Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 118
Filtrar
1.
Rev Esc Enferm USP ; 58: e20230365, 2024.
Artigo em Inglês, Português | MEDLINE | ID: mdl-38743953

RESUMO

OBJECTIVE: To map the evidence in the literature about the relationship between gastrointestinal symptoms and COVID-19 in the pediatric population. METHOD: This is a scoping review following the recommendations of the Joanna Briggs Institute and PRISMA Extension for Scoping Reviews (PRISMA-ScR): Checklist and Explanation. The search was carried out on the following bases: Embase, Google Scholar, PubMed, Scopus, LILACS, CINAHL, Scielo, Web of Science and Virtual Health Library Portal, between July and August 2023. Original studies available in full, in any language, were included. RESULTS: Ten studies were chosen that pointed to three premises: (1) the ACE2 receptor is found in the epithelial cells of the gastrointestinal tract; (2) gastrointestinal symptoms are mediated by stress and infection is justified by the gut-brain axis; (3) it develops the process of Multisystem Inflammatory Syndrome in children, affecting the gastrointestinal tract. CONCLUSION: The synthesis of evidence provided three assumptions which guide the origin of gastrointestinal symptoms. The identification of gastrointestinal symptoms in children affected by COVID-19 can assist in the clinical approach and management of care and treatments.


Assuntos
COVID-19 , Gastroenteropatias , Humanos , COVID-19/complicações , Gastroenteropatias/virologia , Gastroenteropatias/epidemiologia , Criança , Síndrome de Resposta Inflamatória Sistêmica/fisiopatologia , Síndrome de Resposta Inflamatória Sistêmica/diagnóstico , Eixo Encéfalo-Intestino/fisiologia , Enzima de Conversão de Angiotensina 2/metabolismo
2.
J Neuroinflammation ; 21(1): 124, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38730498

RESUMO

Traumatic brain injury (TBI) is a chronic and debilitating disease, associated with a high risk of psychiatric and neurodegenerative diseases. Despite significant advancements in improving outcomes, the lack of effective treatments underscore the urgent need for innovative therapeutic strategies. The brain-gut axis has emerged as a crucial bidirectional pathway connecting the brain and the gastrointestinal (GI) system through an intricate network of neuronal, hormonal, and immunological pathways. Four main pathways are primarily implicated in this crosstalk, including the systemic immune system, autonomic and enteric nervous systems, neuroendocrine system, and microbiome. TBI induces profound changes in the gut, initiating an unrestrained vicious cycle that exacerbates brain injury through the brain-gut axis. Alterations in the gut include mucosal damage associated with the malabsorption of nutrients/electrolytes, disintegration of the intestinal barrier, increased infiltration of systemic immune cells, dysmotility, dysbiosis, enteroendocrine cell (EEC) dysfunction and disruption in the enteric nervous system (ENS) and autonomic nervous system (ANS). Collectively, these changes further contribute to brain neuroinflammation and neurodegeneration via the gut-brain axis. In this review article, we elucidate the roles of various anti-inflammatory pharmacotherapies capable of attenuating the dysregulated inflammatory response along the brain-gut axis in TBI. These agents include hormones such as serotonin, ghrelin, and progesterone, ANS regulators such as beta-blockers, lipid-lowering drugs like statins, and intestinal flora modulators such as probiotics and antibiotics. They attenuate neuroinflammation by targeting distinct inflammatory pathways in both the brain and the gut post-TBI. These therapeutic agents exhibit promising potential in mitigating inflammation along the brain-gut axis and enhancing neurocognitive outcomes for TBI patients.


Assuntos
Anti-Inflamatórios , Lesões Encefálicas Traumáticas , Eixo Encéfalo-Intestino , Humanos , Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Eixo Encéfalo-Intestino/efeitos dos fármacos , Animais , Anti-Inflamatórios/uso terapêutico , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Doenças Neuroinflamatórias/tratamento farmacológico , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/etiologia
3.
Gut Microbes ; 16(1): 2351520, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38717832

RESUMO

Links between the gut microbiota and human health have been supported throughout numerous studies, such as the development of neurological disease disorders. This link is referred to as the "microbiota-gut-brain axis" and is the focus of an emerging field of research. Microbial-derived metabolites and gut and neuro-immunological metabolites regulate this axis in health and many diseases. Indeed, assessing these signals, whether induced by microbial metabolites or neuro-immune mediators, could significantly increase our knowledge of the microbiota-gut-brain axis. However, this will require the development of appropriate techniques and potential models. Methods for studying the induced signals originating from the microbiota remain crucial in this field. This review discusses the methods and techniques available for studies of microbiota-gut-brain interactions. We highlight several much-debated elements of these methodologies, including the widely used in vivo and in vitro models, their implications, and perspectives in the field based on a systematic review of PubMed. Applications of various animal models (zebrafish, mouse, canine, rat, rabbit) to microbiota-gut-brain axis research with practical examples of in vitro methods and innovative approaches to studying gut-brain communications are highlighted. In particular, we extensively discuss the potential of "organ-on-a-chip" devices and their applications in this field. Overall, this review sheds light on the most widely used models and methods, guiding researchers in the rational choice of strategies for studies of microbiota-gut-brain interactions.


Assuntos
Eixo Encéfalo-Intestino , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos , Animais , Microbioma Gastrointestinal/fisiologia , Eixo Encéfalo-Intestino/fisiologia , Humanos , Encéfalo/microbiologia , Encéfalo/metabolismo , Encéfalo/fisiologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/metabolismo , Modelos Animais , Camundongos
4.
Biosens Bioelectron ; 258: 116298, 2024 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-38701537

RESUMO

Wireless activation of the enteric nervous system (ENS) in freely moving animals with implantable optogenetic devices offers a unique and exciting opportunity to selectively control gastrointestinal (GI) transit in vivo, including the gut-brain axis. Programmed delivery of light to targeted locations in the GI-tract, however, poses many challenges not encountered within the central nervous system (CNS). We report here the development of a fully implantable, battery-free wireless device specifically designed for optogenetic control of the GI-tract, capable of generating sufficient light over large areas to robustly activate the ENS, potently inducing colonic motility ex vivo and increased propulsion in vivo. Use in in vivo studies reveals unique stimulation patterns that increase expulsion of colonic content, likely mediated in part by activation of an extrinsic brain-gut motor pathway, via pelvic nerves. This technology overcomes major limitations of conventional wireless optogenetic hardware designed for the CNS, providing targeted control of specific neurochemical classes of neurons in the ENS and brain-gut axis, for direct modulation of GI-transit and associated behaviours in freely moving animals.


Assuntos
Sistema Nervoso Entérico , Optogenética , Tecnologia sem Fio , Animais , Optogenética/instrumentação , Sistema Nervoso Entérico/fisiologia , Camundongos , Tecnologia sem Fio/instrumentação , Eixo Encéfalo-Intestino/fisiologia , Técnicas Biossensoriais/instrumentação , Desenho de Equipamento , Encéfalo/fisiologia , Camundongos Endogâmicos C57BL
5.
Biol Res ; 57(1): 23, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38705984

RESUMO

Obesity, associated with the intake of a high-fat diet (HFD), and anxiety are common among those living in modern urban societies. Recent studies suggest a role of microbiome-gut-brain axis signaling, including a role for brain serotonergic systems in the relationship between HFD and anxiety. Evidence suggests the gut microbiome and the serotonergic brain system together may play an important role in this response. Here we conducted a nine-week HFD protocol in male rats, followed by an analysis of the gut microbiome diversity and community composition, brainstem serotonergic gene expression (tph2, htr1a, and slc6a4), and anxiety-related defensive behavioral responses. We show that HFD intake decreased alpha diversity and altered the community composition of the gut microbiome in association with obesity, increased brainstem tph2, htr1a and slc6a4 mRNA expression, including in the caudal part of the dorsomedial dorsal raphe nucleus (cDRD), a subregion previously associated with stress- and anxiety-related behavioral responses, and, finally, increased anxiety-related defensive behavioral responses. The HFD increased the Firmicutes/Bacteroidetes ratio relative to control diet, as well as higher relative abundances of Blautia, and decreases in Prevotella. We found that tph2, htr1a and slc6a4 mRNA expression were increased in subregions of the dorsal raphe nucleus in the HFD, relative to control diet. Specific bacterial taxa were associated with increased serotonergic gene expression in the cDRD. Thus, we propose that HFD-induced obesity is associated with altered microbiome-gut-serotonergic brain axis signaling, leading to increased anxiety-related defensive behavioral responses in rats.


Assuntos
Ansiedade , Eixo Encéfalo-Intestino , Dieta Hiperlipídica , Microbioma Gastrointestinal , Animais , Masculino , Dieta Hiperlipídica/efeitos adversos , Microbioma Gastrointestinal/fisiologia , Ansiedade/microbiologia , Eixo Encéfalo-Intestino/fisiologia , Ratos , Ratos Sprague-Dawley , Obesidade/microbiologia , Obesidade/psicologia , Obesidade/metabolismo , Transdução de Sinais/fisiologia , Comportamento Animal/fisiologia
6.
Cells ; 13(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38727306

RESUMO

Parkinson's disease (PD) is recognized as the second most prevalent primary chronic neurodegenerative disorder of the central nervous system. Clinically, PD is characterized as a movement disorder, exhibiting an incidence and mortality rate that is increasing faster than any other neurological condition. In recent years, there has been a growing interest concerning the role of the gut microbiota in the etiology and pathophysiology of PD. The establishment of a brain-gut microbiota axis is now real, with evidence denoting a bidirectional communication between the brain and the gut microbiota through metabolic, immune, neuronal, and endocrine mechanisms and pathways. Among these, the vagus nerve represents the most direct form of communication between the brain and the gut. Given the potential interactions between bacteria and drugs, it has been observed that the therapies for PD can have an impact on the composition of the microbiota. Therefore, in the scope of the present review, we will discuss the current understanding of gut microbiota on PD and whether this may be a new paradigm for treating this devastating disease.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Microbioma Gastrointestinal , Doença de Parkinson , Humanos , Doença de Parkinson/microbiologia , Doença de Parkinson/terapia , Encéfalo/microbiologia , Encéfalo/patologia , Eixo Encéfalo-Intestino/fisiologia , Animais
7.
Medicine (Baltimore) ; 103(17): e37960, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38669388

RESUMO

Short-chain fatty acids (SCFAs) are, the metabolic byproducts of intestinal microbiota that, are generated through anaerobic fermentation of undigested dietary fibers. SCFAs play a pivotal role in numerous physiological functions within the human body, including maintaining intestinal mucosal health, modulating immune functions, and regulating energy metabolism. In recent years, extensive research evidence has indicated that SCFAs are significantly involved in the onset and progression of Parkinson disease (PD). However, the precise mechanisms remain elusive. This review comprehensively summarizes the progress in understanding how SCFAs impact PD pathogenesis and the underlying mechanisms. Primarily, we delve into the synthesis, metabolism, and signal transduction of SCFAs within the human body. Subsequently, an analysis of SCFA levels in patients with PD is presented. Furthermore, we expound upon the mechanisms through which SCFAs induce inflammatory responses, oxidative stress, abnormal aggregation of alpha-synuclein, and the intricacies of the gut-brain axis. Finally, we provide a critical analysis and explore the potential therapeutic role of SCFAs as promising targets for treating PD.


Assuntos
Ácidos Graxos Voláteis , Microbioma Gastrointestinal , Doença de Parkinson , Humanos , Doença de Parkinson/metabolismo , Doença de Parkinson/microbiologia , Microbioma Gastrointestinal/fisiologia , Ácidos Graxos Voláteis/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Estresse Oxidativo/fisiologia
8.
Life Sci ; 347: 122650, 2024 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-38631669

RESUMO

AIMS: As a unique iron-dependent non-apoptotic cell death, Ferroptosis is involved in the pathogenesis and development of many human diseases and has become a research hotspot in recent years. However, the regulatory role of ferroptosis in the gut-liver-brain axis has not been elucidated. This paper summarizes the regulatory role of ferroptosis and provides theoretical basis for related research. MATERIALS AND METHODS: We searched PubMed, CNKI and Wed of Science databases on ferroptosis mediated gut-liver-brain axis diseases, summarized the regulatory role of ferroptosis on organ axis, and explained the adverse effects of related regulatory effects on various diseases. KEY FINDINGS: According to our summary, the main way in which ferroptosis mediates the gut-liver-brain axis is oxidative stress, and the key cross-talk of ferroptosis affecting signaling pathway network is Nrf2/HO-1. However, there were no specific marker between different organ axes mediate by ferroptosis. SIGNIFICANCE: Our study illustrates the main ways and key cross-talk of ferroptosis mediating the gut-liver-brain axis, providing a basis for future research.


Assuntos
Encéfalo , Ferroptose , Fígado , Estresse Oxidativo , Ferroptose/fisiologia , Humanos , Estresse Oxidativo/fisiologia , Encéfalo/metabolismo , Fígado/metabolismo , Fígado/patologia , Animais , Eixo Encéfalo-Intestino/fisiologia , Transdução de Sinais , Fator 2 Relacionado a NF-E2/metabolismo
9.
Neurosci Biobehav Rev ; 161: 105653, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38582194

RESUMO

The evolution of the gut-microbiota-brain axis in animals reveals that microbial inputs influence metabolism, the regulation of inflammation and the development of organs, including the brain. Inflammatory, neurodegenerative and psychiatric disorders are more prevalent in people of low socioeconomic status (SES). Many aspects of low SES reduce exposure to the microbial inputs on which we are in a state of evolved dependence, whereas the lifestyle of wealthy citizens maintains these exposures. This partially explains the health deficit of low SES, so focussing on our evolutionary history and on environmental and lifestyle factors that distort microbial exposures might help to mitigate that deficit. But the human microbiota is complex and we have poor understanding of its functions at the microbial and mechanistic levels, and in the brain. Perhaps its composition is more flexible than the microbiota of animals that have restricted habitats and less diverse diets? These uncertainties are discussed in relation to the encouraging but frustrating results of attempts to treat psychiatric disorders by modulating the microbiota.


Assuntos
Evolução Biológica , Microbioma Gastrointestinal , Classe Social , Humanos , Microbioma Gastrointestinal/fisiologia , Animais , Eixo Encéfalo-Intestino/fisiologia , Transtornos Mentais/microbiologia , Saúde Mental , Baixo Nível Socioeconômico
10.
J Neurosci ; 44(20)2024 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-38594069

RESUMO

The brain bidirectionally communicates with the gut to control food intake and energy balance, which becomes dysregulated in obesity. For example, endocannabinoid (eCB) signaling in the small-intestinal (SI) epithelium is upregulated in diet-induced obese (DIO) mice and promotes overeating by a mechanism that includes inhibiting gut-brain satiation signaling. Upstream neural and molecular mechanism(s) involved in overproduction of orexigenic gut eCBs in DIO, however, are unknown. We tested the hypothesis that overactive parasympathetic signaling at the muscarinic acetylcholine receptors (mAChRs) in the SI increases biosynthesis of the eCB, 2-arachidonoyl-sn-glycerol (2-AG), which drives hyperphagia via local CB1Rs in DIO. Male mice were maintained on a high-fat/high-sucrose Western-style diet for 60 d, then administered several mAChR antagonists 30 min prior to tissue harvest or a food intake test. Levels of 2-AG and the activity of its metabolic enzymes in the SI were quantitated. DIO mice, when compared to those fed a low-fat/no-sucrose diet, displayed increased expression of cFos protein in the dorsal motor nucleus of the vagus, which suggests an increased activity of efferent cholinergic neurotransmission. These mice exhibited elevated levels of 2-AG biosynthesis in the SI, that was reduced to control levels by mAChR antagonists. Moreover, the peripherally restricted mAChR antagonist, methylhomatropine bromide, and the peripherally restricted CB1R antagonist, AM6545, reduced food intake in DIO mice for up to 24 h but had no effect in mice conditionally deficient in SI CB1Rs. These results suggest that hyperactivity at mAChRs in the periphery increases formation of 2-AG in the SI and activates local CB1Rs, which drives hyperphagia in DIO.


Assuntos
Dieta Hiperlipídica , Endocanabinoides , Glicerídeos , Camundongos Endogâmicos C57BL , Obesidade , Transdução de Sinais , Transmissão Sináptica , Animais , Endocanabinoides/metabolismo , Masculino , Obesidade/metabolismo , Camundongos , Transmissão Sináptica/fisiologia , Transmissão Sináptica/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Transdução de Sinais/fisiologia , Glicerídeos/metabolismo , Ácidos Araquidônicos/metabolismo , Ingestão de Alimentos/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Antagonistas Muscarínicos/farmacologia , Receptores Muscarínicos/metabolismo , Eixo Encéfalo-Intestino/fisiologia
11.
Front Cell Infect Microbiol ; 14: 1334581, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38644963

RESUMO

Ischemic stroke (IS) is a serious central nervous system disease. Post-IS complications, such as post-stroke cognitive impairment (PSCI), post-stroke depression (PSD), hemorrhagic transformation (HT), gastrointestinal dysfunction, cardiovascular events, and post-stroke infection (PSI), result in neurological deficits. The microbiota-gut-brain axis (MGBA) facilitates bidirectional signal transduction and communication between the intestines and the brain. Recent studies have reported alterations in gut microbiota diversity post-IS, suggesting the involvement of gut microbiota in post-IS complications through various mechanisms such as bacterial translocation, immune regulation, and production of gut bacterial metabolites, thereby affecting disease prognosis. In this review, to provide insights into the prevention and treatment of post-IS complications and improvement of the long-term prognosis of IS, we summarize the interaction between the gut microbiota and IS, along with the effects of the gut microbiota on post-IS complications.


Assuntos
Eixo Encéfalo-Intestino , Microbioma Gastrointestinal , AVC Isquêmico , Humanos , AVC Isquêmico/complicações , AVC Isquêmico/microbiologia , Eixo Encéfalo-Intestino/fisiologia , Animais , Disbiose , Encéfalo/microbiologia , Translocação Bacteriana , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/etiologia
13.
Neurobiol Dis ; 195: 106499, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38588753

RESUMO

The gut-brain axis is a bidirectional communication network linking the gut and the brain, overseeing digestive functions, emotional responses, body immunity, brain development, and overall health. Substantial research highlights a connection between disruptions of the gut-brain axis and various psychiatric and neurological conditions, including depression and Alzheimer's disease. Given the impact of the gut-brain axis on behavior, cognition, and brain diseases, some studies have started to pay attention to the role of the axis in sepsis-associated encephalopathy (SAE), where cognitive impairment is the primary manifestation. SAE emerges as the primary and earliest form of organ dysfunction following sepsis, potentially leading to acute cognitive impairment and long-term cognitive decline in patients. Notably, the neuronal damage in SAE does not stem directly from the central nervous system (CNS) infection but rather from an infection occurring outside the brain. The gut-brain axis is posited as a pivotal factor in this process. This review will delve into the gut-brain axis, exploring four crucial pathways through which inflammatory signals are transmitted and elevate the incidence of SAE. These pathways encompass the vagus nerve pathway, the neuroendocrine pathway involving the hypothalamic-pituitary-adrenal (HPA) axis and serotonin (5-HT) regulation, the neuroimmune pathway, and the microbial regulation. These pathways can operate independently or collaboratively on the CNS to modulate brain activity. Understanding how the gut affects and regulates the CNS could offer the potential to identify novel targets for preventing and treating this condition, ultimately enhancing the prognosis for individuals with SAE.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Encefalopatia Associada a Sepse , Humanos , Eixo Encéfalo-Intestino/fisiologia , Encefalopatia Associada a Sepse/fisiopatologia , Encefalopatia Associada a Sepse/metabolismo , Animais , Encéfalo/fisiopatologia , Encéfalo/metabolismo , Microbioma Gastrointestinal/fisiologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/fisiopatologia , Sistema Hipófise-Suprarrenal/metabolismo , Sepse/fisiopatologia , Sepse/complicações
14.
Clin Nutr ; 43(5): 1181-1189, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38608404

RESUMO

Malnutrition affects 195 million children under the age of five worldwide with long term effects that include impaired cognitive development. Brain development occurs rapidly over the first 36 months of life. Whilst seemingly independent, changes to the brain and gut microbiome are linked by metabolites, hormones, and neurotransmitters as part of the gut-brain axis. In the context of severe malnutrition, the composition of the gut microbiome and the repertoire of biochemicals exchanged via the gut-brain axis vary when compared to healthy individuals. These effects are primarily due to the recognized interacting determinants, macro- and micronutrient deficiencies, infection, infestations and toxins related to poor sanitation, and a dearth of psycho-social stimulation. The standard of care for the treatment of severe acute malnutrition is focused on nutritional repletion and weight restoration through the provision of macro- and micronutrients, the latter usually in excess of recommended dietary allowances (RDA). However, existing formulations and supplements have not been designed to specifically address key recovery requirements for brain and gut microbiome development. Animal model studies indicate that treatments targeting the gut microbiome could improve brain development. Despite this, research on humans targeting the gut microbiome with the aim of restoring brain functionality are scarce. We conclude that there is a need for assessment of cognition and the use of various tools that permit visualization of the brain anatomy and function (e.g., Magnetic resonance imaging (MRI), functional near-infrared spectroscopy (fNIRS), electroencephalogram (EEG)) to understand how interventions targeting the gut microbiome impact brain development.


Assuntos
Cognição , Microbioma Gastrointestinal , Microbioma Gastrointestinal/fisiologia , Humanos , Lactente , Cognição/fisiologia , Desenvolvimento Infantil/fisiologia , Eixo Encéfalo-Intestino/fisiologia , Encéfalo/crescimento & desenvolvimento , Animais , Desnutrição/fisiopatologia , Desnutrição/microbiologia
15.
BMC Microbiol ; 24(1): 147, 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678197

RESUMO

OBJECTIVE: The purpose of this study is to investigate the connection of pre-competition anxiety with gut microbiota and metabolites in wrestlers with different sports performances. METHODS: One week prior to a national competition, 12 wrestlers completed anxiety questionnaires. Faecal and urine samples were collected for the analysis of gut microbiota and metabolites through the high-throughput sequencing of the 16 S rRNA gene in conjunction with untargeted metabolomics technology. The subjects were divided into two groups, namely, achievement (CP) and no-achievement (CnP) wrestlers, on the basis of whether or not their performances placed them in the top 16 at the competition. The relationship amongst the variations in gut microbiota, metabolites, and anxiety indicators was analyzed. RESULTS: (1) The CP group exhibited significantly higher levels of "state self-confidence," "self-confidence," and "somatic state anxiety" than the CnP group. Conversely, the CP group displayed lower levels of "individual failure anxiety" and "sports competition anxiety" than the CnP group. (2) The gut microbiota in the CP group was more diverse and abundant than that in the CnP group. Pre-competition anxiety was linked to Oscillospiraceae UCG_005, Paraprevotella, Ruminococcaceae and TM7x. (3) The functions of differential metabolites in faeces and urine of the CP/CnP group were mainly enriched in caffeine metabolism, lipopolysaccharide biosynthesis and VEGF and mTOR signaling pathways. Common differential metabolites in feces and urine were significantly associated with multiple anxiety indicators. CONCLUSIONS: Wrestlers with different sports performance have different pre-competition anxiety states, gut microbiota distribution and abundance and differential metabolites in faeces and urine. A certain correlation exists between these psychological and physiological indicators.


Assuntos
Ansiedade , Eixo Encéfalo-Intestino , Fezes , Microbioma Gastrointestinal , Luta Romana , Microbioma Gastrointestinal/fisiologia , Humanos , Ansiedade/microbiologia , Masculino , Fezes/microbiologia , Adulto Jovem , Eixo Encéfalo-Intestino/fisiologia , RNA Ribossômico 16S/genética , Bactérias/classificação , Bactérias/genética , Bactérias/metabolismo , Bactérias/isolamento & purificação , Adolescente , Metabolômica/métodos , Desempenho Atlético/fisiologia , Adulto
16.
Medicina (Kaunas) ; 60(4)2024 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-38674247

RESUMO

Mental health disorders and dementia have become a serious public health concern, with a heightened frequency of diagnoses observed in the wake of the global COVID-19 pandemic. Psychobiotics, a novel area of research at the intersection of microbiology and neuroscience, explore the potential of probiotics to influence the nervous system and mental health outcomes. This review explores the intricate mechanisms by which psychobiotics interact with the gut-brain axis, shedding light on their effects on mood, cognition, and the stress response. Through a comprehensive analysis of the current literature and recent advancements, we discuss the therapeutic potential of psychobiotics in various mental health disorders, including depression, anxiety, and neurodegenerative diseases like dementia. The findings from this research highlight the promising potential of psychobiotics as innovative interventions in mental health treatment. Further investigation into their mechanisms of action and clinical applications is warranted to fully realize their therapeutic benefits.


Assuntos
Eixo Encéfalo-Intestino , COVID-19 , Probióticos , Humanos , Probióticos/uso terapêutico , Probióticos/farmacologia , Eixo Encéfalo-Intestino/fisiologia , Transtornos Mentais/terapia , Microbioma Gastrointestinal/fisiologia , SARS-CoV-2 , Demência
17.
Int Immunopharmacol ; 132: 112030, 2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38603861

RESUMO

Mast cells (MCs) play a significant role in various diseases, and their activation and degranulation can trigger inflammatory responses and barrier damage. Several studies have indicated that vagus nerve stimulation (VNS) exerts ameliorates neurological injury, and regulates gut MC degranulation. However, there is limited research on the modulatory effect of VNS on MCs in both the gut and brain in brain ischemia-reperfusion (I/R) injury in this process. We aim to develop a minimally invasive, targeted and convenient VNS approach to assess the impact of VNS and to clarify the relationship between VNS and MCs on the prognosis of acute ischemic stroke. We utilized middle cerebral artery occlusion/reperfusion (MCAO/r) to induce brain I/R injury. After the experiment, the motor function and neurofunctional impairments of the rats were detected, and the gastrointestinal function, blood-brain barrier (BBB) and intestinal barrier damage, and systemic and local inflammation were evaluated by Nissl, TTC staining, Evans blue, immunofluorescence staining, transmission electron microscopy, western blot assays, ELISA, and fecal 16S rRNA sequencing methods. Our research confirmed that our minimally invasive VNS method is a novel approach for stimulating the vagus nerve. VNS alleviated motor deficits and gastrointestinal dysfunction while also suppressing intestinal and neuroinflammation. Additionally, VNS ameliorated gut microbiota dysbiosis in rats. Furthermore, our analysis indicated that VNS reduces chymase secretion by modulating MCs degranulation and improves intestinal and BBB damage. Our results showed that VNS treatment can alleviate the damage of BBB and colonic barrier after cerebral I/R by modulating mast cell degranulation, and alleviates systemic inflammatory responses.


Assuntos
Barreira Hematoencefálica , Eixo Encéfalo-Intestino , Degranulação Celular , Microbioma Gastrointestinal , AVC Isquêmico , Mastócitos , Ratos Sprague-Dawley , Traumatismo por Reperfusão , Estimulação do Nervo Vago , Animais , Mastócitos/imunologia , Estimulação do Nervo Vago/métodos , Masculino , Ratos , Traumatismo por Reperfusão/terapia , Traumatismo por Reperfusão/imunologia , AVC Isquêmico/terapia , Eixo Encéfalo-Intestino/fisiologia , Infarto da Artéria Cerebral Média/terapia , Modelos Animais de Doenças , Isquemia Encefálica/terapia , Isquemia Encefálica/imunologia
18.
Chemosphere ; 356: 141971, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38604519

RESUMO

The environmental prevalence of antibiotic residues poses a potential threat to gut health and may thereby disrupt brain function through the microbiota-gut-brain axis. However, little is currently known about the impacts of antibiotics on gut health and neurotransmitters along the microbiota-gut-brain axis in fish species. Taking enrofloxacin (ENR) as a representative, the impacts of antibiotic exposure on the gut structural integrity, intestinal microenvironment, and neurotransmitters along the microbiota-gut-brain axis were evaluated in zebrafish in this study. Data obtained demonstrated that exposure of zebrafish to 28-day environmentally realistic levels of ENR (6 and 60 µg/L) generally resulted in marked elevation of two intestinal integrity biomarkers (diamine oxidase (DAO) and malondialdehyde (MDA), upregulation of genes that encode inter-epithelial tight junction proteins, and histological alterations in gut as well as increase of lipopolysaccharide (LPS) in plasma, indicating an evident impairment of the structural integrity of gut. Moreover, in addition to significantly altered neurotransmitters, markedly higher levels of LPS while less amount of two short-chain fatty acids (SCFAs), namely acetic acid and valeric acid, were detected in the gut of ENR-exposed zebrafish, suggesting a disruption of gut microenvironment upon ENR exposure. Along with corresponding changes detected in gut, significant disruption of neurotransmitters in brain indicated by marked alterations in the contents of neurotransmitters, the activity of acetylcholin esterase (AChE), and the expression of neurotransmitter-related genes were also observed. These findings suggest exposure to environmental antibiotic residues may impair gut health and disrupt neurotransmitters along the microbiota-gut-brain axis in zebrafish. Considering the prevalence of antibiotic residues in environments and the high homology of zebrafish to other vertebrates including human, the risk of antibiotic exposure to the health of wild animals as well as human deserves more attention.


Assuntos
Antibacterianos , Enrofloxacina , Microbioma Gastrointestinal , Neurotransmissores , Peixe-Zebra , Animais , Neurotransmissores/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Enrofloxacina/toxicidade , Antibacterianos/toxicidade , Antibacterianos/farmacologia , Eixo Encéfalo-Intestino/efeitos dos fármacos , Eixo Encéfalo-Intestino/fisiologia , Poluentes Químicos da Água/toxicidade , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Malondialdeído/metabolismo , Lipopolissacarídeos
19.
Neurogastroenterol Motil ; 36(5): e14779, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38488234

RESUMO

BACKGROUND: Gastric motility disorder is an increasingly common problem among people with diabetes. Neurotransmitters have been recognized as critical regulators in the process of gastric motility. Previous study has shown that herb pair huanglian-banxia (HL-BX) can improve gastric motility, but the underlying mechanism is still unclear. The aim of this study was to further investigate the role of HL-BX in modulating brain-gut neurotransmission to promote gastric motility in diabetic rats, and to explore its possible mechanism. METHODS: The diabetic rats were divided into five groups. Gastric emptying rate, intestinal propulsion rate, body weight, and average food intake were determined. Substance P (SP), 5- hydroxytryptamine (5-HT), and glucagon-like peptide -1 (GLP-1) in the serum were measured by enzyme-linked immunosorbent assay. Dopamine (DA) and norepinephrine (NE) in the brain were analyzed by high-pressure liquid chromatography with a fluorescence detector. Protein expression of the tissues in the stomach and brain was determined by Western blot. KEY RESULTS: HL-BX reduced average food intake significantly, increased body weight, and improved gastric emptying rate and intestinal propulsion rate. HL-BX administration caused a significant increase in SP, GLP-1, and 5-HT, but a significant decrease in DA and NE. Interestingly, HL-BX regulated simultaneously the different expressions of MAPK and its downstream p70S6K/S6 signaling pathway in the stomach and brain. Moreover, berberine exhibited a similar effect to HL-BX. CONCLUSIONS: These results indicated that HL-BX promoted gastric motility by regulating brain-gut neurotransmitters through the MAPK signaling pathway. HL-BX and MAPK provide a potential therapeutic option for the treatment of gastroparesis.


Assuntos
Diabetes Mellitus Experimental , Medicamentos de Ervas Chinesas , Motilidade Gastrointestinal , Sistema de Sinalização das MAP Quinases , Animais , Masculino , Ratos , Encéfalo/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Diabetes Mellitus Experimental/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Motilidade Gastrointestinal/fisiologia , Motilidade Gastrointestinal/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Neurotransmissores/metabolismo
20.
J Microbiol Biotechnol ; 34(4): 747-756, 2024 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-38321650

RESUMO

Chronic gut inflammation promotes the development of metabolic diseases such as obesity. There is growing evidence which suggests that dysbiosis in gut microbiota and metabolites disrupt the integrity of the intestinal barrier and significantly impact the level of inflammation in various tissues, including the liver and adipose tissues. Moreover, dietary sources are connected to the development of leaky gut syndrome through their interaction with the gut microbiota. This review examines the effects of these factors on intestinal microorganisms and the communication pathways between the gut-liver and gut-brain axis. The consumption of diets rich in fats and carbohydrates has been found to weaken the adherence of tight junction proteins in the gastrointestinal tract. Consequently, this allows endotoxins, such as lipopolysaccharides produced by detrimental bacteria, to permeate through portal veins, leading to metabolic endotoxemia and alterations in the gut microbiome composition with reduced production of metabolites, such as short-chain fatty acids. However, the precise correlation between gut microbiota and alternative sweeteners remains uncertain, necessitating further investigation. This study highlights the significance of exploring the impact of diet on gut microbiota and the underlying mechanisms in the gut-liver and gut-brain axis. Nevertheless, limited research on the gut-liver axis poses challenges in comprehending the intricate connections between diet and the gut-brain axis. This underscores the need for comprehensive studies to elucidate the intricate gut-brain mechanisms underlying intestinal health and microbiota.


Assuntos
Dieta , Disbiose , Microbioma Gastrointestinal , Humanos , Bactérias/classificação , Bactérias/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Dieta/efeitos adversos , Disbiose/microbiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Inflamação , Fígado/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA