscular , Músculo Liso/fisiologia , Fator 88 de Diferenciação Mieloide/genética , Neurônios Nitrérgicos/microbiologia , Neurônios Nitrérgicos/fisiologia , Fenótipo , Transdução de Sinais , Receptor 4 Toll-Like/genética
13.
Ageing Res Rev ; 84: 101812, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36455790

RESUMO

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.


Assuntos
Eixo Encéfalo-Intestino , Sistema Nervoso Entérico , Doença de Parkinson , Humanos , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Inflamação/metabolismo , Neuroglia/metabolismo , Neuroglia/patologia , Doença de Parkinson/metabolismo , Doença de Parkinson/patologia , Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/patologia
14.
Trends Microbiol ; 29(8): 686-699, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33309188

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/genética , Regulação Bacteriana da Expressão Gênica , Homeostase , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/microbiologia , Microbioma Gastrointestinal/fisiologia , Humanos , Intestinos/microbiologia , Neurônios/fisiologia
15.
Neuropharmacology ; 197: 108721, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34274348

RESUMO

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'.


Assuntos
Eixo Encéfalo-Intestino , Sistema Nervoso Entérico/fisiopatologia , Microbioma Gastrointestinal , Doenças Neurodegenerativas/microbiologia , Doenças Neurodegenerativas/fisiopatologia , Animais , Sistema Nervoso Entérico/microbiologia , Humanos , Doenças Neurodegenerativas/psicologia
16.
J Clin Invest ; 131(13)2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34196310

RESUMO

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.


Assuntos
Microbioma Gastrointestinal , Esclerose Múltipla/terapia , Animais , Autoimunidade , Modelos Animais de Doenças , Disbiose/imunologia , Disbiose/fisiopatologia , Sistema Endócrino/imunologia , Sistema Endócrino/fisiopatologia , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/fisiopatologia , Transplante de Microbiota Fecal , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/fisiopatologia , Modelos Neurológicos , Esclerose Múltipla/etiologia , Esclerose Múltipla/microbiologia , Neuroimunomodulação , Probióticos/uso terapêutico
17.
Artigo em Inglês | MEDLINE | ID: mdl-30858329

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/fisiologia , Microbioma Gastrointestinal/fisiologia , Intestino Delgado/fisiologia , Animais , Sistema Nervoso Entérico/microbiologia , Homeostase , Humanos , Intestino Delgado/microbiologia
18.
Front Immunol ; 11: 602070, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33552060

RESUMO

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.


Assuntos
Sistema Nervoso Entérico/imunologia , Galanina/imunologia , Trato Gastrointestinal/imunologia , Animais , Brachyspira hyodysenteriae/imunologia , Colite/imunologia , Colite/microbiologia , Colite/patologia , Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/patologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Infecções por Bactérias Gram-Negativas/imunologia , Infecções por Bactérias Gram-Negativas/microbiologia , Infecções por Bactérias Gram-Negativas/patologia , Humanos , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Suínos
19.
Physiol Rep ; 8(21): e14611, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33185323

RESUMO

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.


Assuntos
Bactérias/metabolismo , Sistema Nervoso Entérico/citologia , Sistema Nervoso Entérico/microbiologia , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/microbiologia , Animais , Apoptose , Diferenciação Celular/fisiologia , Células Cultivadas , Sistema Nervoso Entérico/metabolismo , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Gravidez
20.
Front Immunol ; 11: 189, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32256485

RESUMO

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.


Assuntos
Corioamnionite/veterinária , Sistema Nervoso Entérico/lesões , Sistema Nervoso Entérico/microbiologia , Enterocolite Necrosante/veterinária , Feto/microbiologia , Ovinos/embriologia , Infecções por Ureaplasma/complicações , Infecções por Ureaplasma/veterinária , Ureaplasma , Animais , Animais Recém-Nascidos , Corioamnionite/induzido quimicamente , Corioamnionite/microbiologia , Doença Crônica/veterinária , Modelos Animais de Doenças , Sistema Nervoso Entérico/efeitos dos fármacos , Enterocolite Necrosante/induzido quimicamente , Enterocolite Necrosante/microbiologia , Feminino , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/farmacologia , Gravidez , Nascimento Prematuro/veterinária , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo , Ovinos/microbiologia , Ubiquitina Tiolesterase/metabolismo , Infecções por Ureaplasma/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA