Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int J Mol Sci ; 25(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39337263

RESUMO

Changes in maternal gut microbiota due to stress and/or ethanol exposure can have lasting effects on offspring's health, particularly regarding immunity, inflammation response, and susceptibility to psychiatric disorders. The literature search for this review was conducted using PubMed and Scopus, employing keywords and phrases related to maternal stress, ethanol exposure, gut microbiota, microbiome, gut-brain axis, diet, dysbiosis, progesterone, placenta, prenatal development, immunity, inflammation, and depression to identify relevant studies in both preclinical and human research. Only a limited number of reviews were included to support the arguments. The search encompassed studies from the 1990s to the present. This review begins by exploring the role of microbiota in modulating host health and disease. It then examines how disturbances in maternal microbiota can affect the offspring's immune system. The analysis continues by investigating the interplay between stress and dysbiosis, focusing on how prenatal maternal stress influences both maternal and offspring microbiota and its implications for susceptibility to depression. The review also considers the impact of ethanol consumption on gut dysbiosis, with an emphasis on the effects of prenatal ethanol exposure on both maternal and offspring microbiota. Finally, it is suggested that maternal gut microbiota dysbiosis may be significantly exacerbated by the combined effects of stress and ethanol exposure, leading to immune system dysfunction and chronic inflammation, which could increase the risk of depression in the offspring. These interactions underscore the potential for novel mental health interventions that address the gut-brain axis, especially in relation to maternal and offspring health.


Assuntos
Disbiose , Etanol , Microbioma Gastrointestinal , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Humanos , Gravidez , Feminino , Efeitos Tardios da Exposição Pré-Natal/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/imunologia , Etanol/efeitos adversos , Animais , Estresse Psicológico/imunologia , Estresse Psicológico/complicações , Disbiose/imunologia , Transtornos Mentais/etiologia , Transtornos Mentais/imunologia , Transtornos Mentais/microbiologia , Eixo Encéfalo-Intestino
2.
Nutrients ; 16(18)2024 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-39339649

RESUMO

Stress, unhealthy lifestyle, and sleep disturbance worsen cognitive function in mood disorders, prompting a rise in the development of integrative health approaches. The recent investigations in the gut-brain axis field highlight the strong interplay among microbiota, inflammation, and mental health. Thus, this study aimed to investigate a new nutraceutical formulation comprising prebiotics, minerals, and silymarin's impact on microbiota, inflammation, mood, and sleep quality. The study evaluated the LL1 + silymarin capsule supplementation over 180 days in overweight adults. We analyzed the fecal gut microbiota using partial 16S rRNA sequences, measured cytokine expression via CBA, collected anthropometric data, quality of life, and sleep questionnaire responses, and obtained plasma samples for metabolic and hormonal analysis at baseline (T0) and 180 days (T180) post-supplementation. Our findings revealed significant reshaping in gut microbiota composition at the phylum, genus, and species levels, especially in the butyrate-producer bacteria post-supplementation. These changes in gut microbiota were linked to enhancements in sleep quality, mood perception, cytokine expression, and anthropometric measures which microbiota-derived short-chain fatty acids might enhance. The supplementation tested in this study seems to be able to improve microbiota composition, reflecting anthropometrics and inflammation, as well as sleep quality and mood improvement.


Assuntos
Afeto , Eixo Encéfalo-Intestino , Suplementos Nutricionais , Microbioma Gastrointestinal , Silimarina , Qualidade do Sono , Humanos , Microbioma Gastrointestinal/efeitos dos fármacos , Projetos Piloto , Afeto/efeitos dos fármacos , Masculino , Feminino , Silimarina/farmacologia , Adulto , Eixo Encéfalo-Intestino/efeitos dos fármacos , Pessoa de Meia-Idade , Qualidade de Vida , Fezes/microbiologia , Cápsulas , Citocinas/metabolismo , Citocinas/sangue , Sobrepeso , Prebióticos/administração & dosagem , RNA Ribossômico 16S
3.
Life Sci ; 354: 122979, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39147315

RESUMO

Stroke is the second most common cause of death and one of the most common causes of disability worldwide. The intestine is home to several microorganisms that fulfill essential functions for the natural and physiological functioning of the human body. There is an interaction between the central nervous system (CNS) and the gastrointestinal system that enables bidirectional communication between them, the so-called gut-brain axis. Based on the gut-brain axis, there is evidence of a link between the gut microbiota and the regulation of microglial functions through glial activation. This interaction is partly due to the immunological properties of the microbiota and its connection with the CNS, such that metabolites produced by the microbiota can cross the gut barrier, enter the bloodstream and reach the CNS and significantly affect microglia, astrocytes and other cells of the immune system. Studies addressing the effects of short-chain fatty acids (SCFAs) on glial function and the BBB in ischemic stroke are still scarce. Therefore, this review aims to stimulate the investigation of these associations, as well as to generate new studies on this topic that can clarify the role of SCFAs after stroke in a more robust manner.


Assuntos
Barreira Hematoencefálica , Ácidos Graxos Voláteis , Microbioma Gastrointestinal , AVC Isquêmico , Neuroglia , Humanos , Barreira Hematoencefálica/metabolismo , Ácidos Graxos Voláteis/metabolismo , Ácidos Graxos Voláteis/farmacologia , AVC Isquêmico/metabolismo , AVC Isquêmico/fisiopatologia , Animais , Neuroglia/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Isquemia Encefálica/metabolismo
4.
J Neuroimmune Pharmacol ; 19(1): 45, 2024 Aug 19.
Artigo em Inglês | MEDLINE | ID: mdl-39158758

RESUMO

Multiple Sclerosis (MS) is a debilitating disease that severely affects the central nervous system (CNS). Apart from neurological symptoms, it is also characterized by neuropsychiatric comorbidities, such as anxiety and depression. Phosphodiesterase-5 inhibitors (PDE5Is) such as Sildenafil and Tadalafil have been shown to possess antidepressant-like effects, but the mechanisms underpinning such effects are not fully characterized. To address this question, we used the EAE model of MS, behavioral tests, immunofluorescence, immunohistochemistry, western blot, and 16 S rRNA sequencing. Here, we showed that depressive-like behavior in Experimental Autoimmune Encephalomyelitis (EAE) mice is due to neuroinflammation, reduced synaptic plasticity, dysfunction in glutamatergic neurotransmission, glucocorticoid receptor (GR) resistance, increased blood-brain barrier (BBB) permeability, and immune cell infiltration to the CNS, as well as inflammation, increased intestinal permeability, and immune cell infiltration in the distal colon. Furthermore, 16 S rRNA sequencing revealed that behavioral dysfunction in EAE mice is associated with changes in the gut microbiota, such as an increased abundance of Firmicutes and Saccharibacteria and a reduction in Proteobacteria, Parabacteroides, and Desulfovibrio. Moreover, we detected an increased abundance of Erysipelotrichaceae and Desulfovibrionaceae and a reduced abundance of Lactobacillus johnsonii. Surprisingly, we showed that Tadalafil likely exerts antidepressant-like effects by targeting all aforementioned disease aspects. In conclusion, our work demonstrated that anxiety- and depressive-like behavior in EAE is associated with a plethora of neuroimmune and gut microbiota-mediated mechanisms and that Tadalafil exerts antidepressant-like effects probably by targeting these mechanisms. Harnessing the knowledge of these mechanisms of action of Tadalafil is important to pave the way for future clinical trials with depressed patients.


Assuntos
Ansiolíticos , Antidepressivos , Eixo Encéfalo-Intestino , Depressão , Encefalomielite Autoimune Experimental , Inibidores da Fosfodiesterase 5 , Tadalafila , Animais , Feminino , Camundongos , Ansiolíticos/administração & dosagem , Antidepressivos/administração & dosagem , Autoimunidade/efeitos dos fármacos , Eixo Encéfalo-Intestino/efeitos dos fármacos , Depressão/tratamento farmacológico , Encefalomielite Autoimune Experimental/tratamento farmacológico , Encefalomielite Autoimune Experimental/imunologia , Microbioma Gastrointestinal/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Inibidores da Fosfodiesterase 5/administração & dosagem , Tadalafila/administração & dosagem
5.
J Neuroimmune Pharmacol ; 19(1): 36, 2024 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-39042202

RESUMO

Newly conducted research suggests that metabolic disorders, like diabetes and obesity, play a significant role as risk factors for psychiatric disorders. This connection presents a potential avenue for creating novel antidepressant medications by repurposing drugs originally developed to address antidiabetic conditions. Earlier investigations have shown that GLP-1 (Glucagon-like Peptide-1) analogs exhibit neuroprotective qualities in various models of neurological diseases, encompassing conditions such as Alzheimer's disease, Parkinson's disease, and stroke. Moreover, GLP-1 analogs have demonstrated the capability to enhance neurogenesis, a process recognized for its significance in memory formation and the cognitive and emotional aspects of information processing. Nonetheless, whether semaglutide holds efficacy as both an antidepressant and anxiolytic agent remains uncertain. To address this, our study focused on a mouse model of depression linked to type 2 diabetes induced by a High Fat Diet (HFD). In this model, we administered semaglutide (0.05 mg/Kg intraperitoneally) on a weekly basis to evaluate its potential as a therapeutic option for depression and anxiety. Diabetic mice had higher blood glucose, lipidic profile, and insulin resistance. Moreover, mice fed HFD showed higher serum interleukin (IL)-1ß and lipopolysaccharide (LPS) associated with impaired humor and cognition. The analysis of behavioral responses revealed that the administration of semaglutide effectively mitigated depressive- and anxiety-like behaviors, concurrently demonstrating an enhancement in cognitive function. Additionally, semaglutide treatment protected synaptic plasticity and reversed the hippocampal neuroinflammation induced by HFD fed, improving activation of the insulin pathway, demonstrating the protective effects of semaglutide. We also found that semaglutide treatment decreased astrogliosis and microgliosis in the dentate gyrus region of the hippocampus. In addition, semaglutide prevented the DM2-induced impairments of pro-opiomelanocortin (POMC), and G-protein-coupled receptor 43 (GPR43) and simultaneously increased the NeuN + and Glucagon-like Peptide-1 receptor (GLP-1R+) neurons in the hippocampus. Our data also showed that semaglutide increased the serotonin (5-HT) and serotonin transporter (5-HTT) and glutamatergic receptors in the hippocampus. At last, semaglutide changed the gut microbiota profile (increasing Bacterioidetes, Bacteroides acidifaciens, and Blautia coccoides) and decreased leaky gut, improving the gut-brain axis. Taken together, semaglutide has the potential to act as a therapeutic tool for depression and anxiety.


Assuntos
Ansiedade , Eixo Encéfalo-Intestino , Disfunção Cognitiva , Depressão , Diabetes Mellitus Tipo 2 , Microbioma Gastrointestinal , Peptídeos Semelhantes ao Glucagon , Camundongos Endogâmicos C57BL , Animais , Peptídeos Semelhantes ao Glucagon/farmacologia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/psicologia , Diabetes Mellitus Tipo 2/metabolismo , Camundongos , Disfunção Cognitiva/tratamento farmacológico , Disfunção Cognitiva/prevenção & controle , Disfunção Cognitiva/etiologia , Disfunção Cognitiva/metabolismo , Depressão/tratamento farmacológico , Depressão/psicologia , Depressão/metabolismo , Masculino , Ansiedade/tratamento farmacológico , Ansiedade/psicologia , Ansiedade/etiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Eixo Encéfalo-Intestino/efeitos dos fármacos , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/psicologia , Diabetes Mellitus Experimental/metabolismo , Modelos Animais de Doenças , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico
6.
Metab Brain Dis ; 39(5): 967-984, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38848023

RESUMO

The aging of populations is a global phenomenon that follows a possible increase in the incidence of neurodegenerative diseases. Alzheimer's, Parkinson's, Multiple Sclerosis, Amyotrophic Lateral Sclerosis, and Huntington's diseases are some neurodegenerative disorders that aging could initiate or aggravate. Recent research has indicated that intestinal microbiota dysbiosis can trigger metabolism and brain functioning, contributing to the etiopathogenesis of those neurodegenerative diseases. The intestinal microbiota and its metabolites show significant functions in various aspects, such as the immune system modulation (development and maturation), the maintenance of the intestinal barrier integrity, the modulation of neuromuscular functions in the intestine, and the facilitation of essential metabolic processes for both the microbiota and humans. The primary evidence supporting the connection between intestinal microbiota and its metabolites with neurodegenerative diseases are epidemiological observations and animal models experimentation. This paper reviews up-to-date evidence on the correlation between the microbiota-gut-brain axis and neurodegenerative diseases, with a specially focus on gut metabolites. Dysbiosis can increase inflammatory cytokines and bacterial metabolites, altering intestinal and blood-brain barrier permeability and causing neuroinflammation, thus facilitating the pathogenesis of neurodegenerative diseases. Clinical data supporting this evidence still needs to be improved. Most of the works found are descriptive and associated with the presence of phyla or species of bacteria with neurodegenerative diseases. Despite the limitations of recent research, the potential for elucidating clinical questions that have thus far eluded clarification within prevailing pathophysiological frameworks of health and disease is promising through investigation of the interplay between the host and microbiota.


Assuntos
Eixo Encéfalo-Intestino , Disbiose , Microbioma Gastrointestinal , Doenças Neurodegenerativas , Humanos , Microbioma Gastrointestinal/fisiologia , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/microbiologia , Disbiose/metabolismo , Eixo Encéfalo-Intestino/fisiologia , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/metabolismo
7.
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
8.
Nutrients ; 16(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38732599

RESUMO

In this study, a systematic review of randomized clinical trials conducted from January 2000 to December 2023 was performed to examine the efficacy of psychobiotics-probiotics beneficial to mental health via the gut-brain axis-in adults with psychiatric and cognitive disorders. Out of the 51 studies involving 3353 patients where half received psychobiotics, there was a notably high measurement of effectiveness specifically in the treatment of depression symptoms. Most participants were older and female, with treatments commonly utilizing strains of Lactobacillus and Bifidobacteria over periods ranging from 4 to 24 weeks. Although there was a general agreement on the effectiveness of psychobiotics, the variability in treatment approaches and clinical presentations limits the comparability and generalization of the findings. This underscores the need for more personalized treatment optimization and a deeper investigation into the mechanisms through which psychobiotics act. The research corroborates the therapeutic potential of psychobiotics and represents progress in the management of psychiatric and cognitive disorders.


Assuntos
Transtornos Mentais , Probióticos , Ensaios Clínicos Controlados Aleatórios como Assunto , Humanos , Probióticos/uso terapêutico , Feminino , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/terapia , Transtornos Cognitivos/tratamento farmacológico , Masculino , Resultado do Tratamento , Adulto , Eixo Encéfalo-Intestino/efeitos dos fármacos , Pessoa de Meia-Idade , Microbioma Gastrointestinal/efeitos dos fármacos , Lactobacillus , Idoso , Bifidobacterium
9.
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
10.
Am J Gastroenterol ; 119(7): 1272-1284, 2024 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-38595149

RESUMO

Irritable bowel syndrome (IBS) is responsive to treatments using central neuromodulators. Central neuromodulators work by enhancing the synaptic transmission of 5-hydroxytryptamine, noradrenalin, and dopamine, achieving a slower regulation or desensitization of their postsynaptic receptors. Central neuromodulators act on receptors along the brain-gut axis, so they are useful in treating psychiatric comorbidities, modifying gut motility, improving central downregulation of visceral signals, and enhancing neurogenesis in patients with IBS. Choosing a central neuromodulator for treating IBS should be according to the pharmacological properties and predominant symptoms. The first-line treatment for pain management in IBS is using tricyclic antidepressants. An alternative for pain management is the serotonin and noradrenaline reuptake inhibitors. Selective serotonin reuptake inhibitors are useful when symptoms of anxiety and hypervigilance are dominant but are not helpful for treating abdominal pain. The predominant bowel habit is helpful when choosing a neuromodulator to treat IBS; selective serotonin reuptake inhibitors help constipation, not pain, but may cause diarrhea; tricyclic antidepressants help diarrhea but may cause constipation. A clinical response may occur in 6-8 weeks, but long-term treatment (usually 6-12 months) is required after the initial response to prevent relapse. Augmentation therapy may be beneficial when the therapeutic effect of the first agent is incomplete or associated with side effects. It is recommended to reduce the dose of the first agent and add a second complementary treatment. This may include an atypical antipsychotic or brain-gut behavioral treatment. When tapering central neuromodulators, the dose should be reduced slowly over 4 weeks but may take longer when discontinuation effects occur.


Assuntos
Síndrome do Intestino Irritável , Neurotransmissores , Humanos , Síndrome do Intestino Irritável/tratamento farmacológico , Síndrome do Intestino Irritável/fisiopatologia , Neurotransmissores/uso terapêutico , Antidepressivos Tricíclicos/uso terapêutico , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Eixo Encéfalo-Intestino/fisiologia , Inibidores da Recaptação de Serotonina e Norepinefrina/uso terapêutico
11.
CNS Neurol Disord Drug Targets ; 23(11): 1371-1391, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38500273

RESUMO

INTRODUCTION: Multiple illnesses commonly involve both the Central Nervous System (CNS) and the Gastrointestinal Tract (GI) simultaneously. Consistent evidence suggests that neurological disorders impair GI tract function and worsen the symptomatology and pathophysiology of digestive disorders. On the other hand, it has been proposed that early functional changes in the GI tract contribute to the genesis of several CNS illnesses. Additionally, the role played by the gut in these diseases can be seen as a paradigm for how the gut and the brain interact. METHODS: We mentioned significant GI symptoms and discussed how the GI tract affects central nervous system illnesses, including depression, anxiety, Alzheimer's disease, and Parkinson's disease in this study. We also explored potential pathophysiological underpinnings and novel targets for the creation of future therapies targeted at gut-brain connections. RESULTS & DISCUSSION: In this situation, modulating the gut microbiota through the administration of fecal microbiota transplants or probiotics may represent a new therapeutic option for this population, not only to treat GI problems but also behavioral problems, given the role that dysbiosis and leaky gut play in many neurological disorders. CONCLUSION: Accurate diagnosis and treatment of co-existing illnesses also require coordination between psychiatrists, neurologists, gastroenterologists, and other specialties, as well as a thorough history and thorough physical examination.


Assuntos
Ansiedade , Depressão , Gastroenteropatias , Doenças Neurodegenerativas , Humanos , Depressão/terapia , Gastroenteropatias/terapia , Microbioma Gastrointestinal/fisiologia , Eixo Encéfalo-Intestino/fisiologia , Trato Gastrointestinal
12.
Sleep Breath ; 28(1): 561-563, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-37581760

RESUMO

Sleep disruption, especially that resulting from obstructive sleep apnea (OSA) - a widely prevalent sleep disorder - can lead to important systemic repercussions. We raise a subject of current interest, namely the possible relationship between sleep in general, OSA, and irritable bowel syndrome (IBS), an intestinal disease that can be made worse by stressful events. The intermittent hypoxia caused by OSA can induce alterations in the gut microbiota, which can lead to the dysregulation of the gut-brain axis and the worsening of IBS. This may be considered to be a circular relationship, with OSA playing a crucial role in the worsening of bowel symptoms, which in turn have a negative effect on sleep. Thus, based on previous evidence, we suggest that improving sleep quality could be a key to disrupting this relationship of IBS aggravation and OSA.


Assuntos
Microbioma Gastrointestinal , Síndrome do Intestino Irritável , Apneia Obstrutiva do Sono , Humanos , Microbioma Gastrointestinal/fisiologia , Eixo Encéfalo-Intestino , Sono
13.
Neurotox Res ; 42(1): 4, 2023 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-38103074

RESUMO

The gut-brain axis is an essential communication pathway between the central nervous system (CNS) and the gastrointestinal tract. The human microbiota is composed of a diverse and abundant microbial community that compasses more than 100 trillion microorganisms that participate in relevant physiological functions such as host nutrient metabolism, structural integrity, maintenance of the gut mucosal barrier, and immunomodulation. Recent evidence in animal models has been instrumental in demonstrating the possible role of the microbiota in neurodevelopment, neuroinflammation, and behavior. Furthermore, clinical studies suggested that adverse changes in the microbiota can be considered a susceptibility factor for neurological disorders (NDs), such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS). In this review, we will discuss evidence describing the role of gut microbes in health and disease as a relevant risk factor in the pathogenesis of neurodegenerative disorders, including AD, PD, HD, and ALS.


Assuntos
Doença de Alzheimer , Esclerose Lateral Amiotrófica , Microbioma Gastrointestinal , Doença de Huntington , Doenças Neurodegenerativas , Doença de Parkinson , Animais , Humanos , Eixo Encéfalo-Intestino , Microbioma Gastrointestinal/fisiologia , Doenças Neurodegenerativas/patologia , Sistema Nervoso Central , Doença de Parkinson/patologia , Doença de Huntington/patologia
14.
Biomolecules ; 13(10)2023 10 10.
Artigo em Inglês | MEDLINE | ID: mdl-37892186

RESUMO

Diabetic patients are more affected by depression than non-diabetics, and this is related to greater treatment resistance and associated with poorer outcomes. This increase in the prevalence of depression in diabetics is also related to hyperglycemia and hypercortisolism. In diabetics, the hyperactivity of the HPA axis occurs in parallel to gut dysbiosis, weakness of the intestinal permeability barrier, and high bacterial-product translocation into the bloodstream. Diabetes also induces an increase in the permeability of the blood-brain barrier (BBB) and Toll-like receptor 4 (TLR4) expression in the hippocampus. Furthermore, lipopolysaccharide (LPS)-induced depression behaviors and neuroinflammation are exacerbated in diabetic mice. In this context, we propose here that hypercortisolism, in association with gut dysbiosis, leads to an exacerbation of hippocampal neuroinflammation, glutamatergic transmission, and neuronal apoptosis, leading to the development and aggravation of depression and to resistance to treatment of this mood disorder in diabetic patients.


Assuntos
Síndrome de Cushing , Transtorno Depressivo , Diabetes Mellitus Experimental , Humanos , Camundongos , Animais , Eixo Encéfalo-Intestino , Sistema Hipotálamo-Hipofisário/fisiologia , Doenças Neuroinflamatórias , Disbiose , Sistema Hipófise-Suprarrenal/fisiologia
15.
Cells ; 12(13)2023 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-37443769

RESUMO

The term 'perinatal environment' refers to the period surrounding birth, which plays a crucial role in brain development. It has been suggested that dynamic communication between the neuro-immune system and gut microbiota is essential in maintaining adequate brain function. This interaction depends on the mother's status during pregnancy and/or the newborn environment. Here, we show experimental and clinical evidence that indicates that the perinatal period is a critical window in which stress-induced immune activation and altered microbiota compositions produce lasting behavioral consequences, although a clear causative relationship has not yet been established. In addition, we discuss potential early treatments for preventing the deleterious effect of perinatal stress exposure. In this sense, early environmental enrichment exposure (including exercise) and melatonin use in the perinatal period could be valuable in improving the negative consequences of early adversities. The evidence presented in this review encourages the realization of studies investigating the beneficial role of melatonin administration and environmental enrichment exposure in mitigating cognitive alteration in offspring under perinatal stress exposure. On the other hand, direct evidence of microbiota restoration as the main mechanism behind the beneficial effects of this treatment has not been fully demonstrated and should be explored in future studies.


Assuntos
Eixo Encéfalo-Intestino , Encéfalo , Disfunção Cognitiva , Exposição Materna , Efeitos Tardios da Exposição Pré-Natal , Estresse Psicológico , Disfunção Cognitiva/imunologia , Disfunção Cognitiva/microbiologia , Disfunção Cognitiva/prevenção & controle , Humanos , Feminino , Animais , Efeitos Tardios da Exposição Pré-Natal/etiologia , Melatonina/administração & dosagem , Encéfalo/crescimento & desenvolvimento , Neurogênese , Antioxidantes/administração & dosagem , Probióticos/administração & dosagem
16.
Neuroscience ; 526: 21-34, 2023 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-37331688

RESUMO

Parkinson's Disease is a synucleinopathy that primarily affects the dopaminergic cells of the central nervous system, leading to motor and gastrointestinal disturbances. However, intestinal peripheral neurons undergo a similar neurodegeneration process, marked by α-synuclein (αSyn) accumulation and loss of mitochondrial homeostasis. We investigated the metabolic alterations in different biometrics that compose the gut-brain axis (blood, brain, large intestine, and feces) in an MPTP-induced mouse model of sporadic Parkinson's Disease. Animals received escalating administration of MPTP. Tissues and fecal pellets were collected, and the metabolites were identified through the untargeted Nuclear Magnetic Resonance spectroscopic (1H NMR) technique. We found differences in many metabolites from all the tissues evaluated. The differential expression of metabolites in these samples mainly reflects inflammatory aspects, cytotoxicity, and mitochondrial impairment (oxidative stress and energy metabolism) in the animal model used. The direct evaluation of fecal metabolites revealed changes in several classes of metabolites. This data reinforces previous studies showing that Parkinson's disease is associated with metabolic perturbation not only in brain-related tissues, but also in periphery structures such as the gut. In addition, the evaluation of the microbiome and metabolites from gut and feces emerge as promising sources of information for understanding the evolution and progression of sporadic Parkinson's Disease.


Assuntos
Microbioma Gastrointestinal , Intoxicação por MPTP , Doença de Parkinson , Transtornos Parkinsonianos , Camundongos , Animais , Doença de Parkinson/metabolismo , Eixo Encéfalo-Intestino , Microbioma Gastrointestinal/fisiologia , Espectroscopia de Ressonância Magnética , Modelos Animais de Doenças
17.
Arq Gastroenterol ; 60(1): 144-154, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37194773

RESUMO

BACKGROUND: Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disease, characterized by the accumulation of amyloid plaques and neurofibrillary tangles in the brain. Several pathways enable bidirectional communication between the central nervous system (CNS), the intestine and its microbiota, constituting the microbiota-gut-brain axis. OBJECTIVE: Review the pathophysiology of AD, relate it to the microbiota-gut-brain axis and discuss the possibility of using probiotics in the treatment and/or prevention of this disease. METHODS: Search of articles from the PubMed database published in the last 5 years (2017 to 2022) structure the narrative review. RESULTS: The composition of the gut microbiota influences the CNS, resulting in changes in host behavior and may be related to the development of neurodegenerative diseases. Some metabolites produced by the intestinal microbiota, such as trimethylamine N-oxide (TMAO), may be involved in the pathogenesis of AD, while other compounds produced by the microbiota during the fermentation of food in the intestine, such as D-glutamate and fatty acids short chain, are beneficial in cognitive function. The consumption of live microorganisms beneficial to health, known as probiotics, has been tested in laboratory animals and humans to evaluate the effect on AD. CONCLUSION: Although there are few clinical trials evaluating the effect of probiotic consumption in humans with AD, the results to date indicate a beneficial contribution of the use of probiotics in this disease.


Assuntos
Doença de Alzheimer , Microbioma Gastrointestinal , Doenças Neurodegenerativas , Humanos , Animais , Doença de Alzheimer/patologia , Doença de Alzheimer/terapia , Doenças Neurodegenerativas/patologia , Eixo Encéfalo-Intestino , Encéfalo/patologia , Encéfalo/fisiologia , Microbioma Gastrointestinal/fisiologia
18.
J Integr Neurosci ; 22(3): 65, 2023 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-37258450

RESUMO

Depression is the leading cause of disability worldwide, contributing to the global disease burden. From above, it is a priority to investigate models that fully explain its physiopathology to develop new treatments. In the last decade, many studies have shown that gut microbiota (GM) dysbiosis influences brain functions and participate, in association with immunity, in the pathogenesis of depression. Thereby, GM modulation could be a novel therapeutic target for depression. This review aims to evidence how the GM and the immune system influence mental illness, particularly depression. Here, we focus on the communication mechanisms between the intestine and the brain and the impact on the development of neuroinflammation contributing to the development of Major Depressive Disorder (MDD). However, most of the current findings are in animal models, suggesting the need for studies in humans. In addition, more analysis of metabolites and cytokines are needed to identify new pathophysiological mechanisms improving anti-depression treatments.


Assuntos
Transtorno Depressivo Maior , Microbioma Gastrointestinal , Animais , Humanos , Transtorno Depressivo Maior/terapia , Eixo Encéfalo-Intestino , Doenças Neuroinflamatórias , Encéfalo
20.
Physiol Behav ; 260: 114070, 2023 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-36574940

RESUMO

Excessive stress can precipitate depression and anxiety diseases, and damage gastrointestinal functionality and microbiota changes, favoring the development of functional gastrointestinal disorders (FGIDs) - defined by dysregulation in the brain-gut interaction. Therefore, the present study investigated if Emotional-Single Prolonged Stress (E-SPS) induces depressive/anxiety-like phenotype and gut dysfunction in adult Swiss male mice. For this, mice of the E-SPS group were subjected to three stressors sequential exposure: immobilization, swimming, and odor of the predator for 7 days (incubation period). Next, animals performed behavior tests and 24 h later, samples of feces, blood, and colon tissue were collected. E-SPS increased the plasma corticosterone levels, immobility time in the tail suspension and forced swim test, decreased the grooming time in the splash test, OAT%, and OAE% in the elevated plus-maze test, as well as increased anxiety index. Mice of E-SPS had increased % of intestinal transit rate, % of fecal moisture content, and fecal pellets number, and decreased Claudin1 content in the colon. E-SPS decreased the relative abundance of Bacteroidetes phylum, Bacteroidia class, Bacteroidales order, Muribaculaceae and Porphyromonadaceae family, Muribaculum, and Duncaniella genus. However, E-SPS increased Firmicutes and Actinobacteria phylum, Coriobacteriales order, and the ratio of Firmicutes/Bacteroidetes, and demonstrated Mucispirillum genus presence. The present study showed that E-SPS induced depressive/anxiety-like phenotype, predominant diarrhea gut dysfunction, and modulated the gut bacterial microbiota profile in male adult Swiss mice. E-SPS might be a promising model for future studies on the brain-gut interaction and the development of FGIDs with psychological comorbidities.


Assuntos
Encéfalo , Microbiota , Animais , Masculino , Camundongos , Ansiedade , Transtornos de Ansiedade , Bactérias/genética , Estresse Psicológico/psicologia , Eixo Encéfalo-Intestino
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA