Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Autism ; : 13623613241246911, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38666595

RESUMO

LAY ABSTRACT: Autism spectrum disorder is a neurodevelopmental disorder characterized by a wide range of behavioral alterations, including impaired social interaction and repetitive behaviors. Numerous pharmacological interventions have been developed for autism spectrum disorder, often proving ineffective and accompanied by a multitude of side effects. The gut microbiota is the reservoir of bacteria inhabiting our gastrointestinal tract. The gut microbial alterations observed in individuals with autism spectrum disorder, including elevated levels of Bacteroidetes, Firmicutes, and Proteobacteria, as well as reduced levels of Bifidobacterium, provide a basis for further investigation into the role of the gut microbiota in autism spectrum disorder. Recent preclinical studies have shown favorable outcomes with probiotic therapy, including improvements in oxidative stress, anti-inflammatory effects, regulation of neurotransmitters, and restoration of microbial balance. The aim of this review is to explore the potential of probiotics for the management and treatment of autism spectrum disorder, by investigating insights from recent studies in animals.

2.
Depress Anxiety ; 39(5): 407-418, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35357051

RESUMO

BACKGROUND: Major depressive disorder (MDD) is characterized by a high rate of treatment resistance. Omega (ω)-3 polyunsaturated fatty acids (PUFAs) were shown to correlate with depressive phenotype both in rodents and in humans. However, few studies to date have investigated the role of PUFAs in antidepressant response. The primary aim of this study was to assess the link between baseline PUFA composition and changes in depressive symptoms as well as antidepressant response in a multicenter study of depressed patients. METHODS: Sixty depressed adults who met criteria for MDD according to DSM-IV-TR were recruited. Neuropsychiatric evaluations occurred at baseline and after 4 and 8 weeks of treatment with standard antidepressants, including escitalopram (N = 45), sertraline (N = 13) and venlafaxine (N = 2). At study endpoint, patients were stratified into responders (R) or non-responders (NR) based on their MADRS (Montgomery-Åsberg Depression Rating Scale) score. Baseline PUFA levels were assessed and their association with clinical response was determined. RESULTS: Lower ω-3 PUFA levels were associated to worse baseline symptomatology. Baseline levels of PUFAs were significantly different between R and NR, with R exhibiting lower docosahexaenoic acid (DHA), eicosapentaenoic acid (EPA) and ω-3 index; and higher ω-6/ω-3 ratio than NR before the start of antidepressant treatment. DHA levels as well as the ω-3 index and ω-6/ω-3 ratio significantly predicted response to antidepressants at study endpoint. CONCLUSIONS: These results show that baseline levels of PUFAs predict later response to standard antidepressants in depressed subjects. They suggest that PUFA intake and/or metabolism represent a novel modifiable tool for the management of unresponsive depressed patients.


Assuntos
Transtorno Depressivo Maior , Ácidos Graxos Ômega-3 , Antidepressivos/uso terapêutico , Transtorno Depressivo Maior/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ácido Eicosapentaenoico/farmacologia , Ácido Eicosapentaenoico/uso terapêutico , Ácidos Graxos Ômega-3/uso terapêutico , Ácidos Graxos Insaturados/uso terapêutico , Humanos
3.
Int J Obes (Lond) ; 46(4): 885-888, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35001078

RESUMO

Converging data support the role of chronic low-grade inflammation in depressive symptomatology in obesity. One mechanism likely to be involved relies on the effects of inflammation on tryptophan (TRP) metabolism. While recent data document alterations in the indole pathway of TRP metabolism in obesity, the relevance of this mechanism to obesity-related depressive symptoms has not been investigated. The aim of this preliminary study was to assess the association between plasma levels of TRP and indole metabolites and depressive symptoms in 44 subjects with severe or morbid obesity, free of clinically relevant neuropsychiatric disorders. The interaction effect of inflammation, reflected in serum high-sensitive C-reactive protein (hsCRP) levels, and indoles on depressive symptoms was also determined. Higher serum levels of hsCRP and lower concentrations of TRP and indoles, particularly indole-3-carboxaldehyde (IAld), correlated with more severe depressive symptoms. Interestingly, the effect of high hsCRP levels in predicting greater depressive symptoms was potentiated by low IAld levels. These results comfort the link between inflammation, the indole pathway of TRP metabolism, and obesity-related depressive symptoms.


Assuntos
Cinurenina , Triptofano , Proteína C-Reativa/metabolismo , Depressão/metabolismo , Humanos , Indóis , Inflamação/metabolismo , Cinurenina/metabolismo , Obesidade/complicações , Triptofano/metabolismo
4.
Mod Trends Psychiatry ; 32: 113-133, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34032649

RESUMO

The human gut microbiome plays a key role in host physiology in health and disease. There is a growing emphasis on the bidirectional interaction between various medications and the gut microbiome. Here, we will first review how drugs can affect microbiome composition and how the microbiome can alter the pharmacodynamics and potentially pharmacokinetics of psychotropic medications. We will take into consideration different classes of psychotropics, including antipsychotics, antidepressants, antianxiety drugs, anticonvulsants/mood stabilisers, opioid analgesics, drugs of abuse, alcohol, nicotine, and xanthines. The varying effects of these widely used medications on microorganisms are becoming apparent from in vivo and in vitro studies. This has important implications for future drug discovery in psychiatry which will need to consider the host microbiome as a major potential target.


Assuntos
Transtornos Mentais , Psicotrópicos/farmacocinética , Descoberta de Drogas , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Humanos , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/microbiologia , Psiquiatria/tendências , Psicotrópicos/classificação
5.
EBioMedicine ; 66: 103307, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33819741

RESUMO

BACKGROUND: The role of the gut microbiome in the biotransformation of drugs has recently come under scrutiny. It remains unclear whether the gut microbiome directly influences the extent of drug absorbed after oral administration and thus potentially alters clinical pharmacokinetics. METHODS: In this study, we evaluated whether changes in the gut microbiota of male Sprague Dawley rats, as a result of either antibiotic or probiotic administration, influenced the oral bioavailability of two commonly prescribed antipsychotics, olanzapine and risperidone. FINDINGS: The bioavailability of olanzapine, was significantly increased (1.8-fold) in rats that had undergone antibiotic-induced depletion of gut microbiota, whereas the bioavailability of risperidone was unchanged. There was no direct effect of microbiota depletion on the expression of major CYP450 enzymes involved in the metabolism of either drug. However, the expression of UGT1A3 in the duodenum was significantly downregulated. The reduction in faecal enzymatic activity, observed during and after antibiotic administration, did not alter the ex vivo metabolism of olanzapine or risperidone. The relative abundance of Alistipes significantly correlated with the AUC of olanzapine but not risperidone. INTERPRETATION: Alistipes may play a role in the observed alterations in olanzapine pharmacokinetics. The gut microbiome might be an important variable determining the systemic bioavailability of orally administered olanzapine. Additional research exploring the potential implication of the gut microbiota on the clinical pharmacokinetics of olanzapine in humans is warranted. FUNDING: This research is supported by APC Microbiome Ireland, a research centre funded by Science Foundation Ireland (SFI), through the Irish Government's National Development Plan (grant no. 12/RC/2273 P2) and by Nature Research-Yakult (The Global Grants for Gut Health; Ref No. 626891).


Assuntos
Microbioma Gastrointestinal , Olanzapina/farmacocinética , Inibidores Seletivos de Recaptação de Serotonina/farmacocinética , Administração Oral , Animais , Antibacterianos/farmacologia , Biodiversidade , Disponibilidade Biológica , Cromatografia Líquida de Alta Pressão , Monitoramento de Medicamentos , Fezes/microbiologia , Masculino , Estrutura Molecular , Olanzapina/administração & dosagem , Olanzapina/química , Probióticos , Ratos , Inibidores Seletivos de Recaptação de Serotonina/administração & dosagem , Inibidores Seletivos de Recaptação de Serotonina/química
7.
Front Immunol ; 11: 557, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32351500

RESUMO

Background: Obesity is a condition with a complex pathophysiology characterized by both chronic low-grade inflammation and changes in the gut microbial ecosystem. These alterations can affect the metabolism of tryptophan (TRP), an essential amino acid and precursor of serotonin (5-HT), kynurenine (KYN), and indoles. This study aimed to investigate alterations in KYN and microbiota-mediated indole routes of TRP metabolism in obese subjects relatively to non-obese controls and to determine their relationship with systemic inflammation. Methods: Eighty-five obese adults (avg. BMI = 40.48) and 42 non-obese control individuals (avg. BMI = 24.03) were recruited. Plasma levels of TRP catabolites were assessed using Ultra High Performance Liquid Chromatography-ElectroSpray-Ionization-Tandem Mass Spectrometry. High-sensitive C-reactive protein (hsCRP) and high-sensitive interleukin 6 (hsIL-6) were measured in the serum as markers of systemic inflammation using enzyme-linked immunosorbent assay. Results: Both KYN and microbiota-mediated indole routes of TRP metabolism were altered in obese subjects, as reflected in higher KYN/TRP ratio and lower 5-HT and indoles levels, relatively to non-obese controls. HsIL-6 and hsCRP were increased in obesity and were overall associated with TRP metabolic pathways alterations. Conclusion: These results indicate for the first time that KYN and indole TRP metabolic pathways are concomitantly altered in obese subjects and highlight their respective associations with obesity-related systemic inflammation.


Assuntos
Inflamação/metabolismo , Cinurenina/metabolismo , Redes e Vias Metabólicas/imunologia , Obesidade/metabolismo , Triptofano/metabolismo , Adulto , Feminino , Humanos , Inflamação/imunologia , Masculino , Obesidade/imunologia
8.
Harv Rev Psychiatry ; 28(1): 26-39, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31913980

RESUMO

Microorganisms can be found in virtually any environment. In humans, the largest collection of microorganisms is found in the gut ecosystem. The adult gut microbiome consists of more genes than its human host and typically spans more than 60 genera from across the taxonomic tree. In addition, the gut contains the largest number of neurons in the body, after the brain. In recent years, it has become clear that the gut microbiome is in communication with the brain, through the gut-brain axis. A growing body of literature shows that the gut microbiome plays a shaping role in a variety of psychiatric disorders, including major depressive disorder (MDD). In this review, the interplay between the microbiome and MDD is discussed in three facets. First, we discuss factors that affect the onset/development of MDD that also greatly impinge on the composition of the gut microbiota-especially diet and stressful life events. We then examine the interplay between the microbiota and MDD. We examine evidence suggesting that the microbiota is altered in MDD, and we discuss why the microbiota should be considered during MDD treatment. Finally, we look toward the future and examine how the microbiota might become a therapeutic target for MDD. This review is intended to introduce those familiar with the neurological and psychiatric aspects of MDD to the microbiome and its potential role in the disorder. Although research is in its very early days, with much yet to be the understood, the microbiome is offering new avenues for developing potentially novel strategies for managing MDD.


Assuntos
Transtorno Depressivo Maior/metabolismo , Transtorno Depressivo Maior/microbiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/microbiologia , Animais , Encéfalo/metabolismo , Encéfalo/microbiologia , Transtorno Depressivo Maior/dietoterapia , Trato Gastrointestinal/fisiologia , Humanos , Probióticos/uso terapêutico
9.
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
10.
Psychopharmacology (Berl) ; 236(5): 1411-1432, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30806744

RESUMO

The human gut contains trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. Psychotropic medications act on the central nervous system (CNS) and are used in the treatment of various psychiatric disorders. There is increasing emphasis on the bidirectional interaction between drugs and the gut microbiome. An expanding body of evidence supports the notion that microbes can metabolise drugs and vice versa drugs can modify the gut microbiota composition. In this review, we will first give a comprehensive introduction about this bidirectional interaction, then we will take into consideration different classes of psychotropics including antipsychotics, antidepressants, antianxiety drugs, anticonvulsants/mood stabilisers, opioid analgesics, drugs of abuse, alcohol, nicotine and xanthines. The varying effects of these widely used medications on microorganisms are becoming apparent from in vivo and in vitro studies. This has important implications for the future of psychopharmacology pipelines that will routinely need to consider the host microbiome during drug discovery and development.


Assuntos
Antidepressivos/farmacologia , Antipsicóticos/farmacologia , Microbioma Gastrointestinal/efeitos dos fármacos , Psicotrópicos/farmacologia , Antidepressivos/uso terapêutico , Antipsicóticos/uso terapêutico , Microbioma Gastrointestinal/fisiologia , Humanos , Transtornos Mentais/tratamento farmacológico , Transtornos Mentais/psicologia , Microbiota/efeitos dos fármacos , Microbiota/fisiologia , Psicotrópicos/uso terapêutico
11.
Psychopharmacology (Berl) ; 236(5): 1671-1685, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30155748

RESUMO

RATIONALE: Growing evidence supports a role for the microbiota in regulating gut-brain interactions and, thus, psychiatric disorders. Despite substantial scientific efforts to delineate the mechanism of action of psychotropic medications at a central nervous system (CNS) level, there remains a critical lack of understanding on how these drugs might affect the microbiota and gut physiology. OBJECTIVES: We investigated the antimicrobial activity of psychotropics against two bacterial strain residents in the human gut, Lactobacillus rhamnosus and Escherichia coli. In addition, we examined the impact of chronic treatment with these drugs on microbiota and intestinal parameters in the rat. RESULTS: In vitro fluoxetine and escitalopram showed differential antimicrobial effects. Lithium, valproate and aripiprazole administration significantly increased microbial species richness and diversity, while the other treatments were not significantly different from controls. At the genus level, several species belonging to Clostridium, Peptoclostridium, Intestinibacter and Christenellaceae were increased following treatment with lithium, valproate and aripiprazole when compared to the control group. Animals treated with escitalopram, venlafaxine, fluoxetine and aripiprazole exhibited an increased permeability in the ileum. CONCLUSIONS: These data show that psychotropic medications differentially influence the composition of gut microbiota in vivo and that fluoxetine and escitalopram have specific antimicrobial activity in vitro. Interestingly, drugs that significantly altered gut microbial composition did not increase intestinal permeability, suggesting that the two factors are not causally linked. Overall, unravelling the impact of psychotropics on gastrointestinal and microbiota measures offers the potential to provide critical insight into the mechanism of action and side effects of these medications.


Assuntos
Escherichia coli/efeitos dos fármacos , Microbioma Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/efeitos dos fármacos , Lacticaseibacillus rhamnosus/efeitos dos fármacos , Psicotrópicos/farmacologia , Animais , Antibacterianos/farmacologia , Relação Dose-Resposta a Droga , Escherichia coli/fisiologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiologia , Humanos , Lacticaseibacillus rhamnosus/fisiologia , Masculino , Ratos , Ratos Sprague-Dawley
12.
Front Neuroendocrinol ; 51: 80-101, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29753796

RESUMO

The human gut harbours trillions of symbiotic bacteria that play a key role in programming different aspects of host physiology in health and disease. These intestinal microbes are also key components of the gut-brain axis, the bidirectional communication pathway between the gut and the central nervous system (CNS). In addition, the CNS is closely interconnected with the endocrine system to regulate many physiological processes. An expanding body of evidence is supporting the notion that gut microbiota modifications and/or manipulations may also play a crucial role in the manifestation of specific behavioural responses regulated by neuroendocrine pathways. In this review, we will focus on how the intestinal microorganisms interact with elements of the host neuroendocrine system to modify behaviours relevant to stress, eating behaviour, sexual behaviour, social behaviour, cognition and addiction.


Assuntos
Comportamento Aditivo , Encéfalo/fisiologia , Comportamento Alimentar/fisiologia , Microbioma Gastrointestinal/fisiologia , Aprendizagem/fisiologia , Sistemas Neurossecretores/fisiologia , Comportamento Sexual/fisiologia , Comportamento Social , Estresse Psicológico , Animais , Comportamento Aditivo/metabolismo , Comportamento Aditivo/fisiopatologia , Encéfalo/metabolismo , Humanos , Sistemas Neurossecretores/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
13.
Neurosci Lett ; 648: 14-20, 2017 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-28351776

RESUMO

Approximately 50% of depressed individuals fail to achieve remission with first-line antidepressant drugs and a third remain treatment-resistant. When first-line antidepressant treatment is unsuccessful, second-line strategies include dose optimisation, switching to another antidepressant, combination with another antidepressant, or augmentation with a non-antidepressant medication. Much of the evidence for the efficacy of augmentation strategies comes from studies using lithium to augment the effects of tricyclic antidepressants. The neural circuitry underlying the therapeutic effects of lithium augmentation is not yet fully understood. Recently, we reported that chronic treatment with a combination of lithium and the antidepressant desipramine, exerted antidepressant-like behavioural effects in a mouse strain (BALB/cOLaHsd) that did not exhibit an antidepressant-like behavioural response to either drug alone. In the present study, we used this model in combination with ΔFosB/FosB immunohistochemistry to identify brain regions chronically affected by lithium augmentation of desipramine when compared to either treatment alone. The data suggest that the dorsal raphe nucleus and the CA3 regions of the dorsal hippocampus are key nodes in the neural circuitry underlying antidepressant action of lithium augmentation of desipramine. These data give new insight into the neurobiology underlying the mechanism of lithium augmentation in the context of treatment-resistant depression.


Assuntos
Antidepressivos Tricíclicos/administração & dosagem , Desipramina/administração & dosagem , Núcleo Dorsal da Rafe/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Lítio/administração & dosagem , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Contagem de Células , Núcleo Dorsal da Rafe/metabolismo , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Proto-Oncogênicas c-fos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...