Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 79
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Clin Exp Allergy ; 51(9): 1133-1143, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34192396

RESUMEN

BACKGROUND: Cough is a common troublesome symptom in asthma which is neuronally mediated. Limosilactobacillus reuteri DSM-17938 (L. reuteri DSM-17938) is a probiotic shown to be effective in pre-clinical models at suppressing neuronal responses to capsaicin, a transient receptor potential vanilloid agonist (TRPV1). OBJECTIVE: Investigate the effects of DSM-17938 versus matched placebo on capsaicin-evoked coughs in mild allergic asthmatics. METHODS: We performed a 4-visit, randomized, double-blind, placebo-controlled, two-way cross-over study comparing full dose cough responses with inhaled capsaicin in mild allergic asthmatics after 1 month of treatment with DSM-17938 compared with matched placebo. Randomization and allocation to trial group were carried out by a central computer system. Histamine skin prick testing, airway hyper-responsiveness and inflammatory cells in induced sputum were measured at every visit. Blood was collected to extract PBMCs and stimulated with CD3/CD28 to ascertain the effects of DSM-17938 /placebo on T-cell cytokine responses. RESULTS: Seventeen subjects were recruited and 15 completed the study (8 females, mean age 27.3 years). There was no difference in the change in maximum capsaicin-evoked coughs (Emax) after treatment with L. reuteri DSM-17938 compared with placebo [mean difference 2.07 coughs (95% CI -2.77 to 6.91, p = .38) or relative changes in geometric mean ratios for the dose evoking at least half the Emax (ED50) [1.05 (95% CI 0.31-3.58, p = .94)], concentration evoking 2 coughs (C2) [0.63 (0.26-1.53), p = .28] and 5 coughs (C5) [0.79 (0.25-2.50), p = .67]. There was no effect on histamine skin prick wheal size, intensity of itch sensation, methacholine PC20, airway inflammation or T-cell responses after stimulation with CD3/CD28. There were no serious adverse events. One subject developed a mild upper respiratory tract infection and another mild transient nausea whilst on DSM-17938. CONCLUSION: In this small study in adults with mild allergic asthma, we found no evidence that L. reuteri DSM-17938 has any systemic effects on airway nerves, smooth muscle, sputum inflammatory cells, skin responses or T-cell responses after oral consumption. TRIAL REGISTRATION: Clinicaltrials.gov Identifier: NCT03603522.


Asunto(s)
Asma/complicaciones , Tos/etiología , Tos/prevención & control , Limosilactobacillus reuteri , Probióticos/uso terapéutico , Adulto , Estudios Cruzados , Método Doble Ciego , Femenino , Humanos , Masculino , Resultado del Tratamiento
2.
Brain Behav Immun ; 84: 154-163, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31785396

RESUMEN

Growing evidence suggests that environmental disruptors of maternal microbes may have significant detrimental consequences for the developing fetus. Antibiotic exposure during early life can have long-term effects on neurodevelopment in mice and humans. Here we explore whether exposure to low-dose penicillin during only the last week of gestation in mice has long-term effects on offspring behaviour, brain, immune function, and gut microbiota. We found that this treatment had sex-specific effects in the adult mouse offspring. Female, but not male, mice demonstrated decreased anxiety-like behaviours, while male, but not female, mice had abnormal social behaviours which correlated with altered brain expression of AVPR1A, AVPR1B, and OXTR, and decreases in the balance of splenic FOXP3+ regulatory T cells. Prenatal penicillin exposure also led to distinct microbiota compositions that clustered differently by sex. These data suggest that exposure of pregnant mice to even a low dose of penicillin through only the last week before birth is nonetheless sufficient to induce long-term sex-specific developmental changes in both male and female offspring.


Asunto(s)
Conducta Animal/efectos de los fármacos , Microbioma Gastrointestinal/efectos de los fármacos , Inmunidad/efectos de los fármacos , Penicilinas/administración & dosificación , Penicilinas/farmacología , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Caracteres Sexuales , Conducta Social , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Masculino , Ratones , Embarazo , Efectos Tardíos de la Exposición Prenatal/inducido químicamente , Efectos Tardíos de la Exposición Prenatal/microbiología
3.
Brain Behav Immun ; 77: 7-15, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30582961

RESUMEN

The use of antibiotics has recently risen to prominence in neuroscience due to their potential value in studying the microbiota-gut-brain axis. In this context they have been largely employed to illustrate the many influences of the gut microbiota on brain function and behaviour. Much of this research is bolstered by the abnormal behaviour seen in germ-free animals and other well-controlled experiments. However, this literature has largely failed to consider the neuroactive potential of antibiotics themselves, independent from, or in addition to, their microbicidal effects. This is problematic, as clinical as well as experimental literature, largely neglected through the past decade, has clearly demonstrated that broad classes of antibiotics are neuroactive or neurotoxic. This is true even for some antibiotics that are widely regarded as not absorbed in the intestinal tract, and is especially concerning when considering the highly-concentrated and widely-ranging doses that have been used. In this review we will critically survey the clinical and experimental evidence that antibiotics may influence a variety of nervous system functions, from the enteric nervous system through to the brain and resultant behaviour. We will discuss substantial evidence which clearly suggests neuro-activity or -toxicity by most classes of antibiotics. We will conclude that, while evidence for the microbiota-gut-brain axis remains strong, clinical and experimental studies which employ antibiotics to probe it must consider this potential confound.


Asunto(s)
Antibacterianos/farmacología , Encéfalo/efectos de los fármacos , Microbioma Gastrointestinal/fisiología , Animales , Encéfalo/fisiología , Sistema Nervioso Central/efectos de los fármacos , Depresores del Sistema Nervioso Central/metabolismo , Sistema Nervioso Entérico/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Humanos , Intestinos/microbiología , Microbiota/fisiología , Sistema Nervioso/efectos de los fármacos
4.
BMC Med ; 15(1): 7, 2017 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-28073366

RESUMEN

BACKGROUND: Stress-related disorders involve systemic alterations, including disruption of the intestinal microbial community. Given the putative connections between the microbiota, immunity, neural function, and behaviour, we investigated the potential for microbe-induced gut-to-brain signalling to modulate the impact of stress on host behaviour and immunoregulation. METHODS: Male C57BL/6 mice treated orally over 28 days with either Lactobacillus rhamnosus (JB-1) ™ or vehicle were subjected to chronic social defeat and assessed for alterations in behaviour and immune cell phenotype. 16S rRNA sequencing and mass spectrometry were employed to analyse the faecal microbial community and metabolite profile. RESULTS: Treatment with JB-1 decreased stress-induced anxiety-like behaviour and prevented deficits in social interaction with conspecifics. However, JB-1 did not alter development of aggressor avoidance following social defeat. Microbial treatment attenuated stress-related activation of dendritic cells while increasing IL-10+ regulatory T cells. Furthermore, JB-1 modulated the effect of stress on faecal metabolites with neuroactive and immunomodulatory properties. Exposure to social defeat altered faecal microbial community composition and reduced species richness and diversity, none of which was prevented by JB-1. Stress-related microbiota disruptions persisted in vehicle-treated mice for 3 weeks following stressor cessation. CONCLUSIONS: These data demonstrate that despite the complexity of the gut microbiota, exposure to a single microbial strain can protect against certain stress-induced behaviours and systemic immune alterations without preventing dysbiosis. This work supports microbe-based interventions for stress-related disorders.


Asunto(s)
Microbioma Gastrointestinal/efectos de los fármacos , Lacticaseibacillus rhamnosus , Probióticos/administración & dosificación , ARN Ribosómico 16S/efectos de los fármacos , Estrés Psicológico/prevención & control , Animales , Ansiedad/prevención & control , Conducta Animal/efectos de los fármacos , Encéfalo/metabolismo , Interleucina-10/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Probióticos/farmacología , Estrés Psicológico/genética
5.
Brain Behav Immun ; 61: 50-59, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27865949

RESUMEN

BACKGROUND: Preclinical studies have identified certain probiotics as psychobiotics - live microorganisms with a potential mental health benefit. Lactobacillus rhamnosus (JB-1) has been shown to reduce stress-related behaviour, corticosterone release and alter central expression of GABA receptors in an anxious mouse strain. However, it is unclear if this single putative psychobiotic strain has psychotropic activity in humans. Consequently, we aimed to examine if these promising preclinical findings could be translated to healthy human volunteers. OBJECTIVES: To determine the impact of L. rhamnosus on stress-related behaviours, physiology, inflammatory response, cognitive performance and brain activity patterns in healthy male participants. METHODS: An 8week, randomized, placebo-controlled, cross-over design was employed. Twenty-nine healthy male volunteers participated. Participants completed self-report stress measures, cognitive assessments and resting electroencephalography (EEG). Plasma IL10, IL1ß, IL6, IL8 and TNFα levels and whole blood Toll-like 4 (TLR-4) agonist-induced cytokine release were determined by multiplex ELISA. Salivary cortisol was determined by ELISA and subjective stress measures were assessed before, during and after a socially evaluated cold pressor test (SECPT). RESULTS: There was no overall effect of probiotic treatment on measures of mood, anxiety, stress or sleep quality and no significant effect of probiotic over placebo on subjective stress measures, or the HPA response to the SECPT. Visuospatial memory performance, attention switching, rapid visual information processing, emotion recognition and associated EEG measures did not show improvement over placebo. No significant anti-inflammatory effects were seen as assessed by basal and stimulated cytokine levels. CONCLUSIONS: L. rhamnosus was not superior to placebo in modifying stress-related measures, HPA response, inflammation or cognitive performance in healthy male participants. These findings highlight the challenges associated with moving promising preclinical studies, conducted in an anxious mouse strain, to healthy human participants. Future interventional studies investigating the effect of this psychobiotic in populations with stress-related disorders are required.


Asunto(s)
Atención/efectos de los fármacos , Cognición/efectos de los fármacos , Lacticaseibacillus rhamnosus , Probióticos/administración & dosificación , Estrés Psicológico/tratamiento farmacológico , Adulto , Encéfalo/efectos de los fármacos , Cognición/fisiología , Estudios Cruzados , Citocinas/sangre , Método Doble Ciego , Electroencefalografía , Voluntarios Sanos , Humanos , Hidrocortisona/análisis , Masculino , Pruebas Neuropsicológicas , Probióticos/uso terapéutico , Saliva/química , Estrés Psicológico/psicología , Adulto Joven
6.
Neuroimage ; 125: 988-995, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26577887

RESUMEN

The gut microbiome has been shown to regulate the development and functions of the enteric and central nervous systems. Its involvement in the regulation of behavior has attracted particular attention because of its potential translational importance in clinical disorders, however little is known about the pathways involved. We previously have demonstrated that administration of Lactobacillus rhamnosus (JB-1) to healthy male BALB/c mice, promotes consistent changes in GABA-A and -B receptor sub-types in specific brain regions, accompanied by reductions in anxiety and depression-related behaviors. In the present study, using magnetic resonance spectroscopy (MRS), we quantitatively assessed two clinically validated biomarkers of brain activity and function, glutamate+glutamine (Glx) and total N-acetyl aspartate+N-acetyl aspartyl glutamic acid (tNAA), as well as GABA, the chief brain inhibitory neurotransmitter. Mice received 1×10(9) cfu of JB-1 per day for 4weeks and were subjected to MRS weekly and again 4weeks after cessation of treatment to ascertain temporal changes in these neurometabolites. Baseline concentrations for Glx, tNAA and GABA were equal to 10.4±0.3mM, 8.7±0.1mM, and 1.2±0.1mM, respectively. Delayed increases were first seen for Glx (~10%) and NAA (~37%) at 2weeks which persisted only to the end of treatment. However, Glx was still elevated 4weeks after treatment had ceased. Significantly elevated GABA (~25%) was only seen at 4weeks. These results suggest specific metabolic pathways in our pursuit of mechanisms of action of psychoactive bacteria. They also offer through application of standard clinical neurodiagnostic techniques, translational opportunities to assess biomarkers accompanying behavioral changes induced by alterations in the gut microbiome.


Asunto(s)
Ácido Aspártico/análogos & derivados , Encéfalo/metabolismo , Ácido Glutámico/biosíntesis , Lacticaseibacillus rhamnosus , Probióticos/farmacología , Ácido gamma-Aminobutírico/biosíntesis , Animales , Ácido Aspártico/análisis , Ácido Aspártico/biosíntesis , Química Encefálica/efectos de los fármacos , Ensayo de Inmunoadsorción Enzimática , Microbioma Gastrointestinal/fisiología , Ácido Glutámico/análisis , Imagen por Resonancia Magnética , Espectroscopía de Resonancia Magnética , Masculino , Ratones , Ratones Endogámicos BALB C , Ácido gamma-Aminobutírico/análisis
7.
BMC Med ; 14: 58, 2016 Apr 19.
Artículo en Inglés | MEDLINE | ID: mdl-27090095

RESUMEN

INTRODUCTION: The microbiota-gut-brain axis is a term that is commonly used and covers a broad set of functions and interactions between the gut microbiome, endocrine, immune and nervous systems and the brain. The field is not much more than a decade old and so large holes exist in our knowledge. DISCUSSION: At first sight it appears gut microbes are largely responsible for the development, maturation and adult function of the enteric nervous system as well as the blood brain barrier, microglia and many aspects of the central nervous system structure and function. Given the state of the art in this exploding field and the hopes, as well as the skepticism, which have been engendered by its popular appeal, we explore recent examples of evidence in rodents and data derived from studies in humans, which offer insights as to pathways involved. Communication between gut and brain depends on both humoral and nervous connections. Since these are bi-directional and occur through complex communication pathways, it is perhaps not surprising that while striking observations have been reported, they have often either not yet been reproduced or their replication by others has not been successful. CONCLUSIONS: We offer critical and cautionary commentary on the available evidence, and identify gaps in our knowledge that need to be filled so as to achieve translation, where possible, into beneficial application in the clinical setting.


Asunto(s)
Encéfalo , Sistema Nervioso Entérico , Microbioma Gastrointestinal , Tracto Gastrointestinal , Neuroinmunomodulación , Encéfalo/fisiología , Heces , Humanos
8.
FASEB J ; 29(2): 684-95, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25392266

RESUMEN

Ingestion of a commensal bacteria, Lactobacillus rhamnosus JB-1, has potent immunoregulatory effects, and changes nerve-dependent colon migrating motor complexes (MMCs), enteric nerve function, and behavior. How these alterations occur is unknown. JB-1 microvesicles (MVs) are enriched for heat shock protein components such as chaperonin 60 heat-shock protein isolated from Escherichia coli (GroEL) and reproduce regulatory and neuronal effects in vitro and in vivo. Ingested labeled MVs were detected in murine Peyer's patch (PP) dendritic cells (DCs) within 18 h. After 3 d, PP and mesenteric lymph node DCs assumed a regulatory phenotype and increased functional regulatory CD4(+)25(+)Foxp3+ T cells. JB-1, MVs, and GroEL similarly induced phenotypic change in cocultured DCs via multiple pathways including C-type lectin receptors specific intercellular adhesion molecule-3 grabbing non-integrin-related 1 and Dectin-1, as well as TLR-2 and -9. JB-1 and MVs also decreased the amplitude of neuronally dependent MMCs in an ex vivo model of peristalsis. Gut epithelial, but not direct neuronal application of, MVs, replicated functional effects of JB-1 on in situ patch-clamped enteric neurons. GroEL and anti-TLR-2 were without effect in this system, suggesting the importance of epithelium neuron signaling and discrimination between pathways for bacteria-neuron and -immune communication. Together these results offer a mechanistic explanation of how Gram-positive commensals and probiotics may influence the host's immune and nervous systems.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Tracto Gastrointestinal/inervación , Sistema Inmunológico/fisiología , Lacticaseibacillus rhamnosus/inmunología , Animales , Células de la Médula Ósea/citología , Linfocitos T CD4-Positivos/citología , Chaperonina 60/metabolismo , Técnicas de Cocultivo , Células Dendríticas/citología , Células Dendríticas/microbiología , Factores de Transcripción Forkhead/metabolismo , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Lectinas Tipo C/metabolismo , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Neuronas/metabolismo , Peristaltismo , Ganglios Linfáticos Agregados/microbiología , Fenotipo , Probióticos , Proteómica , Transducción de Señal
9.
Can J Psychiatry ; 61(4): 204-13, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-27254412

RESUMEN

Gut bacteria strongly influence our metabolic, endocrine, immune, and both peripheral and central nervous systems. Microbiota do this directly and indirectly through their components, shed and secreted, ranging from fermented and digested dietary and host products to functionally active neurotransmitters including serotonin, dopamine, and γ-aminobutyric acid. Depression has been associated with enhanced levels of proinflammatory biomarkers and abnormal responses to stress. Posttraumatic stress disorder (PTSD) appears to be marked in addition by low cortisol responses, and these factors seem to predict and predispose individuals to develop PTSD after a traumatic event. Dysregulation of the immune system and of the hypothalamic-pituitary-adrenal axis observed in PTSD may reflect prior trauma exposure, especially early in life. Early life, including the prenatal period, is a critical time in rodents, and may well be for humans, for the functional and structural development of the immune and nervous systems. These, in turn, are likely shaped and programmed by gut and possibly other bacteria. Recent experimental and clinical data converge on the hypothesis that imbalanced gut microbiota in early life may have long-lasting immune and other physiologic effects that make individuals more susceptible to develop PTSD after a traumatic event and contribute to the disorder. This suggests that it may be possible to target abnormalities in these systems by manipulation of certain gut bacterial communities directly through supplementation or indirectly by dietary and other novel approaches.


Asunto(s)
Microbioma Gastrointestinal , Trastornos por Estrés Postraumático , Microbioma Gastrointestinal/inmunología , Humanos , Trastornos por Estrés Postraumático/etiología , Trastornos por Estrés Postraumático/inmunología , Trastornos por Estrés Postraumático/microbiología
10.
J Physiol ; 593(17): 3943-57, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26084409

RESUMEN

Certain probiotic bacteria have been shown to reduce distension-dependent gut pain, but the mechanisms involved remain obscure. Live luminal Lactobacillus reuteri (DSM 17938) and its conditioned medium dose dependently reduced jejunal spinal nerve firing evoked by distension or capsaicin, and 80% of this response was blocked by a specific TRPV1 channel antagonist or in TRPV1 knockout mice. The specificity of DSM action on TRPV1 was further confirmed by its inhibition of capsaicin-induced intracellular calcium increases in dorsal root ganglion neurons. Another lactobacillus with ability to reduce gut pain did not modify this response. Prior feeding of rats with DSM inhibited the bradycardia induced by painful gastric distension. These results offer a system for the screening of new and improved candidate bacteria that may be useful as novel therapeutic adjuncts in gut pain. Certain bacteria exert visceral antinociceptive activity, but the mechanisms involved are not determined. Lactobacillus reuteri DSM 17938 was examined since it may be antinociceptive in children. Since transient receptor potential vanilloid 1 (TRPV1) channel activity may mediate nociceptive signals, we hypothesized that TRPV1 current is inhibited by DSM. We tested this by examining the effect of DSM on the firing frequency of spinal nerve fibres in murine jejunal mesenteric nerve bundles following serosal application of capsaicin. We also measured the effects of DSM on capsaicin-evoked increase in intracellular Ca(2+) or ionic current in dorsal root ganglion (DRG) neurons. Furthermore, we tested the in vivo antinociceptive effects of oral DSM on gastric distension in rats. Live DSM reduced the response of capsaicin- and distension-evoked firing of spinal nerve action potentials (238 ± 27.5% vs. 129 ± 17%). DSM also reduced the capsaicin-evoked TRPV1 ionic current in DRG neuronal primary culture from 83 ± 11% to 41 ± 8% of the initial response to capsaicin only. Another lactobacillus (Lactobacillus rhamnosus JB-1) with known visceral anti-nociceptive activity did not have these effects. DSM also inhibited capsaicin-evoked Ca(2+) increase in DRG neurons; an increase in Ca(2+) fluorescence intensity ratio of 2.36 ± 0.31 evoked by capsaicin was reduced to 1.25 ± 0.04. DSM releasable products (conditioned medium) mimicked DSM inhibition of capsaicin-evoked excitability. The TRPV1 antagonist 6-iodonordihydrocapsaicin or the use of TRPV1 knock-out mice revealed that TRPV1 channels mediate about 80% of the inhibitory effect of DSM on mesenteric nerve response to high intensity gut distension. Finally, feeding with DSM inhibited perception in rats of painful gastric distension. Our results identify a specific target channel for a probiotic with potential therapeutic properties.


Asunto(s)
Bradicardia/terapia , Yeyuno/fisiología , Limosilactobacillus reuteri , Probióticos , Gastropatías/terapia , Canales Catiónicos TRPV/fisiología , Analgesia , Animales , Bradicardia/etiología , Bradicardia/fisiopatología , Capsaicina , Ganglios Espinales/fisiología , Yeyuno/inervación , Masculino , Mesenterio/inervación , Mesenterio/fisiología , Ratones Noqueados , Probióticos/farmacología , Probióticos/uso terapéutico , Ratas Sprague-Dawley , Nervios Espinales/fisiología , Gastropatías/complicaciones , Gastropatías/fisiopatología , Canales Catiónicos TRPV/genética
11.
FASEB J ; 28(7): 3064-74, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24719355

RESUMEN

It is generally accepted that intestinal sensory vagal fibers are primary afferent, responding nonsynaptically to luminal stimuli. The gut also contains intrinsic primary afferent neurons (IPANs) that respond to luminal stimuli. A psychoactive Lactobacillus rhamnosus (JB-1) that affects brain function excites both vagal fibers and IPANs. We wondered whether, contrary to its primary afferent designation, the sensory vagus response to JB-1 might depend on IPAN to vagal fiber synaptic transmission. We recorded ex vivo single- and multiunit afferent action potentials from mesenteric nerves supplying mouse jejunal segments. Intramural synaptic blockade with Ca(2+) channel blockers reduced constitutive or JB-1-evoked vagal sensory discharge. Firing of 60% of spontaneously active units was reduced by synaptic blockade. Synaptic or nicotinic receptor blockade reduced firing in 60% of vagal sensory units that were stimulated by luminal JB-1. In control experiments, increasing or decreasing IPAN excitability, respectively increased or decreased nerve firing that was abolished by synaptic blockade or vagotomy. We conclude that >50% of vagal afferents function as interneurons for stimulation by JB-1, receiving input from an intramural functional "sensory synapse." This was supported by myenteric plexus nicotinic receptor immunohistochemistry. These data offer a novel therapeutic target to modify pathological gut-brain axis activity.-Perez-Burgos, A., Mao, Y.-K., Bienenstock, J., Kunze, W. A. The gut-brain axis rewired: adding a functional vagal nicotinic "sensory synapse."


Asunto(s)
Encéfalo/fisiología , Yeyuno/fisiología , Neuronas Aferentes/fisiología , Receptores Nicotínicos/metabolismo , Sinapsis/metabolismo , Nervio Vago/fisiología , Potenciales de Acción/fisiología , Animales , Encéfalo/metabolismo , Yeyuno/inervación , Yeyuno/metabolismo , Yeyuno/microbiología , Lacticaseibacillus rhamnosus/metabolismo , Masculino , Ratones , Nervio Vago/metabolismo
12.
Can J Psychiatry ; 65(4): 278-280, 2020 04.
Artículo en Francés | MEDLINE | ID: mdl-31958990

RESUMEN

OBJECTIVES: Compelling animal data exists examining the impact of the gut microbiome on the brain, but work is required to translate these findings in a clinical population. We sought to do this by exploring the effects of antidepressant medications on the gut microbiota, and establishing a baseline Major Depressive Disorder (MDD) gut phenotype. METHODS: Participants with a primary diagnosis of MDD (n = 15) who were nonmedicated were recruited and followed over 6 months. Stool samples were collected prior to treatment initiation and 3 and 6 months following treatment. 16S rRNA sequencing was employed in order to analyze the gut microbial community profile. Symptom severity was measured by the Beck Depression Inventory. Alpha diversity metrics revealed no significant difference in the community diversity across any of the time-points. RESULTS: Comparison of within-group versus between-group distances revealed a lack of clustering of samples based on time-point, suggesting no significant change in the microbiota across treatment duration. When analyzed based on treatment response, however, patients in the responder group exhibited greater phylogenetic diversity than non-responders (Mann-Whitney U = 5, p = 0.026). At 3-months, 35 Operational Taxonomic Units (OTUs) were significantly different between groups and at 6-months, 42 OTUs were significantly different between responders and non-responders. CONCLUSIONS: These observations indicate that antidepressant medications alter the gut microbiota of patients with MDD, with disparate effects in responders versus non responders. This supports the concept of a microbiota phenotype associate with treatment response in MDD.


Asunto(s)
Antidepresivos de Segunda Generación , Trastorno Depresivo Mayor/tratamiento farmacológico , Trastorno Depresivo Mayor/microbiología , Trastorno Depresivo Mayor/fisiopatología , Microbioma Gastrointestinal , Evaluación de Resultado en la Atención de Salud , Adulto , Antidepresivos de Segunda Generación/farmacología , Citalopram/farmacología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Humanos , Estudios Longitudinales , Masculino , Persona de Mediana Edad , ARN Ribosómico 16S , Inducción de Remisión , Análisis de Secuencia de ARN , Índice de Severidad de la Enfermedad
13.
J Allergy Clin Immunol ; 134(1): 160-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25117802

RESUMEN

BACKGROUND: There is evidence to suggest an association between prenatal maternal stress and the development of asthma or other atopic diseases in offspring. Yet, insights on the lasting effect of multiple, common prenatal stressors are rare, and the effects of prenatal timing are poorly understood. Further, it remains elusive if prenatal life events modify the risk for atopic diseases in the context of a parental predisposition to atopy. OBJECTIVE: We tested whether women's experiences of common, adverse life events during the first or second half of pregnancy predicted the risk of developing atopic diseases in their children and whether a reported parental atopic disease moderated this association. METHODS: We calculated the odds of a child developing asthma, eczema, and/or allergic rhinitis at ages 6 or 14 years, depending on maternal prenatal exposure to negative life events in a sample of 1587 children from the Western Australian Pregnancy Cohort (Raine) Study by using multivariable logistic regression. RESULTS: We observed that the likelihood of asthma and eczema at age 14 years was significantly increased in children of mothers who had experienced adverse life events during the second half of gestation (1 life event: adjusted odds ratio for asthma, 2.08 [95% CI, 1.22-3.54]). A stronger increase in the odds to develop asthma upon prenatal life events was present in children of mothers without asthma compared with mothers with asthma. CONCLUSIONS: Maternal adverse life events during the second half of gestation are linked to an increased risk for the development of atopic disorders, asthma, and eczema, in the case of asthma, particularly in the absence of a maternal asthma.


Asunto(s)
Asma/etiología , Eccema/etiología , Efectos Tardíos de la Exposición Prenatal/psicología , Estrés Psicológico/psicología , Adolescente , Adulto , Asma/inmunología , Asma/patología , Australia , Niño , Eccema/inmunología , Eccema/patología , Femenino , Humanos , Modelos Logísticos , Estudios Longitudinales , Masculino , Oportunidad Relativa , Embarazo , Efectos Tardíos de la Exposición Prenatal/inmunología , Efectos Tardíos de la Exposición Prenatal/patología , Riesgo , Factores de Riesgo , Estrés Psicológico/complicaciones , Estrés Psicológico/inmunología , Estrés Psicológico/patología , Encuestas y Cuestionarios
14.
Adv Exp Med Biol ; 817: 115-33, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24997031

RESUMEN

There is now strong evidence from animal studies that gut microorganism can activate the vagus nerve and that such activation plays a critical role in mediating effects on the brain and behaviour. The vagus appears to differentiate between non-pathogenic and potentially pathogenic bacteria even in the absence of overt inflammation and vagal pathways mediate signals that can induce both anxiogenic and anxiolytic effects, depending on the nature of the stimulus. Certain vagal signals from the gut can instigate an anti-inflammatory reflex with afferent signals to the brain activating an efferent response, releasing mediators including acetylcholine that, through an interaction with immune cells, attenuates inflammation. This immunomodulatory role of the vagus nerve may also have consequences for modulation of brain function and mood.What is currently lacking are relevant data on the electrophysiology of the system. Certainly, important advances in our understanding of the gut-brain and microbiome- gut-brain axis will come from studies of how distinct microbial and nutritional stimuli activate the vagus and the nature of the signals transmitted to the brain that lead to differential changes in the neurochemistry of the brain and behaviour.Understanding the induction and transmission of signals in the vagus nerve may have important implications for the development of microbial-or nutrition based therapeutic strategies for mood disorders.


Asunto(s)
Encéfalo/fisiología , Interacciones Huésped-Patógeno/fisiología , Intestinos/microbiología , Microbiota/fisiología , Nervio Vago/fisiología , Animales , Humanos , Reflejo
15.
Proc Natl Acad Sci U S A ; 108(38): 16050-5, 2011 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-21876150

RESUMEN

There is increasing, but largely indirect, evidence pointing to an effect of commensal gut microbiota on the central nervous system (CNS). However, it is unknown whether lactic acid bacteria such as Lactobacillus rhamnosus could have a direct effect on neurotransmitter receptors in the CNS in normal, healthy animals. GABA is the main CNS inhibitory neurotransmitter and is significantly involved in regulating many physiological and psychological processes. Alterations in central GABA receptor expression are implicated in the pathogenesis of anxiety and depression, which are highly comorbid with functional bowel disorders. In this work, we show that chronic treatment with L. rhamnosus (JB-1) induced region-dependent alterations in GABA(B1b) mRNA in the brain with increases in cortical regions (cingulate and prelimbic) and concomitant reductions in expression in the hippocampus, amygdala, and locus coeruleus, in comparison with control-fed mice. In addition, L. rhamnosus (JB-1) reduced GABA(Aα2) mRNA expression in the prefrontal cortex and amygdala, but increased GABA(Aα2) in the hippocampus. Importantly, L. rhamnosus (JB-1) reduced stress-induced corticosterone and anxiety- and depression-related behavior. Moreover, the neurochemical and behavioral effects were not found in vagotomized mice, identifying the vagus as a major modulatory constitutive communication pathway between the bacteria exposed to the gut and the brain. Together, these findings highlight the important role of bacteria in the bidirectional communication of the gut-brain axis and suggest that certain organisms may prove to be useful therapeutic adjuncts in stress-related disorders such as anxiety and depression.


Asunto(s)
Emociones/fisiología , Lactobacillus/fisiología , Receptores de GABA/genética , Nervio Vago/fisiología , Análisis del Polimorfismo de Longitud de Fragmentos Amplificados , Animales , Ansiedad/fisiopatología , Encéfalo/metabolismo , Encéfalo/fisiopatología , Corticosterona/sangre , ADN Bacteriano/genética , Depresión/fisiopatología , Fiebre/sangre , Fiebre/microbiología , Fiebre/fisiopatología , Expresión Génica , Interacciones Huésped-Patógeno , Hibridación in Situ , Lactobacillus/genética , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos BALB C , Actividad Motora/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de GABA-A/genética , Receptores de GABA-B/genética , Especificidad de la Especie , Vagotomía , Nervio Vago/cirugía
16.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G211-20, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23139216

RESUMEN

Mounting evidence supports the influence of the gut microbiome on the local enteric nervous system and its effects on brain chemistry and relevant behavior. Vagal afferents are involved in some of these effects. We previously showed that ingestion of the probiotic bacterium Lactobacillus rhamnosus (JB-1) caused extensive neurochemical changes in the brain and behavior that were abrogated by prior vagotomy. Because information can be transmitted to the brain via primary afferents encoded as neuronal spike trains, our goal was to record those induced by JB-1 in vagal afferents in the mesenteric nerve bundle and thus determine the nature of the signals sent to the brain. Male Swiss Webster mice jejunal segments were cannulated ex vivo, and serosal and luminal compartments were perfused separately. Bacteria were added intraluminally. We found no evidence for translocation of labeled bacteria across the epithelium during the experiment. We recorded extracellular multi- and single-unit neuronal activity with glass suction pipettes. Within minutes of application, JB-1 increased the constitutive single- and multiunit firing rate of the mesenteric nerve bundle, but Lactobacillus salivarius (a negative control) or media alone were ineffective. JB-1 significantly augmented multiunit discharge responses to an intraluminal distension pressure of 31 hPa. Prior subdiaphragmatic vagotomy abolished all of the JB-1-evoked effects. This detailed exploration of the neuronal spike firing that encodes behavioral signaling to the brain may be useful to identify effective psychoactive bacteria and thereby offer an alternative new perspective in the field of psychiatry and comorbid conditions.


Asunto(s)
Sistema Nervioso Entérico/fisiología , Yeyuno/inervación , Yeyuno/microbiología , Lacticaseibacillus rhamnosus/fisiología , Conducción Nerviosa , Probióticos , Nervio Vago/fisiología , Potenciales de Acción , Vías Aferentes/fisiología , Animales , Sistema Nervioso Entérico/cirugía , Masculino , Mecanotransducción Celular , Ratones , Presión , Tiempo de Reacción , Factores de Tiempo , Vagotomía , Nervio Vago/cirugía
17.
Curr Opin Gastroenterol ; 28(6): 557-62, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23010679

RESUMEN

PURPOSE OF REVIEW: Interest in the microbiota-gut-brain axis is increasing apace and what was, not so long ago, a hypothetical relationship is emerging as a potentially critical factor in the regulation of intestinal and mental health. Studies are now addressing the neural circuitry and mechanisms underlying the influence of gut bacteria on the central nervous system and behavior. RECENT FINDINGS: Gut bacteria influence development of the central nervous systems (CNS) and stress responses. In adult animals, the overall composition of the microbiota or exposure to specific bacterial strains can modulate neural function, peripherally and centrally. Gut bacteria can provide protection from the central effects of infection and inflammation as well as modulate normal behavioral responses. Behavioral effects described to date are largely related to stress and anxiety and an altered hypothalamus-pituitary-adrenal axis response is a common observation in many model systems. The vagus nerve has also emerged as an important means of communicating signals from gut microbes to the CNS. SUMMARY: Studies of microbiota-gut-brain communication are providing us with a deeper understanding of the relationship between the gut bacteria and their hosts while also suggesting the potential for microbial-based therapeutic strategies that may aid in the treatment of mood disorders.


Asunto(s)
Conducta/fisiología , Sistema Nervioso Central/fisiología , Tracto Gastrointestinal/microbiología , Trastornos Mentales/fisiopatología , Metagenoma/fisiología , Sistema Nervioso Periférico/fisiología , Animales , Humanos
18.
J Nutr ; 141(10): 1813-8, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21880952

RESUMEN

Probiotic lactic acid bacteria have been reported to alleviate symptoms in patients with irritable bowel syndrome. However, they have not been tested for use in functional gastric disease. We therefore investigated if strains previously shown to protect from response to colorectal distension (CRD) in rats also modulate response to gastric distension (GD). Healthy, male Sprague-Dawley rats were treated with viable, heat-killed, gamma-irradiated Lactobacillus reuteri or viable Lactobacillus plantarum wild type (WT), L. plantarum Dlt¯mutant, conditioned medium or medium control (9 d), and subjected to GD under anesthesia using an i.g. Teflon catheter. Effects were measured by heart rate (HR) changes during noxious distension (60 mm Hg) compared to baseline HR values. We also investigated the localization of viable, green fluorescent protein-transfected bacteria in the stomach mucosa. Viable L. reuteri decreased the bradycardia induced by noxious GD compared to placebo controls (P < 0.001). Heat-killed or gamma-irradiated L. reuteri and conditioned medium did not have a protective effect in GD. Viable L. plantarum WT and Dlt¯mutant, previously shown to be effective antinociceptive agents in CRD, showed no protective effect in GD. All viable bacteria were associated with the pars glandularis of the rat stomach. Thus, we conclude that the antinociceptive mechanisms of action of probiotic bacteria differ between the stomach and the colon. Symptom alleviation cannot be attributed to the localization of the bacteria in the stomach. Information derived from effects of CRD cannot be extrapolated to effects in the stomach, which are likely to be strain and organ specific.


Asunto(s)
Dilatación Gástrica/dietoterapia , Limosilactobacillus reuteri , Probióticos/uso terapéutico , Dolor Abdominal/prevención & control , Analgesia , Animales , Adhesión Bacteriana , Bradicardia/etiología , Bradicardia/prevención & control , Rayos gamma/efectos adversos , Dilatación Gástrica/patología , Dilatación Gástrica/fisiopatología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiología , Mucosa Gástrica/patología , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Calor/efectos adversos , Lactobacillus plantarum/crecimiento & desarrollo , Limosilactobacillus reuteri/crecimiento & desarrollo , Limosilactobacillus reuteri/metabolismo , Limosilactobacillus reuteri/efectos de la radiación , Masculino , Viabilidad Microbiana , Mutación , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes/metabolismo
19.
FASEB J ; 24(10): 4078-88, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20519636

RESUMEN

Gut commensals modulate host immune, endocrine, and metabolic functions. They also affect peripheral and central neural reflexes and function. We have previously shown that daily ingestion of Lactobacillus reuteri (LR) for 9 d inhibits the pseudoaffective cardiac response and spinal single-fiber discharge evoked by visceral distension, and decreases intestinal motility and myenteric AH cell slow afterhyperpolarization (sAHP) by inhibiting a Ca-activated K (IK(Ca)) channel. We tested whether luminal LR could acutely decrease motility in an ex vivo perfusion model of naive Balb/c jejunum. Live LR dose dependently decreased motor complex pressure wave amplitudes with 9- to 16-min onset latency and an IC(50) of 5 × 10(7) cells/ml Krebs. Heat-killed LR or another live commensal, Lactobacillus salivarius, were without effect. The IK(Ca) channel blocker TRAM-34, but neither the opener (DCEBIO) nor the hyperpolarization-activated cationic channel inhibitor ZD7288 (5 µM) (or TTX 1 µM), mimicked the LR effect on motility acutely ex vivo. We provide evidence for a rapid, strain-specific, dose-dependent action of a live Lactobacillus on small intestinal motility reflexes that recapitulates the long-term effects of LR ingestion. These observations may be useful as a first step to unraveling the pathways involved in bacteria to the nervous system communication.


Asunto(s)
Motilidad Gastrointestinal , Yeyuno/fisiología , Lactobacillus , Probióticos , Animales , Masculino , Ratones , Ratones Endogámicos BALB C
20.
Sci Rep ; 11(1): 13756, 2021 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-34215822

RESUMEN

Intestinal bacteria have diverse and complex influence on their host. Evidence is accumulating that this may be mediated in part by bacterial extracellular membrane vesicles (MV), nanometer-sized particles important for intercellular communication. Little is known about the composition of MV from gram-positive beneficial bacteria nor how they interact with intestinal epithelial cells (IEC). Here we demonstrate that MV from Lacticaseibacillus rhamnosus JB-1 are endocytosed in a likely clathrin-dependent manner by both mouse and human IEC in vitro and by mouse IEC in vivo. We further show that JB-1 MV contain lipoteichoic acid (LTA) that activates Toll-like receptor 2 (TLR2) and induces immunoregulatory interleukin-10 expression by dendritic cells in an internalization-dependent manner. By contrast, neither LTA nor TLR2 appear to be required for JB-1 MV endocytosis by IEC. These results demonstrate a novel mechanism by which bacterial MV can influence host physiology and suggest one potential route for beneficial influence of certain bacteria and probiotics.


Asunto(s)
Vesículas Extracelulares/química , Interleucina-10/genética , Lacticaseibacillus rhamnosus/genética , Receptor Toll-Like 2/genética , Animales , Membrana Celular/efectos de los fármacos , Endocitosis/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Bacterias Grampositivas/química , Humanos , Mucosa Intestinal/efectos de los fármacos , Lacticaseibacillus rhamnosus/química , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA